Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 147(3): 577-89, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036566

RESUMEN

BMP and Wnt signaling pathways control essential cellular responses through activation of the transcription factors SMAD (BMP) and TCF (Wnt). Here, we show that regeneration of hematopoietic lineages following acute injury depends on the activation of each of these signaling pathways to induce expression of key blood genes. Both SMAD1 and TCF7L2 co-occupy sites with master regulators adjacent to hematopoietic genes. In addition, both SMAD1 and TCF7L2 follow the binding of the predominant lineage regulator during differentiation from multipotent hematopoietic progenitor cells to erythroid cells. Furthermore, induction of the myeloid lineage regulator C/EBPα in erythroid cells shifts binding of SMAD1 to sites newly occupied by C/EBPα, whereas expression of the erythroid regulator GATA1 directs SMAD1 loss on nonerythroid targets. We conclude that the regenerative response mediated by BMP and Wnt signaling pathways is coupled with the lineage master regulators to control the gene programs defining cellular identity.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Hematopoyesis , Transducción de Señal , Vía de Señalización Wnt , Animales , Proteínas de Unión al ADN/metabolismo , Humanos , Regeneración , Proteína Smad1/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Pez Cebra
2.
Cell ; 142(1): 133-43, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20603019

RESUMEN

Recent genome-wide studies have demonstrated that pausing of RNA polymerase II (Pol II) occurred on many vertebrate genes. By genetic studies in the zebrafish tif1gamma mutant moonshine we found that loss of function of Pol II-associated factors PAF or DSIF rescued erythroid gene transcription in tif1gamma-deficient animals. Biochemical analysis established physical interactions among TIF1gamma, the blood-specific SCL transcription complex, and the positive elongation factors p-TEFb and FACT. Chromatin immunoprecipitation assays in human CD34(+) cells supported a TIF1gamma-dependent recruitment of positive elongation factors to erythroid genes to promote transcription elongation by counteracting Pol II pausing. Our study establishes a mechanism for regulating tissue cell fate and differentiation through transcription elongation.


Asunto(s)
Eritropoyesis , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Línea Celular Tumoral , Células Cultivadas , Células Eritroides/metabolismo , Humanos , ARN Polimerasa II/metabolismo , Pez Cebra/metabolismo
3.
Dev Biol ; 373(2): 422-30, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23159334

RESUMEN

Transcriptional regulators play critical roles in the regulation of cell fate during hematopoiesis. Previous studies in zebrafish have identified an essential role for the transcriptional intermediary factor TIF1γ in erythropoiesis by regulating the transcription elongation of erythroid genes. To study if TIF1γ plays a similar role in murine erythropoiesis and to assess its function in other blood lineages, we generated mouse models with hematopoietic deletion of TIF1γ. Our results showed a block in erythroid maturation in the bone marrow following tif1γ deletion that was compensated with enhanced spleen erythropoiesis. Further analyses revealed a defect in transcription elongation of erythroid genes in the bone marrow. In addition, loss of TIF1γ resulted in defects in other blood compartments, including a profound loss of B cells, a dramatic expansion of granulocytes and decreased HSC function. TIF1γ exerts its functions in a cell-autonomous manner as revealed by competitive transplantation experiments. Our study therefore demonstrates that TIF1γ plays essential roles in multiple murine blood lineages and that its function in transcription elongation is evolutionally conserved.


Asunto(s)
Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Elongación de la Transcripción Genética , Factores de Transcripción/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Eritroides/citología , Eliminación de Gen , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Granulocitos/citología , Granulocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mielopoyesis/genética , Bazo/metabolismo , Factores de Transcripción/deficiencia
4.
Dev Biol ; 366(2): 185-94, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22537494

RESUMEN

Globin gene switching is a complex, highly regulated process allowing expression of distinct globin genes at specific developmental stages. Here, for the first time, we have characterized all of the zebrafish globins based on the completed genomic sequence. Two distinct chromosomal loci, termed major (chromosome 3) and minor (chromosome 12), harbor the globin genes containing α/ß pairs in a 5'-3' to 3'-5' orientation. Both these loci share synteny with the mammalian α-globin locus. Zebrafish globin expression was assayed during development and demonstrated two globin switches, similar to human development. A conserved regulatory element, the locus control region (LCR), was revealed by analyzing DNase I hypersensitive sites, H3K4 trimethylation marks and GATA1 binding sites. Surprisingly, the position of these sites with relation to the globin genes is evolutionarily conserved, despite a lack of overall sequence conservation. Motifs within the zebrafish LCR include CACCC, GATA, and NFE2 sites, suggesting functional interactions with known transcription factors but not the same LCR architecture. Functional homology to the mammalian α-LCR MCS-R2 region was confirmed by robust and specific reporter expression in erythrocytes of transgenic zebrafish. Our studies provide a comprehensive characterization of the zebrafish globin loci and clarify the regulation of globin switching.


Asunto(s)
Globinas/genética , Región de Control de Posición/genética , Pez Cebra/genética , Animales , Embrión no Mamífero/embriología , Embrión no Mamífero/fisiología , Factor de Transcripción GATA1/genética , Regulación del Desarrollo de la Expresión Génica , Genes de Cambio , Subunidad p45 del Factor de Transcripción NF-E2/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
5.
Nat Commun ; 6: 8146, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26306682

RESUMEN

The vertebrate heart muscle (myocardium) develops from the first heart field (FHF) and expands by adding second heart field (SHF) cells. While both lineages exist already in teleosts, the primordial contributions of FHF and SHF to heart structure and function remain incompletely understood. Here we delineate the functional contribution of the FHF and SHF to the zebrafish heart using the cis-regulatory elements of the draculin (drl) gene. The drl reporters initially delineate the lateral plate mesoderm, including heart progenitors. Subsequent myocardial drl reporter expression restricts to FHF descendants. We harnessed this unique feature to uncover that loss of tbx5a and pitx2 affect relative FHF versus SHF contributions to the heart. High-resolution physiology reveals distinctive electrical properties of each heart field territory that define a functional boundary within the single zebrafish ventricle. Our data establish that the transcriptional program driving cardiac septation regulates physiologic ventricle partitioning, which successively provides mechanical advantages of sequential contraction.


Asunto(s)
Atrios Cardíacos/embriología , Ventrículos Cardíacos/embriología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Cadherinas/genética , Cadherinas/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Elementos Reguladores de la Transcripción/genética , Proteínas y Péptidos Salivales/genética , Proteínas y Péptidos Salivales/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
6.
Elife ; 42015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25714926

RESUMEN

Angiopoietin-like proteins (angptls) are capable of ex vivo expansion of mouse and human hematopoietic stem and progenitor cells (HSPCs). Despite this intriguing ability, their mechanism is unknown. In this study, we show that angptl2 overexpression is sufficient to expand definitive HSPCs in zebrafish embryos. Angptl1/2 are required for definitive hematopoiesis and vascular specification of the hemogenic endothelium. The loss-of-function phenotype is reminiscent of the notch mutant mindbomb (mib), and a strong genetic interaction occurs between angptls and notch. Overexpressing angptl2 rescues mib while overexpressing notch rescues angptl1/2 morphants. Gene expression studies in ANGPTL2-stimulated CD34(+) cells showed a strong MYC activation signature and myc overexpression in angptl1/2 morphants or mib restored HSPCs formation. ANGPTL2 can increase NOTCH activation in cultured cells and ANGPTL receptor interacted with NOTCH to regulate NOTCH cleavage. Together our data provide insight to the angptl-mediated notch activation through receptor interaction and subsequent activation of myc targets.


Asunto(s)
Angiopoyetinas/genética , Células Madre Hematopoyéticas/metabolismo , Receptores Notch/genética , Transducción de Señal/genética , Proteínas de Pez Cebra/genética , Proteína 1 Similar a la Angiopoyetina , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/metabolismo , Animales , Animales Modificados Genéticamente , Western Blotting , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Hematopoyesis/genética , Humanos , Células K562 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Microscopía Confocal , Unión Proteica , Interferencia de ARN , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Imagen de Lapso de Tiempo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
7.
PLoS One ; 8(5): e64969, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23705022

RESUMEN

Epigenetics, or the reversible and heritable marks of gene regulation not including DNA sequence, encompasses chromatin modifications on both the DNA and histones and is as important as the DNA sequence itself. Chromatin-modifying factors are playing an increasingly important role in tumorigenesis, particularly among pediatric rhabdomyosarcomas (RMS), revealing potential novel therapeutic targets. We performed an overexpression screen of chromatin-modifying factors in a KRAS(G12D)-driven zebrafish model for RMS. Here, we describe the identification of a histone H3 lysine 9 histone methyltransferase, SUV39H1, as a suppressor of embryonal RMS formation in zebrafish. This suppression is specific to the histone methyltransferase activity of SUV39H1, as point mutations in the SET domain lacked the effect. SUV39H1-overexpressing and control tumors have a similar proliferation rate, muscle differentiation state, and tumor growth rate. Strikingly, SUV39H1-overexpressing fish initiate fewer tumors, which results in the observed suppressive phenotype. We demonstrate that the delayed tumor onset occurs between 5 and 7 days post fertilization. Gene expression profiling at these stages revealed that in the context of KRAS(G12D) overexpression, SUV39H1 may suppress cell cycle progression. Our studies provide evidence for the role of SUV39H1 as a tumor suppressor.


Asunto(s)
Carcinogénesis/patología , Genes Supresores , Metiltransferasas/metabolismo , Rabdomiosarcoma Embrionario/enzimología , Rabdomiosarcoma Embrionario/patología , Proteínas de Pez Cebra/metabolismo , Animales , Carcinogénesis/genética , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metiltransferasas/química , Metiltransferasas/genética , Músculos/enzimología , Músculos/patología , Estructura Terciaria de Proteína , Rabdomiosarcoma Embrionario/genética , Pez Cebra , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
8.
Stem Cell Reports ; 1(5): 425-36, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24286030

RESUMEN

Deletion of caudal/cdx genes alters hox gene expression and causes defects in posterior tissues and hematopoiesis. Yet, the defects in hox gene expression only partially explain these phenotypes. To gain deeper insight into Cdx4 function, we performed chromatin immunoprecipitation sequencing (ChIP-seq) combined with gene-expression profiling in zebrafish, and identified the transcription factor spalt-like 4 (sall4) as a Cdx4 target. ChIP-seq revealed that Sall4 bound to its own gene locus and the cdx4 locus. Expression profiling showed that Cdx4 and Sall4 coregulate genes that initiate hematopoiesis, such as hox, scl, and lmo2. Combined cdx4/sall4 gene knockdown impaired erythropoiesis, and overexpression of the Cdx4 and Sall4 target genes scl and lmo2 together rescued the erythroid program. These findings suggest that auto- and cross-regulation of Cdx4 and Sall4 establish a stable molecular circuit in the mesoderm that facilitates the activation of the blood-specific program as development proceeds.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , Proteínas de Homeodominio/metabolismo , Mesodermo/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas de Homeodominio/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Mesodermo/citología , Factores de Transcripción/genética , Pez Cebra , Proteínas de Pez Cebra/genética
9.
Nat Cell Biol ; 15(12): 1516-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24240475

RESUMEN

The initiation of cellular programs is orchestrated by key transcription factors and chromatin regulators that activate or inhibit target gene expression. To generate a compendium of chromatin factors that establish the epigenetic code during developmental haematopoiesis, a large-scale reverse genetic screen was conducted targeting orthologues of 425 human chromatin factors in zebrafish. A set of chromatin regulators was identified that target different stages of primitive and definitive blood formation, including factors not previously implicated in haematopoiesis. We identified 15 factors that regulate development of primitive erythroid progenitors and 29 factors that regulate development of definitive haematopoietic stem and progenitor cells. These chromatin factors are associated with SWI/SNF and ISWI chromatin remodelling, SET1 methyltransferase, CBP-p300-HBO1-NuA4 acetyltransferase, HDAC-NuRD deacetylase, and Polycomb repressive complexes. Our work provides a comprehensive view of how specific chromatin factors and their associated complexes play a major role in the establishment of haematopoietic cells in vivo.


Asunto(s)
Hematopoyesis/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Células Eritroides/metabolismo , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/fisiología , Humanos , Morfolinos/genética , Mapas de Interacción de Proteínas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Genética Inversa , Proteínas de Pez Cebra/metabolismo
10.
Dev Cell ; 22(3): 625-38, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22421047

RESUMEN

In mammalian embryonic stem cells, the acquisition of pluripotency is dependent on Nanog, but the in vivo analysis of Nanog has been hampered by its requirement for early mouse development. In an effort to examine the role of Nanog in vivo, we identified a zebrafish Nanog ortholog and found that its knockdown impaired endoderm formation. Genome-wide transcription analysis revealed that nanog-like morphants fail to develop the extraembryonic yolk syncytial layer (YSL), which produces Nodal, required for endoderm induction. We examined the genes that were regulated by Nanog-like and identified the homeobox gene mxtx2, which is both necessary and sufficient for YSL induction. Chromatin immunoprecipitation assays and genetic studies indicated that Nanog-like directly activates mxtx2, which, in turn, specifies the YSL lineage by directly activating YSL genes. Our study identifies a Nanog-like-Mxtx2-Nodal pathway and establishes a role for Nanog-like in regulating the formation of the extraembryonic tissue required for endoderm induction.


Asunto(s)
Endodermo/metabolismo , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos de Señalización Nodal/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Datos de Secuencia Molecular , Proteína Homeótica Nanog , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
11.
Methods Cell Biol ; 104: 341-52, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21924172

RESUMEN

Zebrafish has been used for many years as a model to study development and disease. The ability of zebrafish to produce thousand of embryos in a synchronous manner has made zebrafish an invaluable tool for genetic and chemical screens. Since its emergence as an important model organism the molecular tools for studying zebrafish have been limited. In this chapter, we describe a simple method to identify DNA binding sites and chromatin architecture in erythrocytes from adult zebrafish using chromatin immunoprecipitation coupled with next generation sequencing. This technique has been used extensively and successfully in other systems and it will be a useful tool for studying epigenetics in zebrafish.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Pez Cebra/genética , Animales , Cromatina/metabolismo , Reactivos de Enlaces Cruzados/química , ADN/genética , ADN/aislamiento & purificación , ADN/metabolismo , Epigénesis Genética , Eritrocitos/metabolismo , Formaldehído/química , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Histonas/metabolismo , Unión Proteica , Análisis de Secuencia de ADN/métodos , Sonicación/métodos , Fijación del Tejido/métodos , Factores de Transcripción/metabolismo
12.
Dev Dyn ; 235(3): 747-53, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16372332

RESUMEN

This correspondence is a primer for the zebrafish research community on zebrafish tracks available in the UCSC Genome Browser at http://genome.ucsc.edu based on Sanger's Zv4 assembly. A primary capability of this facility is comparative informatics between humans (as well as many other model organisms) and zebrafish. The zebrafish genome sequencing project has played important roles in mutant mapping and cloning, and comparative genomic research projects. This easy-to-use genome browser aims to display and download useful genome sequence information for zebrafish mutant mapping and cloning projects. Its user-friendly interface expedites annotation of the zebrafish genome sequence.


Asunto(s)
Biología Computacional , Bases de Datos Genéticas , Genoma , Programas Informáticos , Pez Cebra/genética , Animales , Genómica , Humanos , Ratones , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína
13.
Dev Biol ; 281(2): 256-69, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15893977

RESUMEN

The Lmo2 transcription factor, a T-cell oncoprotein, is required for both hematopoiesis and angiogenesis. To investigate the fate of lmo2-expressing cells and the transcriptional regulation of lmo2 in vivo, we generated stable transgenic zebrafish that express green fluorescent protein (EGFP) or DsRed under the control of an lmo2 promoter. A 2.5-kb fragment contains the cis-regulatory elements required to recapitulate endogenous lmo2 expression in embryonic hematopoietic and vascular tissues. We further characterized embryonic Lmo2+ cells through transplantation into vlad tepes (vlt), an erythropoietic mutant. These Lmo2+ primitive wave donor cells differentiated into circulating hematopoietic cells and extended the life span of vlt recipients, but did not demonstrate long-term repopulation of the erythroid lineage. Promoter analysis identified a 174-bp proximal promoter that was sufficient to recapitulate lmo2 expression. This element contains critical ETS-binding sites conserved between zebrafish and pufferfish. Furthermore, we show that ets1 is coexpressed with lmo2, and overexpression experiments indicate that ets1 can activate the lmo2 promoter through this element. Our studies elucidate the transcriptional regulation of this key transcription factor, and provide a transgenic system for the functional analysis of blood and blood vessels in zebrafish.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Sistema Hematopoyético/embriología , Metaloproteínas/biosíntesis , Neovascularización Fisiológica/fisiología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Diferenciación Celular , Proteínas de Unión al ADN/genética , Eritrocitos/metabolismo , Eritrocitos/fisiología , Células Madre Hematopoyéticas/fisiología , Sistema Hematopoyético/citología , Sistema Hematopoyético/metabolismo , Proteínas con Dominio LIM , Metaloproteínas/genética , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-ets-1/biosíntesis , Factores de Transcripción , Pez Cebra/genética , Proteínas de Pez Cebra
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda