Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Biol ; 501: 92-103, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37353106

RESUMEN

During embryonic development, primitive and definitive waves of hematopoiesis take place to provide proper blood cells for each developmental stage, with the possible involvement of epigenetic factors. We previously found that lysine-specific demethylase 1 (LSD1/KDM1A) promotes primitive hematopoietic differentiation by shutting down the gene expression program of hemangioblasts in an Etv2/Etsrp-dependent manner. In the present study, we demonstrated that zebrafish LSD1 also plays important roles in definitive hematopoiesis in the development of hematopoietic stem and progenitor cells. A combination of genetic approaches and imaging analyses allowed us to show that LSD1 promotes the egress of hematopoietic stem and progenitor cells into the bloodstream during the endothelial-to-hematopoietic transition. Analysis of compound mutant lines with Etv2/Etsrp mutant zebrafish revealed that, unlike in primitive hematopoiesis, this function of LSD1 was independent of Etv2/Etsrp. The phenotype of LSD1 mutant zebrafish during the endothelial-to-hematopoietic transition was similar to that of previously reported compound knockout mice of Gfi1/Gfi1b, which forms a complex with LSD1 and represses endothelial genes. Moreover, co-knockdown of zebrafish Gfi1/Gfi1b genes inhibited the development of hematopoietic stem and progenitor cells. We therefore hypothesize that the shutdown of the Gfi1/Gfi1b-target genes during the endothelial-to-hematopoietic transition is one of the key evolutionarily conserved functions of LSD1 in definitive hematopoiesis.


Asunto(s)
Células Madre , Pez Cebra , Animales , Ratones , Diferenciación Celular , Hematopoyesis/genética , Histona Demetilasas/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Methods Mol Biol ; 2475: 313-323, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35451768

RESUMEN

Unlike humans, the zebrafish can repair and regenerate its heart following injury. Understanding the molecular and physiological mechanisms of heart regeneration is critical in developing pro-regenerative strategies for clinical application. The cardiac lymphatic and non-lymphatic vasculature both respond to injury in zebrafish and are instrumental in driving optimal repair and regeneration. However, progress has been impeded by an inability to obtain high resolution images to clearly visualize and thus to fully understand the vascular responses in the injured heart and how this might intersect with successful repair and regeneration in humans.In this chapter, we describe a chemical clearing approach using Clear Unobstructed Brain/Body Imaging Cocktails and Computational analysis (CUBIC), for obtaining high resolution images of the adult zebrafish heart. This approach permits three-dimensional reconstruction of cardiac vasculature throughout the entire organ. By applying CUBIC methodology to tissues from transgenic zebrafish reporter lines or in conjunction with immunofluorescent staining, optical slices can be be generated, negating the need for standard tissue processing and sectioning procedures and yielding higher resolution images. The resultant images enable a holistic view of the coronary blood and lymphatic vasculature during heart injury and regeneration. Herein, we describe our protocol for visualizing vessels in the adult zebrafish heart using these approaches.


Asunto(s)
Vasos Linfáticos , Pez Cebra , Animales , Animales Modificados Genéticamente , Corazón/diagnóstico por imagen , Corazón/fisiología , Imagenología Tridimensional , Vasos Linfáticos/diagnóstico por imagen , Pez Cebra/fisiología
3.
Development ; 147(8)2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32341028

RESUMEN

Runx1 is a transcription factor that plays a key role in determining the proliferative and differential state of multiple cell types, during both development and adulthood. Here, we report how Runx1 is specifically upregulated at the injury site during zebrafish heart regeneration, and that absence of runx1 results in increased myocardial survival and proliferation, and overall heart regeneration, accompanied by decreased fibrosis. Using single cell sequencing, we found that the wild-type injury site consists of Runx1-positive endocardial cells and thrombocytes that induce expression of smooth muscle and collagen genes. Both these populations cannot be identified in runx1 mutant wounds that contain less collagen and fibrin. The reduction in fibrin in the mutant is further explained by reduced myofibroblast formation and upregulation of components of the fibrin degradation pathway, including plasminogen receptor annexin 2A as well as downregulation of plasminogen activator inhibitor serpine1 in myocardium and endocardium, resulting in increased levels of plasminogen. Our findings suggest that Runx1 controls the regenerative response of multiple cardiac cell types and that targeting Runx1 is a novel therapeutic strategy for inducing endogenous heart repair.


Asunto(s)
Cicatriz/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Corazón/fisiopatología , Miocardio/patología , Regeneración , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Anexina A2/metabolismo , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Endocardio/patología , Regulación del Desarrollo de la Expresión Génica , Músculo Liso/metabolismo , Mutación/genética , Miofibroblastos/metabolismo , Miofibroblastos/patología , Cadenas Pesadas de Miosina/metabolismo , Regulación hacia Arriba/genética , Proteínas de Pez Cebra/genética
4.
Commun Biol ; 3(1): 71, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32054973

RESUMEN

Gata2 is a key transcription factor required to generate Haematopoietic Stem and Progenitor Cells (HSPCs) from haemogenic endothelium (HE); misexpression of Gata2 leads to haematopoietic disorders. Here we deleted a conserved enhancer (i4 enhancer) driving pan-endothelial expression of the zebrafish gata2a and showed that Gata2a is required for HE programming by regulating expression of runx1 and of the second Gata2 orthologue, gata2b. By 5 days, homozygous gata2aΔi4/Δi4 larvae showed normal numbers of HSPCs, a recovery mediated by Notch signalling driving gata2b and runx1 expression in HE. However, gata2aΔi4/Δi4 adults showed oedema, susceptibility to infections and marrow hypo-cellularity, consistent with bone marrow failure found in GATA2 deficiency syndromes. Thus, gata2a expression driven by the i4 enhancer is required for correct HE programming in embryos and maintenance of steady-state haematopoietic stem cell output in the adult. These enhancer mutants will be useful in exploring further the pathophysiology of GATA2-related deficiencies in vivo.


Asunto(s)
Reprogramación Celular/genética , Secuencia Conservada , Endotelio/metabolismo , Elementos de Facilitación Genéticos , Factor de Transcripción GATA2/genética , Hematopoyesis/genética , Eliminación de Secuencia , Factores de Edad , Animales , Secuencia de Bases , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Sitios Genéticos , Células Madre Hematopoyéticas/metabolismo , Pez Cebra
5.
Dev Cell ; 52(5): 574-590.e6, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32084358

RESUMEN

The epicardium is essential during cardiac development, homeostasis, and repair, and yet fundamental insights into its underlying cell biology, notably epicardium formation, lineage heterogeneity, and functional cross-talk with other cell types in the heart, are currently lacking. In this study, we investigated epicardial heterogeneity and the functional diversity of discrete epicardial subpopulations in the developing zebrafish heart. Single-cell RNA sequencing uncovered three epicardial subpopulations with specific genetic programs and distinctive spatial distribution. Perturbation of unique gene signatures uncovered specific functions associated with each subpopulation and established epicardial roles in cell adhesion, migration, and chemotaxis as a mechanism for recruitment of leukocytes into the heart. Understanding which mechanisms epicardial cells employ to establish a functional epicardium and how they communicate with other cardiovascular cell types during development will bring us closer to repairing cellular relationships that are disrupted during cardiovascular disease.


Asunto(s)
Linaje de la Célula , Pericardio/citología , Transcriptoma , Animales , Regulación del Desarrollo de la Expresión Génica , Pericardio/embriología , Pericardio/metabolismo , RNA-Seq , Análisis de la Célula Individual , Pez Cebra
6.
IUBMB Life ; 72(1): 39-44, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31778014

RESUMEN

GATA factors play central roles in the programming of blood and cardiac cells during embryonic development. Using the experimentally accessible Xenopus and zebrafish models, we report observations regarding the roles of GATA-2 in the development of blood stem cells and GATA-4, -5, and -6 in cardiac development. We show that blood stem cells develop from the dorsal lateral plate mesoderm and GATA-2 is required at multiple stages. Firstly, GATA-2 is required to make the cells responsive to VEGF-A signalling by driving the synthesis of its receptor, FLK-1/KDR. This leads to differentiation into the endothelial cells that form the dorsal aorta. GATA-2 is again required for the endothelial-to-haematopoietic transition that takes place later in the floor of the dorsal aorta. GATA-2 expression is dependent on BMP signalling for each of these inputs into blood stem cell programming. GATA-4, -5, and -6 work together to ensure the specification of cardiac cells during development. We have demonstrated redundancy within the family and also some evolution of the functions of the different family members. Interestingly, one of the features that varies in evolution is the timing of expression relative to other key regulators such as Nkx2.5 and BMP. We show that the GATA factors, Nkx2.5 and BMP regulate each other and it would appear that what is critical is the mutually supportive network of expression rather than the order of expression of each of the component genes. In Xenopus and zebrafish, the cardiac mesoderm is adjacent to an anterior population of cells giving rise to blood and endothelium. This population is not present in mammals and we have shown that, like the cardiac population, the blood and endothelial precursors require GATA-4, -5, and -6 for their development. Later, blood-specific or cardiac-specific regulators determine the ultimate fate of the cells, and we show that these regulators act cross-antagonistically. Fibroblast growth factor (FGF) signalling drives the cardiac fate, and we propose that the anterior extension of the FGF signalling field during evolution led to the recruitment of the blood and endothelial precursors into the heart field ultimately resulting in a larger four chambered heart. Zebrafish are able to successfully regenerate their hearts after injury. To understand the pathways involved, with a view to determining why humans cannot do this, we profiled gene expression in the cardiomyocytes before and after injury, and compared those proximal to the injury with those more distal. We were able to identify an enhancement of the expression of regulators of the canonical Wnt pathway proximal to the injury, suggesting that changes in Wnt signalling are responsible for the repair response to injury.


Asunto(s)
Sangre/metabolismo , Diferenciación Celular , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Miocitos Cardíacos/citología , Animales , Factores de Transcripción GATA/genética , Humanos , Miocitos Cardíacos/metabolismo
7.
Nat Commun ; 10(1): 3577, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31395869

RESUMEN

Haematopoietic stem cells are generated from the haemogenic endothelium (HE) located in the floor of the dorsal aorta (DA). Despite being integral to arteries, it is controversial whether HE and arterial endothelium share a common lineage. Here, we present a transgenic zebrafish runx1 reporter line to isolate HE and aortic roof endothelium (ARE)s, excluding non-aortic endothelium. Transcriptomic analysis of these populations identifies Runx1-regulated genes and shows that HE initially expresses arterial markers at similar levels to ARE. Furthermore, runx1 expression depends on prior arterial programming by the Notch ligand dll4. Runx1-/- mutants fail to downregulate arterial genes in the HE, which remains integrated within the DA, suggesting that Runx1 represses the pre-existing arterial programme in HE to allow progression towards the haematopoietic fate. These findings strongly suggest that, in zebrafish, aortic endothelium is a precursor to HE, with potential implications for pluripotent stem cell differentiation protocols for the generation of transplantable HSCs.


Asunto(s)
Arterias/embriología , Endotelio Vascular/embriología , Hemangioblastos/fisiología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Arterias/citología , Arterias/metabolismo , Linaje de la Célula , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Embrión no Mamífero , Desarrollo Embrionario , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
J Comput Biol ; 26(7): 719-725, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31140835

RESUMEN

Blood is an example of a highly regenerative tissue and its regeneration depends on the presence of stem cells residing in the bone marrow in humans. A better understanding of how these stem cells are programmed would benefit their use in clinical practice and shed light on the mechanisms by which the unique properties of stem cells are established. Our approach is to delineate the gene regulatory networks (GRNs) that specify these cells during their development in the embryo, and we use the amphibian experimental model because a wealth of evidence shows that the mechanisms used are conserved in mammals including humans. Blood stem cells are made during the intraembryonic wave of hematopoiesis during embryonic development where they emerge from endothelial precursors in the floor of the dorsal aorta (DA). These cells are derived from lateral plate mesoderm and so we have focused on the subset of cells in the lateral plate mesoderm fated to become blood and endothelium known as definitive hemangioblasts. We have found that their programming results from the activities of vascular endothelial growth factor A (VEGFA) and bone morphogenetic protein (BMP) signaling and the inhibition by miRNA of transforming growth factor beta signaling. VEGFA is first generated in the somites adjacent to the lateral plate mesoderm, and one of the responses of the lateral plate mesoderm is to activate endogenous VEGFA expression. BMP has multiple inputs into the programming of these cells via the activation of the transcription factor (TF), Gata2, and of the VEGFA receptor. These actions culminate in the expression of the leukemia-associated TF, Scl/Tal1, which is essential for blood fate specification. The activity of VEGFA in driving endothelial development resides in the small isoform, but the medium and large isoforms are required to initiate the blood stem cell program in the floor of the DA. The expression of the small isoform is dependent on the blood TF with leukemia connections, Tel1/Etv6, whereas the larger isoforms depend on another transcription-associated factor with leukemia connections, Eto2, raising the possibility that the regulation of VEGFA expression may be the mode of action of these leukemic factors. The action of Tel1/Etv6 in directly activating VEGFA expression in the somites was unexpected because this TF had only been reported to repress transcription. Using chromatin immunoprecipitation technology, we were able to show that Tel1/Etv6 does indeed work by repressing the expression of a VEGFA repressor, FoxC3, but it also acts directly to activate VEGFA expression, working together with Klf4. Finally, we have also looked at the mesodermal population that gives rise to the earlier waves of hematopoiesis, which do not generate a stem cell. We find significant differences including differential use of TFs of the E-Twenty-Six (ETS) family. In conclusion, we have elucidated the GRN responsible for preparing the lateral mesoderm for blood stem cell production.


Asunto(s)
Sangre/metabolismo , Redes Reguladoras de Genes , Regeneración/genética , Animales , Embrión de Mamíferos/metabolismo , Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
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
10.
PLoS Comput Biol ; 14(8): e1006077, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30157169

RESUMEN

The precise anatomical location of gene expression is an essential component of the study of gene function. For most model organisms this task is usually undertaken via visual inspection of gene expression images by interested researchers. Computational analysis of gene expression has been developed in several model organisms, notably in Drosophila which exhibits a uniform shape and outline in the early stages of development. Here we address the challenge of computational analysis of gene expression in Xenopus, where the range of developmental stages of interest encompasses a wide range of embryo size and shape. Embryos may have different orientation across images, and, in addition, embryos have a pigmented epidermis that can mask or confuse underlying gene expression. Here we report the development of a set of computational tools capable of processing large image sets with variable characteristics. These tools efficiently separate the Xenopus embryo from the background, separately identify both histochemically stained and naturally pigmented regions within the embryo, and can sort images from the same gene and developmental stage according to similarity of gene expression patterns without information about relative orientation. We tested these methods on a large, but highly redundant, collection of 33,289 in situ hybridization images, allowing us to select representative images of expression patterns at different embryo orientations. This has allowed us to put a much smaller subset of these images into the public domain in an effective manner. The 'isimage' module and the scripts developed are implemented in Python and freely available on https://pypi.python.org/pypi/isimage/.


Asunto(s)
Biología Computacional/métodos , Curaduría de Datos/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Animales , Embrión no Mamífero/metabolismo , Expresión Génica , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ/métodos , Hibridación Fluorescente in Situ/métodos , Programas Informáticos , Transcriptoma , Xenopus laevis/embriología
11.
Biol Open ; 7(4)2018 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-29535102

RESUMEN

Advances in genome engineering have resulted in the generation of numerous zebrafish mutant lines. A commonly used method to assess gene expression in the mutants is in situ hybridisation (ISH). Because the embryos can be distinguished by genotype after ISH, comparing gene expression between wild-type and mutant siblings can be done blinded and in parallel. Such experimental design reduces the technical variation between samples and minimises the risk of bias. This approach, however, requires an efficient method of genomic DNA extraction from post-ISH fixed zebrafish samples to ascribe phenotype to genotype. Here we describe a method to obtain PCR-quality DNA from 95-100% of zebrafish embryos, suitable for genotyping after ISH. In addition, we provide an image analysis protocol for quantifying gene expression of ISH-probed embryos, adaptable for the analysis of different expression patterns. Finally, we show that intensity-based image analysis enables accurate representation of the variability of gene expression detected by ISH and that it can complement quantitative methods like qRT-PCR. By combining genotyping after ISH and computer-based image analysis, we have established a high-confidence, unbiased methodology to assign gene expression levels to specific genotypes, and applied it to the analysis of molecular phenotypes of newly generated lmo4a mutants.

12.
Proc Natl Acad Sci U S A ; 114(23): 5814-5821, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28584091

RESUMEN

Hematopoietic stem cells (HSCs) that sustain lifelong blood production are created during embryogenesis. They emerge from a specialized endothelial population, termed hemogenic endothelium (HE), located in the ventral wall of the dorsal aorta (DA). In Xenopus, we have been studying the gene regulatory networks (GRNs) required for the formation of HSCs, and critically found that the hemogenic potential is defined at an earlier time point when precursors to the DA express hematopoietic as well as endothelial genes, in the definitive hemangioblasts (DHs). The GRN for DH programming has been constructed and, here, we show that bone morphogenetic protein (BMP) signaling is essential for the initiation of this GRN. BMP2, -4, and -7 are the principal ligands expressed in the lineage forming the HE. To investigate the requirement and timing of all BMP signaling in HSC ontogeny, we have used a transgenic line, which inducibly expresses an inhibitor of BMP signaling, Noggin, as well as a chemical inhibitor of BMP receptors, DMH1, and described the inputs from BMP signaling into the DH GRN and the HE, as well as into primitive hematopoiesis. BMP signaling is required in at least three points in DH programming: first to initiate the DH GRN through gata2 expression, then for kdr expression to enable the DH to respond to vascular endothelial growth factor A (VEGFA) ligand from the somites, and finally for gata2 expression in the DA, but is dispensable for HE specification after hemangioblasts have been formed.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/genética , Linaje de la Célula , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/citología , Animales , Animales Modificados Genéticamente , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Xenopus laevis
13.
Sci Rep ; 7(1): 2917, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28592901

RESUMEN

The adult zebrafish is a well-established model for studying heart regeneration, but due to its tissue opaqueness, repair has been primarily assessed using destructive histology, precluding repeated investigations of the same animal. We present a high-resolution, non-invasive in vivo magnetic resonance imaging (MRI) method incorporating a miniature respiratory and anaesthetic perfusion set-up for live adult zebrafish, allowing for visualization of scar formation and heart regeneration in the same animal over time at an isotropic 31 µm voxel resolution. To test the method, we compared well and poorly healing cardiac ventricles using a transgenic fish model that exhibits heat-shock (HS) inducible impaired heart regeneration. HS-treated groups revealed persistent scar tissue for 10 weeks, while control groups were healed after 4 weeks. Application of the advanced MRI technique allowed clear discrimination of levels of repair following cryo- and resection injury for several months. It further provides a novel tool for in vivo time-lapse imaging of adult fish for non-cardiac studies, as the method can be readily applied to image wound healing in other injured or diseased tissues, or to monitor tissue changes over time, thus expanding the range of questions that can be addressed in adult zebrafish and other small aquatic species.


Asunto(s)
Corazón/diagnóstico por imagen , Corazón/fisiología , Imagen por Resonancia Magnética , Regeneración , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Cardiopatías/diagnóstico por imagen , Cardiopatías/patología , Pez Cebra
14.
Nat Immunol ; 17(12): 1424-1435, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27695000

RESUMEN

The final stages of restriction to the T cell lineage occur in the thymus after the entry of thymus-seeding progenitors (TSPs). The identity and lineage potential of TSPs remains unclear. Because the first embryonic TSPs enter a non-vascularized thymic rudiment, we were able to directly image and establish the functional and molecular properties of embryonic thymopoiesis-initiating progenitors (T-IPs) before their entry into the thymus and activation of Notch signaling. T-IPs did not include multipotent stem cells or molecular evidence of T cell-restricted progenitors. Instead, single-cell molecular and functional analysis demonstrated that most fetal T-IPs expressed genes of and had the potential to develop into lymphoid as well as myeloid components of the immune system. Moreover, studies of embryos deficient in the transcriptional regulator RBPJ demonstrated that canonical Notch signaling was not involved in pre-thymic restriction to the T cell lineage or the migration of T-IPs.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Células Progenitoras Linfoides/fisiología , Células Progenitoras Mieloides/fisiología , Receptores Notch/metabolismo , Linfocitos T/fisiología , Timo/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Células Cultivadas , Feto , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
15.
Dev Cell ; 38(4): 358-70, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27499523

RESUMEN

Hematopoietic stem cells (HSCs) are self-renewing multipotent stem cells that generate mature blood lineages throughout life. They, together with hematopoietic progenitor cells (collectively known as HSPCs), emerge from hemogenic endothelium in the floor of the embryonic dorsal aorta by an endothelial-to-hematopoietic transition (EHT). Here we demonstrate that transforming growth factor ß (TGFß) is required for HSPC specification and that it regulates the expression of the Notch ligand Jagged1a in endothelial cells prior to EHT, in a striking parallel with the epithelial-to-mesenchymal transition (EMT). The requirement for TGFß is two fold and sequential: autocrine via Tgfß1a and Tgfß1b produced in the endothelial cells themselves, followed by a paracrine input of Tgfß3 from the notochord, suggesting that the former programs the hemogenic endothelium and the latter drives EHT. Our findings have important implications for the generation of HSPCs from pluripotent cells in vitro.


Asunto(s)
Endotelio Vascular/embriología , Células Madre Hematopoyéticas/citología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Transición Epitelial-Mesenquimal , Proteína Jagged-1/biosíntesis , Proteína Jagged-1/genética , Morfolinos/genética , Células Madre Multipotentes/citología , Notocorda/embriología , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta3/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
FEBS Lett ; 590(22): 4002-4015, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27531714

RESUMEN

Haematopoietic stem cells (HSCs) emerge from the haemogenic endothelium (HE) localised in the ventral wall of the embryonic dorsal aorta (DA). The HE generates HSCs through a process known as the endothelial to haematopoietic transition (EHT), which has been visualised in live embryos and is currently under intense study. However, EHT is the culmination of multiple programming events, which are as yet poorly understood, that take place before the specification of HE. A number of haematopoietic precursor cells have been described before the emergence of definitive HSCs, but only one haematovascular progenitor, the definitive haemangioblast (DH), gives rise to the DA, HE and HSCs. DHs emerge in the lateral plate mesoderm (LPM) and have a distinct origin and genetic programme compared to other, previously described haematovascular progenitors. Although DHs have so far only been established in Xenopus embryos, evidence for their existence in the LPM of mouse and chicken embryos is discussed here. We also review the current knowledge of the origins, lineage relationships, genetic programming and differentiation of the DHs that leads to the generation of HSCs. Importantly, we discuss the significance of the gene regulatory network (GRN) that controls the programming of DHs, a better understanding of which may aid in the establishment of protocols for the de novo generation of HSCs in vitro.


Asunto(s)
Desarrollo Embrionario/genética , Células Madre Hematopoyéticas , Mesodermo/crecimiento & desarrollo , Xenopus laevis/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Embrión de Pollo , Embrión de Mamíferos , Embrión no Mamífero , Hemangioblastos/metabolismo , Mesodermo/embriología , Ratones , Xenopus laevis/embriología
17.
Nucleic Acids Res ; 44(14): 6693-706, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27084945

RESUMEN

DNA methylation is a repressive epigenetic modification that covers vertebrate genomes. Regions known as CpG islands (CGIs), which are refractory to DNA methylation, are often associated with gene promoters and play central roles in gene regulation. Yet how CGIs in their normal genomic context evade the DNA methylation machinery and whether these mechanisms are evolutionarily conserved remains enigmatic. To address these fundamental questions we exploited a transchromosomic animal model and genomic approaches to understand how the hypomethylated state is formed in vivo and to discover whether mechanisms governing CGI formation are evolutionarily conserved. Strikingly, insertion of a human chromosome into mouse revealed that promoter-associated CGIs are refractory to DNA methylation regardless of host species, demonstrating that DNA sequence plays a central role in specifying the hypomethylated state through evolutionarily conserved mechanisms. In contrast, elements distal to gene promoters exhibited more variable methylation between host species, uncovering a widespread dependence on nucleotide frequency and occupancy of DNA-binding transcription factors in shaping the DNA methylation landscape away from gene promoters. This was exemplified by young CpG rich lineage-restricted repeat sequences that evaded DNA methylation in the absence of co-evolved mechanisms targeting methylation to these sequences, and species specific DNA binding events that protected against DNA methylation in CpG poor regions. Finally, transplantation of mouse chromosomal fragments into the evolutionarily distant zebrafish uncovered the existence of a mechanistically conserved and DNA-encoded logic which shapes CGI formation across vertebrate species.


Asunto(s)
Secuencia Conservada/genética , Islas de CpG/genética , Metilación de ADN/genética , ADN/genética , Evolución Molecular , Animales , Línea Celular , Cromosomas Humanos Par 21/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Unión Proteica/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Especificidad de la Especie , Factores de Transcripción/metabolismo , Vertebrados/genética
18.
Open Biol ; 5(8)2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26289800

RESUMEN

Vertebrate NCoR-family co-repressors play central roles in the timing of embryo and stem cell differentiation by repressing the activity of a range of transcription factors. They interact with nuclear receptors using short linear motifs (SLiMs) termed co-repressor for nuclear receptor (CoRNR) boxes. Here, we identify the pathway leading to increasing co-repressor diversity across the deuterostomes. The final complement of CoRNR boxes arose in an ancestral cephalochordate, and was encoded in one large exon; the urochordates and vertebrates then split this region between 10 and 12 exons. In Xenopus, alternative splicing is prevalent in NCoR2, but absent in NCoR1. We show for one NCoR1 exon that alternative splicing can be recovered by a single point mutation, suggesting NCoR1 lost the capacity for alternative splicing. Analyses in Xenopus and zebrafish identify that cellular context, rather than gene sequence, predominantly determines species differences in alternative splicing. We identify a pathway to diversity for the NCoR family beginning with the addition of a SLiM, followed by gene duplication, the generation of alternatively spliced isoforms and their differential deployment.


Asunto(s)
Empalme Alternativo , Secuencias de Aminoácidos , Proteínas Co-Represoras/química , Proteínas Co-Represoras/genética , Exones , Animales , Secuencia de Bases , Secuencia Conservada , Datos de Secuencia Molecular , Co-Represor 1 de Receptor Nuclear/química , Co-Represor 1 de Receptor Nuclear/genética , Posición Específica de Matrices de Puntuación , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Xenopus laevis/genética
19.
PLoS Biol ; 13(2): e1002051, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25665164

RESUMEN

As some of the most widely utilised intercellular signalling molecules, transforming growth factor ß (TGFß) superfamily members play critical roles in normal development and become disrupted in human disease. Establishing appropriate levels of TGFß signalling involves positive and negative feedback, which are coupled and driven by the same signal transduction components (R-Smad transcription factor complexes), but whether and how the regulation of the two can be distinguished are unknown. Genome-wide comparison of published ChIP-seq datasets suggests that LIM domain binding proteins (Ldbs) co-localise with R-Smads at a substantial subset of R-Smad target genes including the locus of inhibitory Smad7 (I-Smad7), which mediates negative feedback for TGFß signalling. We present evidence suggesting that zebrafish Ldb2a binds and directly activates the I-Smad7 gene, whereas it binds and represses the ligand gene, Squint (Sqt), which drives positive feedback. Thus, the fine tuning of TGFß signalling derives from positive and negative control by Ldb2a. Expression of ldb2a is itself activated by TGFß signals, suggesting potential feed-forward loops that might delay the negative input of Ldb2a to the positive feedback, as well as the positive input of Ldb2a to the negative feedback. In this way, precise gene expression control by Ldb2a enables an initial build-up of signalling via a fully active positive feedback in the absence of buffering by the negative feedback. In Ldb2a-deficient zebrafish embryos, homeostasis of TGFß signalling is perturbed and signalling is stably enhanced, giving rise to excess mesoderm and endoderm, an effect that can be rescued by reducing signalling by the TGFß family members, Nodal and BMP. Thus, Ldb2a is critical to the homeostatic control of TGFß signalling and thereby embryonic patterning.


Asunto(s)
Tipificación del Cuerpo/genética , Retroalimentación Fisiológica , Proteínas con Dominio LIM/genética , Ligandos de Señalización Nodal/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Animales , Secuencia de Bases , Embrión no Mamífero , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas con Dominio LIM/antagonistas & inhibidores , Proteínas con Dominio LIM/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Microinyecciones , Datos de Secuencia Molecular , Morfolinos/genética , Morfolinos/metabolismo , Ligandos de Señalización Nodal/genética , Alineación de Secuencia , Transducción de Señal , Proteína smad7/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/genética , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/deficiencia
20.
Nat Commun ; 5: 5588, 2014 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-25429520

RESUMEN

Haematopoietic stem cells (HSCs) are produced during embryogenesis from the floor of the dorsal aorta. The localization of HSCs is dependent on the presence of instructive signals on the ventral side of the vessel. The nature of the extrinsic molecular signals that control the aortic haematopoietic niche is currently poorly understood. Here we demonstrate a novel requirement for FGF signalling in the specification of aortic haemogenic endothelium. Our results demonstrate that FGF signalling normally acts to repress BMP activity in the subaortic mesenchyme through transcriptional inhibition of bmp4, as well as through activation of two BMP antagonists, noggin2 and gremlin1a. Taken together, these findings demonstrate a key role for FGF signalling in establishment of the developmental HSC niche via its regulation of BMP activity in the subaortic mesenchyme. These results should help inform strategies to recapitulate the development of HSCs in vitro from pluripotent precursors.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Proteínas Portadoras/genética , Diferenciación Celular , Endotelio Vascular/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas de Pez Cebra/genética , Animales , Aorta/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/metabolismo , Mesodermo/metabolismo , Transducción de Señal , Nicho de Células Madre , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...