Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 588(7837): 337-343, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33239788

RESUMEN

The zebrafish (Danio rerio) has been widely used in the study of human disease and development, and about 70% of the protein-coding genes are conserved between the two species1. However, studies in zebrafish remain constrained by the sparse annotation of functional control elements in the zebrafish genome. Here we performed RNA sequencing, assay for transposase-accessible chromatin using sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing, whole-genome bisulfite sequencing, and chromosome conformation capture (Hi-C) experiments in up to eleven adult and two embryonic tissues to generate a comprehensive map of transcriptomes, cis-regulatory elements, heterochromatin, methylomes and 3D genome organization in the zebrafish Tübingen reference strain. A comparison of zebrafish, human and mouse regulatory elements enabled the identification of both evolutionarily conserved and species-specific regulatory sequences and networks. We observed enrichment of evolutionary breakpoints at topologically associating domain boundaries, which were correlated with strong histone H3 lysine 4 trimethylation (H3K4me3) and CCCTC-binding factor (CTCF) signals. We performed single-cell ATAC-seq in zebrafish brain, which delineated 25 different clusters of cell types. By combining long-read DNA sequencing and Hi-C, we assembled the sex-determining chromosome 4 de novo. Overall, our work provides an additional epigenomic anchor for the functional annotation of vertebrate genomes and the study of evolutionarily conserved elements of 3D genome organization.


Asunto(s)
Genoma/genética , Imagenología Tridimensional , Imagen Molecular , Secuencias Reguladoras de Ácidos Nucleicos/genética , Pez Cebra/genética , Animales , Encéfalo/metabolismo , Secuencia Conservada/genética , Metilación de ADN , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Evolución Molecular , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/genética , Heterocromatina/química , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Masculino , Ratones , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Análisis de la Célula Individual , Especificidad de la Especie
2.
J Cell Mol Med ; 26(3): 764-775, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35019227

RESUMEN

Skin can be mechanically stimulated to grow through a clinical procedure called tissue expansion (TE). Using a porcine TE model, we determined that expansion promptly activates transcription of SFRP2 in skin and we revealed that in the epidermis, this protein is secreted by Langerhans cells (LCs). Similar to well-known mechanosensitive genes, the increase in SFRP2 expression was proportional to the magnitude of tension, showing a spike at the apex of the expanded skin. This implies that SFRP2 might be a newly discovered effector of mechanotransduction pathways. In addition, we found that acute stretching induces accumulation of b-catenin in the nuclei of basal keratinocytes (KCs) and LCs, indicating Wnt signalling activation, followed by cell proliferation. Moreover, TE-activated LCs proliferate and migrate into the suprabasal layer of skin, suggesting that LCs rebuild their steady network within the growing epidermis. We demonstrated that in vitro hrSFRP2 treatment on KCs inhibits Wnt/b-catenin signalling and stimulates KC differentiation. In parallel, we observed an accumulation of KRT10 in vivo in the expanded skin, pointing to TE-induced, SFRP2-augmented KC maturation. Overall, our results reveal that a network of LCs delivers SFRP2 across the epidermis to fine-tune Wnt/b-catenin signalling to restore epidermal homeostasis disrupted by TE.


Asunto(s)
Células de Langerhans , beta Catenina , Animales , Epidermis/metabolismo , Mecanotransducción Celular , Porcinos , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
3.
Development ; 145(17)2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30082271

RESUMEN

During embryogenesis, the germ layers, including the endoderm, undergo convergence and extension movements to narrow and elongate the body plan. In zebrafish, the dorsal migration of endodermal cells during gastrulation is controlled by chemokine signaling, but little is known about how they migrate during segmentation. Here, we show that glypican 4 (Gpc4), a member of the heparin sulfate proteoglycan family, is required for efficient migration of anterior endodermal cells during early segmentation, regulating Rac activation to maintain polarized actin-rich lamellipodia. An endoderm transplantation assay showed that Gpc4 regulates endoderm migration in a non-cell-autonomous fashion. Further analyses revealed that the impaired endoderm migration in gpc4 mutants results from increases in the expression and assembly of fibronectin and laminin, major components of the extracellular matrix (ECM). Notably, we found that matrix metalloproteinase 14 (Mmp14a/b) is required for the control of ECM expression during endoderm migration, with Gpc4 acting through Mmp14a/b to limit ECM expression. Our results suggest that Gpc4 is crucial for generating the environment required for efficient migration of endodermal cells, uncovering a novel function of Gpc4 during development.


Asunto(s)
Tipificación del Cuerpo/fisiología , Movimiento Celular/fisiología , Endodermo/embriología , Glipicanos/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Movimiento Celular/genética , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Gastrulación/fisiología , Glipicanos/genética , Laminina/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Seudópodos/metabolismo , Proteínas de Unión al GTP rac/metabolismo
4.
Development ; 145(9)2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29650589

RESUMEN

Zebrafish mutants with increased retinoic acid (RA) signaling due to the loss of the RA-inactivating enzyme Cyp26b1 develop a hyper-mineralized spine with gradually fusing vertebral body precursors (centra). However, the underlying cellular mechanisms remain incompletely understood. Here, we show that cells of the notochord epithelium named chordoblasts are sensitive to RA signaling. Chordoblasts are uniformly distributed along the anteroposterior axis and initially generate the continuous collagenous notochord sheath. However, subsequently and iteratively, subsets of these cells undergo further RA-dependent differentiation steps, acquire a stellate-like shape, downregulate expression of the collagen gene col2a1a, switch on cyp26b1 expression and trigger metameric sheath mineralization. This mineralization fails to appear upon chordoblast-specific cell ablation or RA signal transduction blockade. Together, our data reveal that, despite their different developmental origins, the activities and regulation of chordoblasts are very similar to those of osteoblasts, including their RA-induced transition from osteoid-producing cells to osteoid-mineralizing ones. Furthermore, our data point to a requirement for locally controlled RA activity within the chordoblast layer in order to generate the segmented vertebral column.


Asunto(s)
Calcificación Fisiológica/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Notocorda/embriología , Columna Vertebral/embriología , Tretinoina/metabolismo , Pez Cebra/embriología , Animales , Colágeno/biosíntesis , Colágeno/genética , Notocorda/citología , Ácido Retinoico 4-Hidroxilasa/genética , Ácido Retinoico 4-Hidroxilasa/metabolismo , Columna Vertebral/citología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Haematologica ; 104(1): 13-24, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30573510

RESUMEN

Inherited bone marrow failure syndromes are experiments of nature characterized by impaired hematopoiesis with cancer and leukemia predisposition. The mutations associated with inherited bone marrow failure syndromes affect fundamental cellular pathways, such as DNA repair, telomere maintenance, or proteostasis. How these disturbed pathways fail to produce sufficient blood cells and lead to leukemogenesis are not understood. The rarity of inherited cytopenias, the paucity of affected primary human hematopoietic cells, and the sometime inadequacy of murine or induced pluripotential stem cell models mean it is difficult to acquire a greater understanding of them. Zebrafish offer a model organism to study gene functions. As vertebrates, zebrafish share with humans many orthologous genes involved in blood disorders. As a model organism, zebrafish provide advantages that include rapid development of transparent embryos, high fecundity (providing large numbers of mutant and normal siblings), and a large collection of mutant and transgenic lines useful for investigating the blood system and other tissues during development. Importantly, recent advances in genomic editing in zebrafish can speedily validate the new genes or novel variants discovered in clinical investigation as causes for marrow failure. Here we review zebrafish as a model organism that phenocopies Fanconi anemia, Diamond-Blackfan anemia, dyskeratosis congenita, Shwachman-Diamond syndrome, congenital amegakaryocytic thrombocytopenia, and severe congenital neutropenia. Two important insights, provided by modeling inherited cytopenias in zebrafish, widen understanding of ribosome biogenesis and TP53 in mediating marrow failure and non-hematologic defects. They suggest that TP53-independent pathways contribute to marrow failure. In addition, zebrafish provide an attractive model organism for drug development.


Asunto(s)
Trastornos de Fallo de la Médula Ósea , Enfermedades Genéticas Congénitas , Proteína p53 Supresora de Tumor , Proteínas de Pez Cebra , Pez Cebra , Animales , Trastornos de Fallo de la Médula Ósea/genética , Trastornos de Fallo de la Médula Ósea/metabolismo , Trastornos de Fallo de la Médula Ósea/patología , Modelos Animales de Enfermedad , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Humanos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Pediatr Blood Cancer ; 64(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28398638

RESUMEN

PURPOSE: Malignant rhabdoid tumors (MRTs) are deadly embryonal tumors of the infancy. With poor survival and modest response to available therapies, more effective and less toxic treatments are needed. We hypothesized that a systematic screening of the kinome will reveal kinases that drive rhabdoid tumors and can be targeted by specific inhibitors. METHODS: We individually mutated 160 kinases in a well-characterized rhabdoid tumor cell line (MON) using lentiviral clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9). The kinase that most significantly impaired cell growth was further validated. Its expression was evaluated by microarray gene expression (GE) within 111 pediatric tumors, and functional assays were performed. A small molecule inhibitor was tested in multiple rhabdoid tumor cell lines and its toxicity evaluated in zebrafish larvae. RESULTS: The Polo-like kinase 4 (PLK4) was identified as the kinase that resulted in higher impairment of cell proliferation when mutated by CRISPR/Cas9. PLK4 CRISPR-mutated rhabdoid cells demonstrated significant decrease in proliferation, viability, and survival. GE showed upregulation of PLK4 in rhabdoid tumors and other embryonal tumors of the brain. The PLK4 inhibitor CFI-400945 showed cytotoxic effects on rhabdoid tumor cell lines while sparing non-neoplastic human fibroblasts and developing zebrafish larvae. CONCLUSIONS: Our findings indicate that rhabdoid tumor cell proliferation is highly dependent on PLK4 and suggest that targeting PLK4 with small-molecule inhibitors may hold a novel strategy for the treatment of MRT and possibly other embryonal tumors of the brain. This is the first time that PLK4 has been described as a potential target for both brain and pediatric tumors.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Sistemas CRISPR-Cas/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Indazoles/farmacología , Indoles/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Tumor Rabdoide/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Larva/crecimiento & desarrollo , Larva/metabolismo , Mutación/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Alineación de Secuencia , Células Tumorales Cultivadas , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
7.
J Neurosci ; 35(15): 6038-50, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878277

RESUMEN

Mechanisms underlying motor neuron degeneration in spinal muscular atrophy (SMA), the leading inherited cause of infant mortality, remain largely unknown. Many studies have established the importance of hyperphosphorylation of the microtubule-associated protein tau in various neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. However, tau phosphorylation in SMA pathogenesis has yet to be investigated. Here we show that tau phosphorylation on serine 202 (S202) and threonine 205 (T205) is increased significantly in SMA motor neurons using two SMA mouse models and human SMA patient spinal cord samples. Interestingly, phosphorylated tau does not form aggregates in motor neurons or neuromuscular junctions (NMJs), even at late stages of SMA disease, distinguishing it from other tauopathies. Hyperphosphorylation of tau on S202 and T205 is mediated by cyclin-dependent kinase 5 (Cdk5) in SMA disease condition, because tau phosphorylation at these sites is significantly reduced in Cdk5 knock-out mice; genetic knock-out of Cdk5 activating subunit p35 in an SMA mouse model also leads to reduced tau phosphorylation on S202 and T205 in the SMA;p35(-/-) compound mutant mice. In addition, expression of the phosphorylation-deficient tauS202A,T205A mutant alleviates motor neuron defects in a zebrafish SMA model in vivo and mouse motor neuron degeneration in culture, whereas expression of phosphorylation-mimetic tauS202E,T205E promotes motor neuron defects. More importantly, genetic knock-out of tau in SMA mice rescues synapse stripping on motor neurons, NMJ denervation, and motor neuron degeneration in vivo. Altogether, our findings suggest a novel mechanism for SMA pathogenesis in which hyperphosphorylation of non-aggregating tau by Cdk5 contributes to motor neuron degeneration.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal , Degeneración Nerviosa/etiología , Médula Espinal/patología , Proteínas tau/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Inmunoprecipitación , Lactante , Recién Nacido , Masculino , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Músculo Esquelético/patología , Atrofia Muscular Espinal/complicaciones , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Proteínas del Tejido Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Proteínas Nucleares/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Fosforilación , Proteínas Represoras/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Pez Cebra , Proteínas tau/deficiencia , Proteínas tau/genética
8.
Development ; 140(21): 4362-74, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24067352

RESUMEN

Morphogenesis of the semicircular canal ducts in the vertebrate inner ear is a dramatic example of epithelial remodelling in the embryo, and failure of normal canal development results in vestibular dysfunction. In zebrafish and Xenopus, semicircular canal ducts develop when projections of epithelium, driven by extracellular matrix production, push into the otic vesicle and fuse to form pillars. We show that in the zebrafish, extracellular matrix gene expression is high during projection outgrowth and then rapidly downregulated after fusion. Enzymatic disruption of hyaluronan in the projections leads to their collapse and a failure to form pillars: as a result, the ears swell. We have cloned a zebrafish mutant, lauscher (lau), identified by its swollen ear phenotype. The primary defect in the ear is abnormal projection outgrowth and a failure of fusion to form the semicircular canal pillars. Otic expression of extracellular matrix components is highly disrupted: several genes fail to become downregulated and remain expressed at abnormally high levels into late larval stages. The lau mutations disrupt gpr126, an adhesion class G protein-coupled receptor gene. Expression of gpr126 is similar to that of sox10, an ear and neural crest marker, and is partially dependent on sox10 activity. Fusion of canal projections and downregulation of otic versican expression in a hypomorphic lau allele can be restored by cAMP agonists. We propose that Gpr126 acts through a cAMP-mediated pathway to control the outgrowth and adhesion of canal projections in the zebrafish ear via the regulation of extracellular matrix gene expression.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Morfogénesis/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Canales Semicirculares/embriología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , AMP Cíclico/metabolismo , Matriz Extracelular/metabolismo , Genotipo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Hibridación in Situ , Repeticiones de Microsatélite/genética , Faloidina , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción SOXE/metabolismo , Canales Semicirculares/anomalías , Análisis de Secuencia de ADN , Versicanos/metabolismo
9.
Dev Biol ; 386(1): 72-85, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24333517

RESUMEN

Congenital vertebral malformations (CVM) occur in 1 in 1000 live births and in many cases can cause spinal deformities, such as scoliosis, and result in disability and distress of affected individuals. Many severe forms of the disease, such as spondylocostal dystostosis, are recessive monogenic traits affecting somitogenesis, however the etiologies of the majority of CVM cases remain undetermined. Here we demonstrate that morphological defects of the notochord in zebrafish can generate congenital-type spine defects. We characterize three recessive zebrafish leviathan/col8a1a mutant alleles ((m531, vu41, vu105)) that disrupt collagen type VIII alpha1a (col8a1a), and cause folding of the embryonic notochord and consequently adult vertebral column malformations. Furthermore, we provide evidence that a transient loss of col8a1a function or inhibition of Lysyl oxidases with drugs during embryogenesis was sufficient to generate vertebral fusions and scoliosis in the adult spine. Using periodic imaging of individual zebrafish, we correlate focal notochord defects of the embryo with vertebral malformations (VM) in the adult. Finally, we show that bends and kinks in the notochord can lead to aberrant apposition of osteoblasts normally confined to well-segmented areas of the developing vertebral bodies. Our results afford a novel mechanism for the formation of VM, independent of defects of somitogenesis, resulting from aberrant bone deposition at regions of misshapen notochord tissue.


Asunto(s)
Colágeno Tipo VIII/fisiología , Regulación del Desarrollo de la Expresión Génica , Columna Vertebral/anomalías , Pez Cebra/embriología , Alelos , Animales , Colágeno Tipo VIII/genética , Cruzamientos Genéticos , Hibridación in Situ , Meiosis , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mutación , Notocorda/anomalías , Osteoblastos/citología , Osteoblastos/metabolismo , Proteína-Lisina 6-Oxidasa/metabolismo , Factores de Tiempo , Pez Cebra/genética
10.
Development ; 139(13): 2416-25, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22669824

RESUMEN

Multiple developmental processes require tightly controlled Wnt signaling, and its misregulation leads to congenital abnormalities and diseases. Glypicans are extracellular proteins that modulate the Wnt pathway. In addition to interacting with Wnts, these glycosophosphotidylinositol (GPI)-anchored, heparan-sulfate proteoglycans bind ligands of several other signaling pathways in both vertebrates and invertebrates. In Drosophila, Notum, a secreted α/ß-hydrolase, antagonizes the signaling of the prototypical Wnt Wingless (Wg), by releasing glypicans from the cell surface. Studies of mammalian Notum indicate promiscuous target specificity in cell culture, but the role of Notum in vertebrate development has not been studied. Our work shows that zebrafish Notum 1a, an ortholog of mammalian Notum, contributes to a self-regulatory loop that restricts Wnt/ß-catenin signaling. Notum 1a does not interact with Glypican 4, an essential component of the Wnt/planar cell polarity (PCP) pathway. Our results suggest a surprising specific role of Notum in the developing vertebrate embryo.


Asunto(s)
Hidrolasas/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , beta Catenina/metabolismo , Animales , Tipificación del Cuerpo , Polaridad Celular , Desarrollo Embrionario , Glipicanos/metabolismo , Hidrolasas/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
11.
J Neurosci ; 33(5): 2177-87, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23365253

RESUMEN

To form complex neuronal networks, growth cones use intermediate targets as guideposts on the path to more distant targets. In the developing zebrafish (Danio rerio), the muscle pioneers (MPs) are intermediate targets for primary motor neurons (PMNs) that innervate the trunk musculature. The mechanisms regulating PMN axon guidance at the MPs are not fully understood. We have identified a new member of the Notum family in zebrafish, Notum 2, which is expressed exclusively in the MPs during primary motor innervation. While homologs of Notum, including zebrafish Notum 1a, negatively regulate the Wnt/ß-catenin signaling pathway, we discovered a novel function of Notum 2 in regulating motor axon guidance. Knockdown of Notum 2 resulted in a failure of caudal primary (CaP) axons to migrate beyond the MPs, despite the proper specification of the intermediate target. In contrast, mosaic Notum 2 overexpression induced branching of PMN axons. This effect is specific to Notum 2, as overexpression of Notum 1a does not affect PMN axon trajectory. Ectopic expression of Notum 2 by cells contacting the growing CaP axon induced the highest frequency of branching, suggesting that localized Notum 2 expression affects axon behavior. We propose a model where Notum 2 expression at the MPs provides a cue to release CaP motor axons from their intermediate targets, allowing growth cones to proceed to secondary targets in the ventral muscle. This work demonstrates an unexpected role for a Notum homolog in regulating growth cone migration, separate from the well established functions of other Notum homologs in Wnt signaling.


Asunto(s)
Hidrolasas/metabolismo , Neuronas Motoras/metabolismo , Músculo Esquelético/inervación , Proteínas de Pez Cebra/metabolismo , Animales , Axones/metabolismo , Movimiento Celular/fisiología , Conos de Crecimiento/metabolismo , Hidrolasas/genética , Músculo Esquelético/metabolismo , Vía de Señalización Wnt/fisiología , Pez Cebra , Proteínas de Pez Cebra/genética
12.
Blood ; 119(11): 2679-87, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22207734

RESUMEN

We discovered that miR-27b controls 2 critical vascular functions: it turns the angiogenic switch on by promoting endothelial tip cell fate and sprouting and it promotes venous differentiation. We have identified its targets, a Notch ligand Delta-like ligand 4 (Dll4) and Sprouty homologue 2 (Spry2). miR-27b knockdown in zebrafish and mouse tissues severely impaired vessel sprouting and filopodia formation. Moreover, miR-27b was necessary for the formation of the first embryonic vein in fish and controlled the expression of arterial and venous markers in human endothelium, including Ephrin B2 (EphB2), EphB4, FMS-related tyrosine kinase 1 (Flt1), and Flt4. In zebrafish, Dll4 inhibition caused increased sprouting and longer intersegmental vessels and exacerbated tip cell migration. Blocking Spry2 caused premature vessel branching. In contrast, Spry2 overexpression eliminated the tip cell branching in the intersegmental vessels. Blockade of Dll4 and Spry2 disrupted arterial specification and augmented the expression of venous markers. Blocking either Spry2 or Dll4 rescued the miR-27b knockdown phenotype in zebrafish and in mouse vascular explants, pointing to essential roles of these targets downstream of miR-27b. Our study identifies critical role of miR-27b in the control of endothelial tip cell fate, branching, and venous specification and determines Spry2 and Dll4 as its essential targets.


Asunto(s)
Arterias/embriología , Endotelio Vascular/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/fisiología , Neovascularización Fisiológica , Pez Cebra/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Aorta/citología , Aorta/metabolismo , Arterias/metabolismo , Biomarcadores/metabolismo , Northern Blotting , Western Blotting , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular , Movimiento Celular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas , Seudópodos/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/metabolismo
13.
Dev Growth Differ ; 55(2): 282-300, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23350700

RESUMEN

The zebrafish maxillary barbel is an integumentary organ containing skin, glands, pigment cells, taste buds, nerves, and endothelial vessels. The maxillary barbel can regenerate (LeClair & Topczewski 2010); however, little is known about its molecular regulation. We have studied fibroblast growth factor (FGF) pathway molecules during barbel regeneration, comparing this system to a well-known regenerating appendage, the zebrafish caudal fin. Multiple FGF ligands (fgf20a, fgf24), receptors (fgfr1-4) and downstream targets (pea3, il17d) are expressed in normal and regenerating barbel tissue, confirming FGF activation. To test if specific FGF pathways were required for barbel regeneration, we performed simultaneous barbel and caudal fin amputations in two temperature-dependent zebrafish lines. Zebrafish homozygous for a point mutation in fgf20a, a factor essential for caudal fin blastema formation, regrew maxillary barbels normally, indicating that the requirement for this ligand is appendage-specific. Global overexpression of a dominant negative FGF receptor, Tg(hsp70l:dn-fgfr1:EGFP)(pd1) completely blocked fin outgrowth but only partially inhibited barbel outgrowth, suggesting reduced requirements for FGFs in barbel tissue. Maxillary barbels expressing dn-fgfr1 regenerated peripheral nerves, dermal connective tissue, endothelial tubes, and a glandular epithelium; in contrast to a recent report in which dn-fgfr1 overexpression blocks pharyngeal taste bud formation in zebrafish larvae (Kapsimali et al. 2011), we observed robust formation of calretinin-positive tastebuds. These are the first experiments to explore the molecular mechanisms of maxillary barbel regeneration. Our results suggest heterogeneous requirements for FGF signaling in the regeneration of different zebrafish appendages (caudal fin versus maxillary barbel) and taste buds of different embryonic origin (pharyngeal endoderm versus barbel ectoderm).


Asunto(s)
Aletas de Animales/fisiología , Estructuras Animales/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Regeneración/fisiología , Proteínas de Pez Cebra/fisiología , Amputación Quirúrgica , Aletas de Animales/metabolismo , Aletas de Animales/cirugía , Estructuras Animales/metabolismo , Estructuras Animales/cirugía , Animales , Animales Modificados Genéticamente , Femenino , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Microscopía Fluorescente , Mutación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Regeneración/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Temperatura , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética
14.
Nat Med ; 12(8): 925-32, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16892036

RESUMEN

Bidirectional cellular communication is integral to both cancer progression and embryological development. In addition, aggressive tumor cells are phenotypically plastic, sharing many properties with embryonic cells. Owing to the similarities between these two types of cells, the developing zebrafish can be used as a biosensor for tumor-derived signals. Using this system, we show that aggressive melanoma cells secrete Nodal (a potent embryonic morphogen) and consequently can induce ectopic formation of the embryonic axis. We further show that Nodal is present in human metastatic tumors, but not in normal skin, and thus may be involved in melanoma pathogenesis. Inhibition of Nodal signaling reduces melanoma cell invasiveness, colony formation and tumorigenicity. Nodal inhibition also promotes the reversion of melanoma cells toward a melanocytic phenotype. These data suggest that Nodal signaling has a key role in melanoma cell plasticity and tumorigenicity, thereby providing a previously unknown molecular target for regulating tumor progression.


Asunto(s)
Melanoma/patología , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Blástula/trasplante , Línea Celular Tumoral , Embrión no Mamífero , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Melanocitos/metabolismo , Melanocitos/patología , Proteínas de la Membrana/antagonistas & inhibidores , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Oligonucleótidos Antisentido/farmacología , Trasplante Heterólogo , Proteínas de Pez Cebra/antagonistas & inhibidores
15.
Dev Biol ; 357(2): 518-31, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21723274

RESUMEN

Zebrafish (Danio rerio) is an excellent model organism for the study of vertebrate development including skeletogenesis. Studies of mammalian cartilage formation were greatly advanced through the use of a cartilage specific regulatory element of the Collagen type II alpha 1 (Col2a1) gene. In an effort to isolate such an element in zebrafish, we compared the expression of two col2a1 homologues and found that expression of col2a1b, a previously uncharacterized zebrafish homologue, only partially overlaps with col2a1a. We focused our analysis on col2a1a, as it is expressed in both the stacked chondrocytes and the perichondrium. By comparing the genomic sequence surrounding the predicted transcriptional start site of col2a1a among several species of teleosts we identified a small highly conserved sequence (R2) located 1.7 kb upstream of the presumptive transcriptional initiation site. Interestingly, neither the sequence nor location of this element is conserved between teleost and mammalian Col2a1. We generated transient and stable transgenic lines with just the R2 element or the entire 1.7 kb fragment 5' of the transcriptional initiation site. The identified regulatory elements enable the tracking of cellular development in various tissues by driving robust reporter expression in craniofacial cartilage, ear, notochord, floor plate, hypochord and fins in a pattern similar to the expression of endogenous col2a1a. Using a reporter gene driven by the R2 regulatory element, we analyzed the morphogenesis of the notochord sheath cells as they withdraw from the stack of initially uniform cells and encase the inflating vacuolated notochord cells. Finally, we show that like endogenous col2a1a, craniofacial expression of these reporter constructs depends on Sox9a transcription factor activity. At the same time, notochord expression is maintained after Sox9a knockdown, suggesting that other factors can activate expression through the identified regulatory element in this tissue.


Asunto(s)
Colágeno Tipo II/genética , Secuencia Conservada/genética , Evolución Molecular , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Aletas de Animales/embriología , Aletas de Animales/metabolismo , Animales , Cartílago/embriología , Cartílago/metabolismo , Colágeno Tipo II/metabolismo , Regulación hacia Abajo/genética , Oído/embriología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Genoma/genética , Disco Intervertebral/embriología , Disco Intervertebral/metabolismo , Larva/genética , Notocorda/embriología , Notocorda/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Homología de Secuencia de Aminoácido , Sintenía/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
16.
Dev Biol ; 337(2): 233-45, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19895804

RESUMEN

Tbx5 is involved in congenital heart disease, however, the mechanisms leading to organ malformation are greatly unknown. We hypothesized a model by which the Tbx5 binding protein Pdlim7 controls nuclear/cytoplasmic shuttling and function of the transcription factor. Using the zebrafish, we present in vivo significance for an essential role of Tbx5/Pdlim7 protein interaction in the regulation of cardiac formation. Knock-down of Pdlim7 results in a non-looped heart, strikingly reminiscent of the tbx5 heartstrings mutant phenotype. However, while misregulation of Pdlim7 and Tbx5 produce similar aberrant cardiac morphology, molecular and histological analysis uncovered that the Pdlim7 and Tbx5 cardiac phenotypes are due to opposite effects on valve development. Loss of Pdlim7 function causes no valve tissue to develop while lack of Tbx5 results in increased valve tissue. These opposing defects are evident before valve formation and are the result of distinct gene misregulation during specification of the atrio-ventricular (AV) boundary. We show that Pdlim7/Tbx5 interactions affect the expression of Tbx5 target genes nppa and tbx2b at the AV boundary, and their domains of misexpression directly correlate with the identified valve defects. These studies demonstrate that controlling the correct balance of Tbx5 activity is crucial for the specification of the AV boundary and valve formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Tipificación del Cuerpo , Atrios Cardíacos/embriología , Válvulas Cardíacas/embriología , Ventrículos Cardíacos/embriología , Proteínas de Dominio T Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Proteínas Adaptadoras Transductoras de Señales/genética , Estructuras Animales/metabolismo , Animales , Tipificación del Cuerpo/genética , Células COS , Diferenciación Celular , Chlorocebus aethiops , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Atrios Cardíacos/metabolismo , Válvulas Cardíacas/metabolismo , Ventrículos Cardíacos/metabolismo , Miocardio/citología , Miocardio/metabolismo , Especificidad de Órganos/genética , Unión Proteica , Proteínas de Dominio T Box/genética , Factores de Tiempo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
17.
Nat Cell Biol ; 4(8): 610-5, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12105418

RESUMEN

Embryonic morphogenesis is driven by a suite of cell behaviours, including coordinated shape changes, cellular rearrangements and individual cell migrations, whose molecular determinants are largely unknown. In the zebrafish, Dani rerio, trilobite mutant embryos have defects in gastrulation movements and posterior migration of hindbrain neurons. Here, we have used positional cloning to demonstrate that trilobite mutations disrupt the transmembrane protein Strabismus (Stbm)/Van Gogh (Vang), previously associated with planar cell polarity (PCP) in Drosophila melanogaster, and PCP and canonical Wnt/beta-catenin signalling in vertebrates. Our genetic and molecular analyses argue that during gastrulation, trilobite interacts with the PCP pathway without affecting canonical Wnt signalling. Furthermore, trilobite may regulate neuronal migration independently of PCP molecules. We show that trilobite mediates polarization of distinct movement behaviours. During gastrulation convergence and extension movements, trilobite regulates mediolateral cell polarity underlying effective intercalation and directed dorsal migration at increasing velocities. In the hindbrain, trilobite controls effective migration of branchiomotor neurons towards posterior rhombomeres. Mosaic analyses show trilobite functions cell-autonomously and non-autonomously in gastrulae and the hindbrain. We propose Trilobite/Stbm mediates cellular interactions that confer directionality on distinct movements during vertebrate embryogenesis.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Movimiento Celular , Gástrula/metabolismo , Proteínas de la Membrana/genética , Mutación , Neuronas/citología , Neuronas/metabolismo , Transducción de Señal , Pez Cebra/genética , Proteínas de Pez Cebra/genética
18.
J Cell Biol ; 169(5): 777-87, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15928205

RESUMEN

Galpha(12/13) have been implicated in numerous cellular processes, however, their roles in vertebrate gastrulation are largely unknown. Here, we show that during zebrafish gastrulation, suppression of both Galpha(12) and Galpha(13) signaling by overexpressing dominant negative proteins and application of antisense morpholino-modified oligonucleotide translation interference disrupted convergence and extension without changing embryonic patterning. Analyses of mesodermal cell behaviors revealed that Galpha(12/13) are required for cell elongation and efficient dorsalward migration during convergence independent of noncanonical Wnt signaling. Furthermore, Galpha(12/13) function cell-autonomously to mediate mediolateral cell elongation underlying intercalation during notochord extension, likely acting in parallel to noncanonical Wnt signaling. These findings provide the first evidence that Galpha(12) and Galpha(13) have overlapping and essential roles in distinct cell behaviors that drive vertebrate gastrulation.


Asunto(s)
Desarrollo Embrionario/fisiología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Gástrula/metabolismo , Mesodermo/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Tipificación del Cuerpo/fisiología , Comunicación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Forma de la Célula/fisiología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Gástrula/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/citología , Datos de Secuencia Molecular , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt
19.
Dev Dyn ; 238(10): 2550-63, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19777561

RESUMEN

The heparan sulfate proteoglycan Glypican 4 (Gpc4) is part of the Wnt/planar cell polarity pathway, which is required for convergence and extension during zebrafish gastrulation. To observe Glypican 4-deficient phenotypes at later stages, we rescued gpc4(-/-) (knypek) homozygotes and raised them for more than one year. Adult mutants showed diverse cranial malformations of both dermal and endochondral bones, ranging from shortening of the rostral-most skull to loss of the symplectic. Additionally, the adult palatoquadrate cartilage was disorganized, with abnormal chondrocyte orientation. To understand how the palatoquadrate cartilage normally develops, we examined a juvenile series of wild type and mutant specimens. This identified two novel domains of elongated chondrocytes in the larval palatoquadrate, which normally form prior to endochondral ossification. In contrast, gpc4(-/-) larvae never form these domains, suggesting a failure of chondrocyte orientation, though not differentiation. Our findings implicate Gpc4 in the regulation of zebrafish cartilage and bone morphogenesis.


Asunto(s)
Tipificación del Cuerpo , Cartílago , Huesos Faciales , Glipicanos/metabolismo , Cráneo , Proteínas de Pez Cebra/metabolismo , Pez Cebra , Animales , Animales Modificados Genéticamente , Cartílago/anomalías , Cartílago/anatomía & histología , Cartílago/crecimiento & desarrollo , Huesos Faciales/anomalías , Huesos Faciales/anatomía & histología , Huesos Faciales/crecimiento & desarrollo , Técnicas de Silenciamiento del Gen , Glipicanos/genética , Humanos , Fenotipo , Cráneo/anomalías , Cráneo/anatomía & histología , Cráneo/crecimiento & desarrollo , Pez Cebra/anomalías , Pez Cebra/anatomía & histología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética
20.
Dis Model Mech ; 13(6)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32430393

RESUMEN

Human disorders of the post-squalene cholesterol biosynthesis pathway frequently result in skeletal abnormalities, yet our understanding of the mechanisms involved is limited. In a forward-genetic approach, we have found that a late-onset skeletal mutant, named kolibernu7 , is the result of a cis-acting regulatory mutation leading to loss of methylsterol monooxygenase 1 (msmo1) expression within pre-hypertrophic chondrocytes. Generated msmo1nu81 knockdown mutation resulted in lethality at larval stage. We demonstrated that this is a result of both cholesterol deprivation and sterol intermediate accumulation by creating a mutation eliminating activity of Lanosterol synthase (Lss). Our results indicate that double lssnu60;msmo1nu81 and single lssnu60 mutants survive significantly longer than msmo1nu81 homozygotes. Liver-specific restoration of either Msmo1 or Lss in corresponding mutant backgrounds suppresses larval lethality. Rescued mutants develop dramatic skeletal abnormalities, with a loss of Msmo1 activity resulting in a more-severe patterning defect of a near-complete loss of hypertrophic chondrocytes marked by col10a1a expression. Our analysis suggests that hypertrophic chondrocytes depend on endogenous cholesterol synthesis, and blocking C4 demethylation exacerbates the cholesterol deficiency phenotype. Our findings offer new insight into the genetic control of bone development and provide new zebrafish models for human disorders of the cholesterol biosynthesis pathway.


Asunto(s)
Enfermedades del Desarrollo Óseo/metabolismo , Huesos/metabolismo , Colesterol/biosíntesis , Condrocitos/metabolismo , Hígado/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/patología , Huesos/patología , Condrocitos/patología , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Transferasas Intramoleculares/genética , Transferasas Intramoleculares/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Mutación , Fenotipo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA