Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Dev Biol ; 477: 22-34, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34004181

RESUMEN

Branching networks are a very common feature of multicellular animals and underlie the formation and function of numerous organs including the nervous system, the respiratory system, the vasculature and many internal glands. These networks range from subcellular structures such as dendritic trees to large multicellular tissues such as the lungs. The production of branched structures by single cells, so called subcellular branching, which has been better described in neurons and in cells of the respiratory and vascular systems, involves complex cytoskeletal remodelling events. In Drosophila, tracheal system terminal cells (TCs) and nervous system dendritic arborisation (da) neurons are good model systems for these subcellular branching processes. During development, the generation of subcellular branches by single-cells is characterized by extensive remodelling of the microtubule (MT) network and actin cytoskeleton, followed by vesicular transport and membrane dynamics. In this review, we describe the current knowledge on cytoskeletal regulation of subcellular branching, based on the terminal cells of the Drosophila tracheal system, but drawing parallels with dendritic branching and vertebrate vascular subcellular branching.


Asunto(s)
Diferenciación Celular/fisiología , Citoesqueleto/fisiología , Drosophila melanogaster/embriología , Morfogénesis , Neurogénesis/fisiología , Actinas/fisiología , Animales , Comunicación Celular , Drosophila melanogaster/citología , Endotelio/embriología , Humanos , Microtúbulos/fisiología , Análisis de la Célula Individual , Tráquea/citología , Tráquea/embriología
2.
Angiogenesis ; 24(2): 199-211, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33783643

RESUMEN

Hematopoiesis in vertebrate embryos occurs in temporally and spatially overlapping waves in close proximity to blood vascular endothelial cells. Initially, yolk sac hematopoiesis produces primitive erythrocytes, megakaryocytes, and macrophages. Thereafter, sequential waves of definitive hematopoiesis arise from yolk sac and intraembryonic hemogenic endothelia through an endothelial-to-hematopoietic transition (EHT). During EHT, the endothelial and hematopoietic transcriptional programs are tightly co-regulated to orchestrate a shift in cell identity. In the yolk sac, EHT generates erythro-myeloid progenitors, which upon migration to the liver differentiate into fetal blood cells, including erythrocytes and tissue-resident macrophages. In the dorsal aorta, EHT produces hematopoietic stem cells, which engraft the fetal liver and then the bone marrow to sustain adult hematopoiesis. Recent studies have defined the relationship between the developing vascular and hematopoietic systems in animal models, including molecular mechanisms that drive the hemato-endothelial transcription program for EHT. Moreover, human pluripotent stem cells have enabled modeling of fetal human hematopoiesis and have begun to generate cell types of clinical interest for regenerative medicine.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Linaje de la Célula/fisiología , Células Endoteliales/metabolismo , Endotelio/embriología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales/citología , Endotelio/citología , Células Madre Hematopoyéticas/citología , Humanos
3.
Pathol Res Pract ; 216(11): 153225, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32987302

RESUMEN

The in vitro 3D model established from murine pluripotential stem cells (i.e., embryoid bodies (EBs)) is a dynamic model for endothelial differentiation. The aim of the present study was to investigate whether digital image analysis (DIA) can be applied on histological sections of EBs in order to quantify endothelial differentiation over time. The EBs were established in suspension cultures for 21 days in three independent replicate experiments. At day 4, 6, 9, 14, 18, and 21, the EBs were fixed in formaldehyde, embedded in paraffin and immunohistochemically (IHC) stained for CD31. The IHC-stained slides were digitally scanned and analysed using the Visiopharm® Quantitative Digital Pathology software Oncotopix™. The EBs developed CD31+ vascular-like structures during their differentiation. The quantitative DIA of the EBs showed that the log10 values of the relative CD31+ areas increased from -0.574 ± 0.470 (mean ± SD) at day 4 to 0.093 ± 0.688 (mean ± SD) at day 21 (p < 0.001). The approach presented in this study is a fast, quantitative and reproducible alternative method for an otherwise time-consuming and observer-dependent histological investigation. The future perspectives for such a system would be implementation of a modified version of the method on different 3D cultures and IHC markers.


Asunto(s)
Diferenciación Celular/fisiología , Cuerpos Embrioides/citología , Endotelio/embriología , Células Madre Pluripotentes/citología , Animales , Procesamiento de Imagen Asistido por Computador , Ratones
4.
Cells ; 9(5)2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32438714

RESUMEN

Different sources have been claimed for the embryonic origin of the coronary endothelium. Recently, the potential of circulating cells as progenitors of the cardiac endothelium has also been suggested. In a previous study we have shown that circulating progenitors are recruited by the embryonic endocardium and incorporated into the coronary vessels. These progenitors derive from a mesodermal lineage characterized by the expression of Gata4 under control of the enhancer G2. Herein, we aim to trace this specific lineage throughout postnatal stages. We have found that more than 50% of the adult cardiac endothelium derives from the G2-GATA4 lineage. This percentage increases from embryos to adults probably due to differential proliferation and postnatal recruitment of circulating endothelial progenitors. In fact, injection of fetal liver or placental cells in the blood stream of neonates leads to incorporation of G2-GATA4 lineage cells to the coronary endothelium. On the other hand, labeling of the hematopoietic lineage by the stage E7.5 also resulted in positive coronary endothelial cells from both, embryos and adults. Our results suggest that early hematopoietic progenitors recruited by the embryonic ventricular endocardium can become the predominant source of definitive endothelium during the vascularization of the heart.


Asunto(s)
Envejecimiento/metabolismo , Linaje de la Célula , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio/metabolismo , Factor de Transcripción GATA4/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Animales Recién Nacidos , Embrión de Mamíferos/citología , Endotelio/embriología , Riñón/citología , Riñón/embriología , Mesodermo/embriología , Ratones , Miocardio/citología
5.
Blood ; 136(7): 845-856, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32392346

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) in the bone marrow are derived from a small population of hemogenic endothelial (HE) cells located in the major arteries of the mammalian embryo. HE cells undergo an endothelial to hematopoietic cell transition, giving rise to HSPCs that accumulate in intra-arterial clusters (IAC) before colonizing the fetal liver. To examine the cell and molecular transitions between endothelial (E), HE, and IAC cells, and the heterogeneity of HSPCs within IACs, we profiled ∼40 000 cells from the caudal arteries (dorsal aorta, umbilical, vitelline) of 9.5 days post coitus (dpc) to 11.5 dpc mouse embryos by single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing. We identified a continuous developmental trajectory from E to HE to IAC cells, with identifiable intermediate stages. The intermediate stage most proximal to HE, which we term pre-HE, is characterized by increased accessibility of chromatin enriched for SOX, FOX, GATA, and SMAD motifs. A developmental bottleneck separates pre-HE from HE, with RUNX1 dosage regulating the efficiency of the pre-HE to HE transition. A distal candidate Runx1 enhancer exhibits high chromatin accessibility specifically in pre-HE cells at the bottleneck, but loses accessibility thereafter. Distinct developmental trajectories within IAC cells result in 2 populations of CD45+ HSPCs; an initial wave of lymphomyeloid-biased progenitors, followed by precursors of hematopoietic stem cells (pre-HSCs). This multiomics single-cell atlas significantly expands our understanding of pre-HSC ontogeny.


Asunto(s)
Diferenciación Celular , Endotelio/embriología , Hemangioblastos/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Diferenciación Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Embrión de Mamíferos , Endotelio/citología , Endotelio/metabolismo , Femenino , Dosificación de Gen/fisiología , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos/citología , Hematopoyesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , RNA-Seq/métodos
6.
Int J Mol Sci ; 21(1)2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31861391

RESUMEN

The endothelium is an additional cell layer, differentiating from the inner epidermis of the ovule integument. In tomato (Solanum lycopersicum L.), after fertilization, the endothelium separates from integument and becomes an independent tissue developing next to the growing embryo sac. In the absence of fertilization, the endothelium may proliferate and form pseudo-embryo. However, the course of the reorganization of endothelium into pseudo-embryo in tomato ovules is poorly understood. We aimed to investigate specific features of endothelium differentiation and the role of the endothelium in the development of fertilized and unfertilized tomato ovules. The ovules of tomato plants ("YaLF" line), produced by vegetative growth plants of transgenic tomato line expressing the ac gene, encoding chitin-binding protein from Amaranthus caudatus L., were investigated using light and transmission electron microscopy. We showed that in the fertilized ovule of normally developing fruit and in the unfertilized ovule of parthenocarpic fruit, separation of the endothelium from integument occurs via programmed death of cells of the integumental parenchyma, adjacent to the endothelium. Endothelial cells in normally developing ovules change their structural and functional specialization from meristematic to secretory and back to meristematic, and proliferate until seeds fully mature. The secretory activity of the endothelium is necessary for the lysis of dying cells of the integument and provides the space for the growth of the new sporophyte. However, in ovules of parthenocarpic fruits, pseudo-embryo cells do not change their structural and functional organization and remain meristematic, no zone of lysis is formed, and pseudo-embryo cells undergo programmed cell death. Our data shows the key role of the endothelium as a protective and secretory tissue, needed for the normal development of ovules.


Asunto(s)
Endotelio/embriología , Endotelio/metabolismo , Células Germinativas de las Plantas/citología , Células Germinativas de las Plantas/metabolismo , Desarrollo de la Planta , Solanum lycopersicum/fisiología , Diferenciación Celular , Endotelio/citología , Fertilización , Flores , Regulación de la Expresión Génica de las Plantas , Células Germinativas de las Plantas/ultraestructura , Solanum lycopersicum/ultraestructura , Plantas Modificadas Genéticamente
7.
Nat Commun ; 10(1): 1083, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842454

RESUMEN

VEGFA signaling controls physiological and pathological angiogenesis and hematopoiesis. Although many context-dependent signaling pathways downstream of VEGFA have been uncovered, vegfa transcriptional regulation in vivo remains unclear. Here, we show that the ETS transcription factor, Etv6, positively regulates vegfa expression during Xenopus blood stem cell development through multiple transcriptional inputs. In agreement with its established repressive functions, Etv6 directly inhibits expression of the repressor foxo3, to prevent Foxo3 from binding to and repressing the vegfa promoter. Etv6 also directly activates expression of the activator klf4; reflecting a genome-wide paucity in ETS-binding motifs in Etv6 genomic targets, Klf4 then recruits Etv6 to the vegfa promoter to activate its expression. These two mechanisms (double negative gate and feed-forward loop) are classic features of gene regulatory networks specifying cell fates. Thus, Etv6's dual function, as a transcriptional repressor and activator, controls a major signaling pathway involved in endothelial and blood development in vivo.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Represoras/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/fisiología , Animales , Embrión no Mamífero , Endotelio/embriología , Endotelio/metabolismo , Proteína Forkhead Box O3/antagonistas & inhibidores , Proteína Forkhead Box O3/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/genética , Morfolinos/genética , Oligonucleótidos Antisentido/genética , Proteínas Proto-Oncogénicas c-ets/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal/fisiología , Somitos/embriología , Somitos/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Xenopus/antagonistas & inhibidores , Proteínas de Xenopus/genética , Proteína ETS de Variante de Translocación 6
8.
Nature ; 566(7745): 490-495, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30787436

RESUMEN

Across the animal kingdom, gastrulation represents a key developmental event during which embryonic pluripotent cells diversify into lineage-specific precursors that will generate the adult organism. Here we report the transcriptional profiles of 116,312 single cells from mouse embryos collected at nine sequential time points ranging from 6.5 to 8.5 days post-fertilization. We construct a molecular map of cellular differentiation from pluripotency towards all major embryonic lineages, and explore the complex events involved in the convergence of visceral and primitive streak-derived endoderm. Furthermore, we use single-cell profiling to show that Tal1-/- chimeric embryos display defects in early mesoderm diversification, and we thus demonstrate how combining temporal and transcriptional information can illuminate gene function. Together, this comprehensive delineation of mammalian cell differentiation trajectories in vivo represents a baseline for understanding the effects of gene mutations during development, as well as a roadmap for the optimization of in vitro differentiation protocols for regenerative medicine.


Asunto(s)
Diferenciación Celular/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Gastrulación , Organogénesis , Análisis de la Célula Individual , Animales , Linaje de la Célula/genética , Quimera/embriología , Quimera/genética , Quimera/metabolismo , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Endotelio/citología , Endotelio/embriología , Endotelio/metabolismo , Femenino , Gastrulación/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/genética , Masculino , Mesodermo/citología , Mesodermo/embriología , Ratones , Mutación/genética , Células Mieloides/citología , Organogénesis/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/embriología , Proteína 1 de la Leucemia Linfocítica T Aguda/deficiencia , Proteína 1 de la Leucemia Linfocítica T Aguda/genética
9.
Curr Top Dev Biol ; 132: 177-220, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30797509

RESUMEN

Organogenesis requires tightly coordinated and patterned growth of numerous cell types to form a fully mature and vascularized organ. Endothelial cells (ECs) that line blood vessels develop alongside the growing organ, but only recently has their role in directing epithelial and stromal growth been appreciated. Endothelial, epithelial, and stromal cells in embryonic organs actively communicate with one another throughout development to ensure that the organ forms appropriately. What signals tell blood vessel progenitors where to go? How are tissues influenced by the vasculature that pervades it? In this chapter, we review the ways in which crosstalk between ECs and epithelial or stromal cells during development leads to a fully patterned pancreas, lung, or kidney. ECs in all of these organs are necessary for proper epithelial and stromal growth, but how they direct this process is organ- and time-specific, highlighting the concept of dynamic EC heterogeneity. We end with a discussion on how understanding cell-cell crosstalk during development can be applied therapeutically through the generation of transplantable miniature organ-like tissues called "organoids." We will discuss the current state of organoid technology and highlight the major challenges in forming a properly patterned vascular network that will be critical in transforming them into a viable therapeutic option.


Asunto(s)
Vasos Sanguíneos/embriología , Células Endoteliales/fisiología , Endotelio/embriología , Organogénesis , Animales , Vasos Sanguíneos/citología , Biología Evolutiva/métodos , Biología Evolutiva/tendencias , Endotelio/citología , Humanos , Riñón/irrigación sanguínea , Riñón/citología , Riñón/embriología , Pulmón/irrigación sanguínea , Pulmón/citología , Pulmón/embriología , Páncreas/irrigación sanguínea , Páncreas/citología , Páncreas/embriología , Medicina Regenerativa/métodos , Medicina Regenerativa/tendencias
11.
Elife ; 72018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29555020

RESUMEN

Recent advances in single-cell transcriptomics techniques have opened the door to the study of gene regulatory networks (GRNs) at the single-cell level. Here, we studied the GRNs controlling the emergence of hematopoietic stem and progenitor cells from mouse embryonic endothelium using a combination of single-cell transcriptome assays. We found that a heptad of transcription factors (Runx1, Gata2, Tal1, Fli1, Lyl1, Erg and Lmo2) is specifically co-expressed in an intermediate population expressing both endothelial and hematopoietic markers. Within the heptad, we identified two sets of factors of opposing functions: one (Erg/Fli1) promoting the endothelial cell fate, the other (Runx1/Gata2) promoting the hematopoietic fate. Surprisingly, our data suggest that even though Fli1 initially supports the endothelial cell fate, it acquires a pro-hematopoietic role when co-expressed with Runx1. This work demonstrates the power of single-cell RNA-sequencing for characterizing complex transcription factor dynamics.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Análisis de la Célula Individual/métodos , Factores de Transcripción/genética , Animales , Análisis por Conglomerados , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Endotelio/citología , Endotelio/embriología , Endotelio/metabolismo , Redes Reguladoras de Genes , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Proto-Oncogénica c-fli-1/genética
12.
J Clin Invest ; 127(3): 1099-1114, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28218627

RESUMEN

Microvascular endothelial cells (ECs) are increasingly recognized as organ-specific gatekeepers of their microenvironment. Microvascular ECs instruct neighboring cells in their organ-specific vascular niches through angiocrine factors, which include secreted growth factors (angiokines), extracellular matrix molecules, and transmembrane proteins. However, the molecular regulators that drive organ-specific microvascular transcriptional programs and thereby regulate angiodiversity are largely elusive. In contrast to other ECs, which form a continuous cell layer, liver sinusoidal ECs (LSECs) constitute discontinuous, permeable microvessels. Here, we have shown that the transcription factor GATA4 controls murine LSEC specification and function. LSEC-restricted deletion of Gata4 caused transformation of discontinuous liver sinusoids into continuous capillaries. Capillarization was characterized by ectopic basement membrane deposition, formation of a continuous EC layer, and increased expression of VE-cadherin. Correspondingly, ectopic expression of GATA4 in cultured continuous ECs mediated the downregulation of continuous EC-associated transcripts and upregulation of LSEC-associated genes. The switch from discontinuous LSECs to continuous ECs during embryogenesis caused liver hypoplasia, fibrosis, and impaired colonization by hematopoietic progenitor cells, resulting in anemia and embryonic lethality. Thus, GATA4 acts as master regulator of hepatic microvascular specification and acquisition of organ-specific vascular competence, which are indispensable for liver development. The data also establish an essential role of the hepatic microvasculature in embryonic hematopoiesis.


Asunto(s)
Diferenciación Celular/fisiología , Embrión de Mamíferos/enzimología , Células Endoteliales/metabolismo , Endotelio/embriología , Factor de Transcripción GATA4/metabolismo , Hematopoyesis/fisiología , Hígado/embriología , Animales , Capilares/embriología , Factor de Transcripción GATA4/genética , Hígado/irrigación sanguínea , Ratones , Ratones Transgénicos , Especificidad de Órganos/fisiología
13.
Dev Biol ; 421(2): 108-117, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27955943

RESUMEN

Oxygenated blood from the heart is directed into the systemic circulation through the aortic arch arteries (AAAs). The AAAs arise by remodeling of three symmetrical pairs of pharyngeal arch arteries (PAAs), which connect the heart with the paired dorsal aortae at mid-gestation. Aberrant PAA formation results in defects frequently observed in patients with lethal congenital heart disease. How the PAAs form in mammals is not understood. The work presented in this manuscript shows that the second heart field (SHF) is the major source of progenitors giving rise to the endothelium of the pharyngeal arches 3 - 6, while the endothelium in the pharyngeal arches 1 and 2 is derived from a different source. During the formation of the PAAs 3 - 6, endothelial progenitors in the SHF extend cellular processes toward the pharyngeal endoderm, migrate from the SHF and assemble into a uniform vascular plexus. This plexus then undergoes remodeling, whereby plexus endothelial cells coalesce into a large PAA in each pharyngeal arch. Taken together, our studies establish a platform for investigating cellular and molecular mechanisms regulating PAA formation and alterations that lead to disease.


Asunto(s)
Región Branquial/embriología , Endotelio/embriología , Corazón/embriología , Animales , Aorta/embriología , Región Branquial/citología , Supervivencia Celular , Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Endotelio/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Ratones Transgénicos , Células Madre/citología , Factores de Tiempo
14.
Stem Cell Reports ; 7(4): 802-816, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27693424

RESUMEN

A major hurdle for in vitro culturing of primary endothelial cells (ECs) is that they readily dedifferentiate, hampering their use for therapeutic applications. Human embryonic stem cells (hESCs) may provide an unlimited cell source; however, most current protocols deriving endothelial progenitor cells (EPCs) from hESCs use direct differentiation approaches albeit on undefined matrices, yet final yields are insufficient. We developed a method to culture monolayer hESCs on stem cell niche laminin (LN) LN511 or LN521 matrix. Here, we report a chemically defined, xeno-free protocol for differentiation of hESCs to EPCs using LN521 as the main culture substrate. We were able to generate ∼95% functional EPCs defined as VEGFR2+CD34+CD31+VE-Cadherin+. RNA-sequencing analyses of hESCs, EPCs, and primary human umbilical vein endothelial cells showed differentiation-related EC expression signatures, regarding basement membrane composition, cell-matrix interactions, and changes in endothelial lineage markers. Our results may facilitate production of stable ECs for the treatment of vascular diseases and in vitro cell modeling.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Laminina/metabolismo , Biomarcadores , Diferenciación Celular/genética , Línea Celular , Análisis por Conglomerados , Endotelio/embriología , Endotelio/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Mesodermo/embriología , Mesodermo/metabolismo , Transducción de Señal , Transcriptoma
15.
Mech Dev ; 140: 41-52, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26916723

RESUMEN

Among gap junction-encoding genes, the loss of connexin (Cx) 45 most profoundly obstructs embryogenesis through an endocardial cushion defect and conduction block. However, the interdependence of these defects is not known, and the details of conduction block have not been elucidated. Here, we examined mouse embryos with a region-specific deletion of Cx45 in the myocardium (CA-Cre; Cx45(flox/flox)) or endothelium (Tie2-Cre; Cx45(flox/flox)). Although the deletion of Cx45 in the myocardium was heterogeneous, the CA-Cre; Cx45(flox/flox) embryos were lethal at the same stage as the constitutive Cx45-deficient (Cx45(-/-)) embryos. We determined the onset and patterns of their conduction block through point-tracking in video recordings of embryonic heart contractions. An incomplete conduction block at the atrioventricular canal appeared at embryonic day (E) 8.5 and was predominant around the lethal E9.5 stage in both the Cx45(-/-) and CA-Cre; Cx45(flox/flox) embryos. Although the Cx45(-/-) hearts showed a consistently severe reduction in atrioventricular conduction velocity, the CA-Cre; Cx45(flox/flox) hearts had delay times within the normal range and showed frequent retrograde conduction. As previously reported, the Cx45(-/-) endocardial cushion was consistently defective, and nuclear factor of activated T-cells cytoplasmic (NFATc)1 within the endocardium showed inactive cytoplasmic distribution. In CA-Cre; Cx45(flox/flox), however, the endocardial cushion was partially formed, with active NFATc1 within the endocardium. There was no developmental abnormality in the Tie2-Cre; Cx45(flox/flox) embryos. These results indicate that myocardial dysfunction is responsible for most of the reported defects in Cx45(-/-), which are alleviated by sporadic Cx45 expression in the CA-Cre; Cx45(flox/flox) myocardium.


Asunto(s)
Conexinas/metabolismo , Endocardio/embriología , Endocardio/metabolismo , Corazón/embriología , Miocardio/metabolismo , Animales , Desarrollo Embrionario/fisiología , Endotelio/embriología , Endotelio/metabolismo , Uniones Comunicantes/metabolismo , Ratones , Factores de Transcripción NFATC/metabolismo
16.
Mol Cell Biol ; 35(23): 3990-4005, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26391956

RESUMEN

p21-activated kinases (Paks) have been shown to regulate cytoskeleton rearrangements, cell proliferation, attachment, and migration in a variety of cellular contexts, including endothelial cells. However, the role of endothelial Pak in embryo development has not been reported, and currently, there is no consensus on the endothelial function of individual Pak isoforms, in particular p21-activated kinase 2 (Pak2), the main Pak isoform expressed in endothelial cells. In this work, we employ genetic and molecular studies that show that Pak2, but not Pak1, is a critical mediator of development and maintenance of endothelial cell function. Endothelial depletion of Pak2 leads to early embryo lethality due to flawed blood vessel formation in the embryo body and yolk sac. In adult endothelial cells, Pak2 depletion leads to severe apoptosis and acute angiogenesis defects, and in adult mice, endothelial Pak2 deletion leads to increased vascular permeability. Furthermore, ubiquitous Pak2 deletion is lethal in adult mice. We show that many of these defects are mediated through a newly unveiled Pak2/Bmk1 pathway. Our results demonstrate that endothelial Pak2 is essential during embryogenesis and also for adult blood vessel maintenance, and they also pinpoint the Bmk1/Erk5 pathway as a critical mediator of endothelial Pak2 signaling.


Asunto(s)
Endotelio/embriología , Endotelio/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Animales , Permeabilidad Capilar , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Sistema Cardiovascular/embriología , Sistema Cardiovascular/metabolismo , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Pérdida del Embrión , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Endotelio/citología , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Masculino , Ratones Endogámicos C57BL , Interferencia de ARN , Quinasas p21 Activadas/genética
17.
Development ; 142(17): 2951-61, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26253401

RESUMEN

Vessel formation has been extensively studied at the tissue level, but the difficulty in imaging the endothelium with cellular resolution has hampered study of the morphogenesis and behavior of endothelial cells (ECs) in vivo. We are using endothelial-specific transgenes and high-resolution imaging to examine single ECs in zebrafish. By generating mosaics with transgenes that simultaneously mark endothelial nuclei and membranes we are able to definitively identify and study the morphology and behavior of individual ECs during vessel sprouting and lumen formation. Using these methods, we show that developing trunk vessels are composed of ECs of varying morphology, and that single-cell analysis can be used to quantitate alterations in morphology and dynamics in ECs that are defective in proper guidance and patterning. Finally, we use single-cell analysis of intersegmental vessels undergoing lumen formation to demonstrate the coexistence of seamless transcellular lumens and single or multicellular enclosed lumens with autocellular or intercellular junctions, suggesting that heterogeneous mechanisms contribute to vascular lumen formation in vivo. The tools that we have developed for single EC analysis should facilitate further rigorous qualitative and quantitative analysis of EC morphology and behavior in vivo.


Asunto(s)
Endotelio/citología , Endotelio/embriología , Morfogénesis , Análisis de la Célula Individual/métodos , Pez Cebra/embriología , Animales , Polaridad Celular , Embrión no Mamífero/citología , Células Endoteliales/citología , Endotelio/irrigación sanguínea , Imagenología Tridimensional , Uniones Intercelulares , Espacio Intracelular/metabolismo , Fusión de Membrana , Neovascularización Fisiológica , Reproducibilidad de los Resultados , Torso/irrigación sanguínea , Torso/embriología , Vacuolas/metabolismo
18.
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
19.
Int J Mol Med ; 34(3): 669-76, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24969754

RESUMEN

Endothelial-mesenchymal transition (EndoMT) is a process in which endothelial cells lose their cell-type­specific characteristics and gain a mesenchymal cell phenotype. The Notch signaling pathway is crucial in the regulation of EndoMT; however, its roles have not been fully studied in vivo. In a previous study, we reported the generation of transgenic mice with a floxed ß-geo/stop signal between a CMV promoter and the constitutively active intracellular domain of Notch1 (IC-Notch1) linked with a human placental alkaline phosphatase (hPLAP) reporter (ZAP-IC-Notch1). In this study, we examined the results of activating IC-Notch1 in endothelial cells. ZAP-IC­Notch1 mice were crossed with Tie2-Cre mice to activate IC-Notch1 expression specifically in endothelial cells. The ZAP-IC-Notch1/Tie2-Cre double transgenic embryos died at E9.5-10.5 with disruption of vasculature and enlargement of myocardium. VE-cadherin expression was decreased and EphrinB2 expression was increased in the heart of these embryos. Mesenchymal cell marker α-smooth muscle actin (SMA) was expressed in IC-Notch1­expressing endothelial cells. In addition, upregulation of Snail, the key effector in mediating EndoMT, was identified in the cardiac cushion of the double transgenic murine embryo heart. The results of the present study demonstrate that constitutively active Notch signaling promotes EndoMT and differentially regulates endothelial/mesenchymal cell markers during cardiac development.


Asunto(s)
Endotelio/embriología , Mesodermo/embriología , Receptor Notch1/metabolismo , Transducción de Señal , Animales , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Endotelio/citología , Efrina-B2/metabolismo , Corazón/embriología , Humanos , Integrasas/metabolismo , Mesodermo/citología , Ratones Transgénicos , Modelos Animales , Estructura Terciaria de Proteína , Receptor Notch1/química , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
20.
Stem Cells Dev ; 23(17): 2004-13, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24762086

RESUMEN

Vasculogenesis/angiogenesis is one of the earliest processes that occurs during embryogenesis. ETV2 and SOX7 were previously shown to play a role in endothelial development; however, their mechanistic interaction has not been defined. In the present study, concomitant expression of Etv2 and Sox7 in endothelial progenitor cells was verified. ETV2 was shown to be a direct upstream regulator of Sox7 that binds to ETV2 binding elements in the Sox7 upstream regulatory region and activates transcription. We observed that SOX7 over-expression can mimic ETV2 and increase endothelial progenitor cells in embryonic bodies (EBs), while knockdown of Sox7 is able to block ETV2-induced increase in endothelial progenitor cell formation. Angiogenic sprouting was increased by ETV2 over-expression in EBs, and it was significantly decreased in the presence of Sox7 shRNA. Collectively, these studies support the conclusion that ETV2 directly regulates Sox7, and that ETV2 governs endothelial development by regulating transcriptional networks which include Sox7.


Asunto(s)
Sistema Cardiovascular/citología , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Sistema Cardiovascular/embriología , Sistema Cardiovascular/metabolismo , Células Cultivadas , Cuerpos Embrioides/metabolismo , Células Progenitoras Endoteliales/fisiología , Endotelio/citología , Endotelio/embriología , Endotelio/metabolismo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Factores de Transcripción SOXF/genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA