Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Traffic ; 17(1): 66-79, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26456025

RESUMEN

The differential distribution of lipids between apical and basolateral membranes is necessary for many epithelial cell functions, but how this characteristic membrane organization is integrated within the polarity network during ductal organ development is poorly understood. Here we quantified membrane order in the gut, kidney and liver ductal epithelia in zebrafish larvae at 3-11 days post fertilization (dpf) with Laurdan 2-photon microscopy. We then applied a combination of Laurdan imaging, antisense knock-down and analysis of polarity markers to understand the relationship between membrane order and apical-basal polarity. We found a reciprocal relationship between membrane order and the cell polarity network. Reducing membrane condensation by exogenously added oxysterol or depletion of cholesterol reduced apical targeting of the polarity protein, aPKC. Conversely, using morpholino knock down in zebrafish, we found that membrane order was dependent upon the Crb3 and Par3 polarity protein expression in ductal epithelia. Hence our data suggest that the biophysical property of membrane lipid packing is a regulatory element in apical basal polarity.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Epitelio/metabolismo , Organogénesis , Proteína Quinasa C/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Colesterol/metabolismo , Epitelio/embriología , Proteínas de la Membrana , Pez Cebra
2.
J Neurosci ; 37(32): 7782-7794, 2017 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-28687604

RESUMEN

The neurodegenerative disease Machado-Joseph disease (MJD), also known as spinocerebellar ataxin-3, affects neurons of the brain and spinal cord, disrupting control of the movement of muscles. We have successfully established the first transgenic zebrafish (Danio rerio) model of MJD by expressing human ataxin-3 protein containing either 23 glutamines (23Q, wild-type) or 84Q (MJD-causing) within neurons. Phenotypic characterization of the zebrafish (male and female) revealed that the ataxin-3-84Q zebrafish have decreased survival compared with ataxin-3-23Q and develop ataxin-3 neuropathology, ataxin-3 cleavage fragments and motor impairment. Ataxin-3-84Q zebrafish swim shorter distances than ataxin-3-23Q zebrafish as early as 6 days old, even if expression of the human ataxin-3 protein is limited to motor neurons. This swimming phenotype provides a valuable readout for drug treatment studies. Treating the EGFP-ataxin-3-84Q zebrafish with the calpain inhibitor compound calpeptin decreased levels of ataxin-3 cleavage fragments, but also removed all human ataxin-3 protein (confirmed by ELISA) and prevented the early MJD zebrafish motor phenotype. We identified that this clearance of ataxin-3 protein by calpeptin treatment resulted from an increase in autophagic flux (indicated by decreased p62 levels and increased LC3II). Cotreatment with the autophagy inhibitor chloroquine blocked the decrease in human ataxin-3 levels and the improved movement produced by calpeptin treatment. This study demonstrates that this first transgenic zebrafish model of MJD is a valuable tool for testing potential treatments for MJD. Calpeptin treatment is protective in this model of MJD and removal of human ataxin-3 through macro-autophagy plays an important role in this beneficial effect.SIGNIFICANCE STATEMENT We have established the first transgenic zebrafish model of the neurodegenerative disease MJD, and identified relevant disease phenotypes, including impaired movement from an early age, which can be used in rapid drug testing studies. We have found that treating the MJD zebrafish with the calpain inhibitor compound calpeptin produces complete removal of human ataxin-3 protein, due to induction of the autophagy quality control pathway. This improves the movement of the MJD zebrafish. Artificially blocking the autophagy pathway prevents the removal of human ataxin-3 and improved movement produced by calpeptin treatment. These findings indicate that induction of autophagy, and removal of ataxin-3 protein, plays an important role in the protective effects of calpain inhibition for the treatment of MJD.


Asunto(s)
Ataxina-3/metabolismo , Autofagia/fisiología , Calpaína/metabolismo , Modelos Animales de Enfermedad , Glicoproteínas/farmacología , Enfermedad de Machado-Joseph/metabolismo , Proteínas Represoras/metabolismo , Animales , Animales Modificados Genéticamente , Ataxina-3/genética , Autofagia/efectos de los fármacos , Calpaína/antagonistas & inhibidores , Calpaína/genética , Femenino , Glicoproteínas/uso terapéutico , Humanos , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/prevención & control , Masculino , Proteínas Represoras/genética , Pez Cebra
3.
Hum Mol Genet ; 25(9): 1728-38, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908606

RESUMEN

Spinal muscular atrophy (SMA) is an autosomal recessive disease linked to survival motor neuron (SMN) protein deficiency. While SMN protein is expressed ubiquitously, its deficiency triggers tissue-specific hallmarks, including motor neuron death and muscle atrophy, leading to impaired motor functions and premature death. Here, using stable miR-mediated knockdown technology in zebrafish, we developed the first vertebrate system allowing transgenic spatio-temporal control of the smn1 gene. Using this new model it is now possible to investigate normal and pathogenic SMN function(s) in specific cell types, independently or in synergy with other cell populations. We took advantage of this new system to first test the effect of motor neuron or muscle-specific smn1 silencing. Anti-smn1 miRNA expression in motor neurons, but not in muscles, reproduced SMA hallmarks, including abnormal motor neuron development, poor motor function and premature death. Interestingly, smn1 knockdown in motor neurons also induced severe late-onset phenotypes including scoliosis-like body deformities, weight loss, muscle atrophy and, seen for the first time in zebrafish, reduction in the number of motor neurons, indicating motor neuron degeneration. Taken together, we have developed a new transgenic system allowing spatio-temporal control of smn1 expression in zebrafish, and using this model, we have demonstrated that smn1 silencing in motor neurons alone is sufficient to reproduce SMA hallmarks in zebrafish. It is noteworthy that this research is going beyond SMA as this versatile gene-silencing transgenic system can be used to knockdown any genes of interest, filling the gap in the zebrafish genetic toolbox and opening new avenues to study gene functions in this organism.


Asunto(s)
Embrión no Mamífero/citología , MicroARNs/genética , Neuronas Motoras/patología , Músculo Esquelético/patología , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Neuronas Motoras/metabolismo , Músculo Esquelético/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/antagonistas & inhibidores , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Pez Cebra
4.
Hum Mol Genet ; 24(20): 5789-804, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26231217

RESUMEN

Correct morphogenesis and differentiation are critical in development and maintenance of the lens, which is a classic model system for epithelial development and disease. Through germline genomic analyses in patients with lens and eye abnormalities, we discovered functional mutations in the Signal Induced Proliferation Associated 1 Like 3 (SIPA1L3) gene, which encodes a previously uncharacterized member of the Signal Induced Proliferation Associated 1 (SIPA1 or SPA1) family, with a role in Rap1 signalling. Patient 1, with a de novo balanced translocation, 46,XY,t(2;19)(q37.3;q13.1), had lens and ocular anterior segment abnormalities. Breakpoint mapping revealed transection of SIPA1L3 at 19q13.1 and reduced SIPA1L3 expression in patient lymphoblasts. SIPA1L3 downregulation in 3D cell culture revealed morphogenetic and cell polarity abnormalities. Decreased expression of Sipa1l3 in zebrafish and mouse caused severe lens and eye abnormalities. Sipa1l3(-/-) mice showed disrupted epithelial cell organization and polarity and, notably, abnormal epithelial to mesenchymal transition in the lens. Patient 2 with cataracts was heterozygous for a missense variant in SIPA1L3, c.442G>T, p.Asp148Tyr. Examination of the p.Asp148Tyr mutation in an epithelial cell line showed abnormal clustering of actin stress fibres and decreased formation of adherens junctions. Our findings show that abnormalities of SIPA1L3 in human, zebrafish and mouse contribute to lens and eye defects, and we identify a critical role for SIPA1L3 in epithelial cell morphogenesis, polarity, adhesion and cytoskeletal organization.


Asunto(s)
Catarata/fisiopatología , Polaridad Celular , Citoesqueleto/ultraestructura , Anomalías del Ojo/fisiopatología , Proteínas Activadoras de GTPasa/genética , Mutación , Proteínas de Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Catarata/genética , Catarata/metabolismo , Polaridad Celular/genética , Análisis Mutacional de ADN , Transición Epitelial-Mesenquimal/genética , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Transducción de Señal , Pez Cebra/genética , Proteínas de Unión al GTP rap1/metabolismo
5.
Gen Comp Endocrinol ; 246: 233-240, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28041791

RESUMEN

Serotonin has been implicated in the inhibition of food intake in vertebrates. However, the mechanisms through which serotonin acts has yet to be elucidated. Recently, ETV5 (ets variant gene 5) has been associated with obesity and food intake control mechanisms in mammals. We have analyzed a putative physiological function of the two etv5 paralogous genes (etv5a and etv5b) in neuronal food intake control in adult zebrafish that have been exposed to different nutritional conditions. A feeding assay was established and fluoxetine, a selective serotonin re-uptake inhibitor (SSRI), was applied. Gene expression changes in the hypothalamus were determined using real-time PCR. Fasting induced an up-regulation of etv5a and etv5b in the hypothalamus, whereas increased serotonin levels in the fasted fish counteracted the increase in expression. To investigate potential mechanisms the expression of further food intake control genes was determined. The results show that an increase of serotonin in fasting fish causes a reduction in the activity of genes stimulating food intake. This is in line with a previously demonstrated anorexigenic function of serotonin. Our results suggest that obesity-associated ETV5 has a food intake stimulating function and that this function is modulated through serotonin.


Asunto(s)
Ayuno/fisiología , Proteínas de Peces/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Agonistas de Receptores de Serotonina/farmacología , Serotonina/farmacología , Pez Cebra/metabolismo , Animales , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hipotálamo/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Serotonina/química , Receptores de Serotonina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra/crecimiento & desarrollo
6.
Genesis ; 53(5): 321-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25864959

RESUMEN

miR218-1 and miR218-2 are embedded in introns of SLIT2 and SLIT3, respectively, an arrangement conserved throughout vertebrate genomes. Both miR218 genes are predicted to be transcribed in the same orientation as their host genes and were assumed to be spliced from Slit2/3 primary transcripts. In zebrafish miR218 is active in cranial nerve motor nuclei and spinal cord motor neurons, while slit2 and slit3 are expressed predominantly in the midline. This differential expression pattern suggested independent regulation of miR218 genes by distinct enhancers. We tested conserved noncoding elements for regulatory activity by reporter gene transgenesis in zebrafish. Two human enhancers, 76 kb and 130 kb distant from miR218-2, were identified that drove GFP expression in zebrafish in an almost complete miR218 expression pattern. In the zebrafish slit3 locus, two enhancers with identical activity were discovered. In human SLIT2 one enhancer 52 kb upstream of miR218-1 drove an expression pattern very similar to the enhancers of miR218-2. This establishes that miR218-1/-2 regulatory units are nested within SLIT2/3 and that they are duplicates of an ancestral single locus. Due to the strong activity of the enhancers, unique transgenic lines were created that facilitate morphological and gene functional genetic experiments in motor neurons.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Intrones , MicroARNs/genética , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
7.
Genesis ; 53(10): 640-51, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26271004

RESUMEN

Single Nucleotide Polymorphisms in FTO intron 1 have been associated with obesity risk, leading to the hypothesis that FTO is the obesity-related gene. However, other studies have shown that the FTO gene is part of the regulatory domain of the neighboring IRX3 gene and that enhancers in FTO intron 1 regulate IRX3. While Irx3 activity was shown to be necessary in the hypothalamus for the metabolic function of Irx3 in mouse, no enhancers with hypothalamic activity have been demonstrated in the risk-associated region within FTO. In order to identify potential enhancers at the human FTO locus in vivo, we tested regulatory activity in FTO intron 1 using BAC transgenesis in zebrafish. A minimal gata2 promoter-GFP cassette was inserted 1.3 kb upstream of the obesity associated SNP rs9939609 in a human FTO BAC plasmid. In addition to the previously identified expression domains in notochord and kidney, human FTO BAC:GFP transgenic zebrafish larvae expressed GFP in the ventral posterior tuberculum, the posterior hypothalamus and the anterior brainstem, which are also expression domains of zebrafish irx3a. In contrast, an in-frame insertion of a GFP cassette at the FTO start codon resulted in weak ubiquitous GFP expression indicating that the promoter of FTO does likely not react to enhancers located in the obesity risk-associated region.


Asunto(s)
Hipotálamo/metabolismo , Obesidad/genética , Polimorfismo de Nucleótido Simple , Proteínas/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Animales Modificados Genéticamente , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Inmunohistoquímica , Hibridación in Situ , Intrones/genética , Larva/genética , Larva/metabolismo , Microscopía Confocal , Regiones Promotoras Genéticas/genética , Proteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/metabolismo
8.
Hum Genet ; 134(11-12): 1163-82, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26337422

RESUMEN

Protein-coding mutations in the transcription factor-encoding gene ARX cause various forms of intellectual disability (ID) and epilepsy. In contrast, variations in surrounding non-coding sequences are correlated with milder forms of non-syndromic ID and autism and had suggested the importance of ARX gene regulation in the etiology of these disorders. We compile data on several novel and some already identified patients with or without ID that carry duplications of ARX genomic region and consider likely genetic mechanisms underlying the neurodevelopmental defects. We establish the long-range regulatory domain of ARX and identify its brain region-specific autoregulation. We conclude that neurodevelopmental disturbances in the patients may not simply arise from increased dosage due to ARX duplication. This is further exemplified by a small duplication involving a non-functional ARX copy, but with duplicated enhancers. ARX enhancers are located within a 504-kb region and regulate expression specifically in the forebrain in developing and adult zebrafish. Transgenic enhancer-reporter lines were used as in vivo tools to delineate a brain region-specific negative and positive autoregulation of ARX. We find autorepression of ARX in the telencephalon and autoactivation in the ventral thalamus. Fluorescently labeled brain regions in the transgenic lines facilitated the identification of neuronal outgrowth and pathfinding disturbances in the ventral thalamus and telencephalon that occur when arxa dosage is diminished. In summary, we have established a model for how breakpoints in long-range gene regulation alter the expression levels of a target gene brain region-specifically, and how this can cause subtle neuronal phenotypes relating to the etiology of associated neuropsychiatric disease.


Asunto(s)
Variaciones en el Número de Copia de ADN , Duplicación de Gen , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Adulto , Animales , Animales Modificados Genéticamente , Encéfalo/embriología , Encéfalo/metabolismo , Estudios de Casos y Controles , Embrión no Mamífero , Femenino , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Factores de Transcripción/metabolismo , Pez Cebra
9.
Methods ; 62(3): 216-25, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23542551

RESUMEN

We detail an approach for the identification of human tissue-specific transcriptional enhancers involving three steps: delineation of search space around a locus or target gene, in silico identification and size definition of putative candidate sequences, and testing through several independent genomic insertions in a transgenic zebrafish reporter assay. Candidate sequences are defined through evolutionary conservation, transcription factor binding and chromatin marks (e.g. ENCODE data) and are amplified from genomic DNA, cloned into basal promoter:fluorescent protein reporter vectors based on the Tol2 transposon system and are microinjected into fertilized zebrafish eggs. After raising injected founders to sexual maturity, fluorescent screening identifies positive founder fish whose offspring undergo a detailed expression analysis to determine tissue specificity and reproducibility of specific enhancers.


Asunto(s)
Animales Modificados Genéticamente , Elementos de Facilitación Genéticos , Genoma , Transgenes , Pez Cebra/genética , Animales , Elementos Transponibles de ADN , Femenino , Efecto Fundador , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Sitios Genéticos , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Microinyecciones , Pez Cebra/crecimiento & desarrollo , Cigoto/crecimiento & desarrollo , Cigoto/metabolismo
10.
BMC Dev Biol ; 12: 37, 2012 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-23244389

RESUMEN

BACKGROUND: Genetic studies in mouse have demonstrated the crucial function of PAX4 in pancreatic cell differentiation. This transcription factor specifies ß- and δ-cell fate at the expense of α-cell identity by repressing Arx gene expression and ectopic expression of PAX4 in α-cells is sufficient to convert them into ß-cells. Surprisingly, no Pax4 orthologous gene can be found in chicken and Xenopus tropicalis raising the question of the function of pax4 gene in lower vertebrates such as in fish. In the present study, we have analyzed the expression and the function of the orthologous pax4 gene in zebrafish. RESULTS: pax4 gene is transiently expressed in the pancreas of zebrafish embryos and is mostly restricted to endocrine precursors as well as to some differentiating δ- and ε-cells but was not detected in differentiating ß-cells. pax4 knock-down in zebrafish embryos caused a significant increase in α-cells number while having no apparent effect on ß- and δ-cell differentiation. This rise of α-cells is due to an up-regulation of the Arx transcription factor. Conversely, knock-down of arx caused to a complete loss of α-cells and a concomitant increase of pax4 expression but had no effect on the number of ß- and δ-cells. In addition to the mutual repression between Arx and Pax4, these two transcription factors negatively regulate the transcription of their own gene. Interestingly, disruption of pax4 RNA splicing or of arx RNA splicing by morpholinos targeting exon-intron junction sites caused a blockage of the altered transcripts in cell nuclei allowing an easy characterization of the arx- and pax4-deficient cells. Such analyses demonstrated that arx knock-down in zebrafish does not lead to a switch of cell fate, as reported in mouse, but rather blocks the cells in their differentiation process towards α-cells. CONCLUSIONS: In zebrafish, pax4 is not required for the generation of the first ß- and δ-cells deriving from the dorsal pancreatic bud, unlike its crucial role in the differentiation of these cell types in mouse. On the other hand, the mutual repression between Arx and Pax4 is observed in both mouse and zebrafish. These data suggests that the main original function of Pax4 during vertebrate evolution was to modulate the number of pancreatic α-cells and its role in ß-cells differentiation appeared later in vertebrate evolution.


Asunto(s)
Embrión no Mamífero/citología , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Paired Box/metabolismo , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Morfolinos/farmacología , Factores de Transcripción Paired Box/biosíntesis , Factores de Transcripción Paired Box/genética , Páncreas/embriología , Empalme del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/biosíntesis , Proteínas de Pez Cebra/biosíntesis
11.
Dev Biol ; 340(2): 269-82, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20144609

RESUMEN

Hox genes are key regulators of anterior-posterior axis patterning and have a major role in hindbrain development. The zebrafish Hox4 paralogs have strong overlapping activities in hindbrain rhombomeres 7 and 8, in the spinal cord and in the pharyngeal arches. With the aim to predict enhancers that act on the hoxa4a, hoxb4a, hoxc4a and hoxd4a genes, we used sequence conservation around the Hox4 genes to analyze all fish:human conserved non-coding sequences by reporter assays in stable zebrafish transgenesis. Thirty-four elements were functionally tested in GFP reporter gene constructs and more than 100 F1 lines were analyzed to establish a correlation between sequence conservation and cis-regulatory function, constituting a catalog of Hox4 CNEs. Sixteen tissue-specific enhancers could be identified. Multiple alignments of the CNEs revealed paralogous cis-regulatory sequences, however, the CNE sequence similarities were found not to correlate with tissue specificity. To identify ancestral enhancers that direct Hox4 gene activity, genome sequence alignments of mammals, teleosts, horn shark and the cephalochordate amphioxus, which is the most basal extant chordate possessing a single prototypical Hox cluster, were performed. Three elements were identified and two of them exhibited regulatory activity in transgenic zebrafish, however revealing no specificity. Our data show that the approach to identify cis-regulatory sequences by genome sequence alignments and subsequent testing in zebrafish transgenesis can be used to define enhancers within the Hox clusters and that these have significantly diverged in their function during evolution.


Asunto(s)
Genes Homeobox , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Secuencia Conservada/genética , ADN/genética , ADN/aislamiento & purificación , Embrión no Mamífero/metabolismo , Elementos de Facilitación Genéticos , Evolución Molecular , Duplicación de Gen , Genes Reporteros , Genoma , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Datos de Secuencia Molecular , Familia de Multigenes , Filogenia , Alineación de Secuencia , Análisis de Secuencia de ADN , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
12.
Dev Neurobiol ; 76(5): 507-18, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26184457

RESUMEN

During early development of the central nervous system (CNS), a subset of yolk-sac derived myeloid cells populate the brain and provide the seed for the microglial cell population, which will self-renew throughout life. As development progresses, individual microglial cells transition from a phagocytic amoeboid state through a transitional morphing phase into the sessile, ramified, and normally nonphagocytic microglia observed in the adult CNS under healthy conditions. The molecular drivers of this tissue-specific maturation profile are not known. However, a survey of tissue resident macrophages identified miR-124 to be expressed in microglia. In this study, we used transgenic zebrafish to overexpress miR-124 in the mpeg1 expressing yolk-sac-derived myeloid cells that seed the microglia. In addition, a systemic sponge designed to neutralize the effects of miR-124 was used to assess microglial development in a miR-124 loss-of-function environment. Following the induction of miR-124 overexpression, microglial motility and phagocytosis of apoptotic cells were significantly reduced. miR-124 overexpression in microglia resulted in the accumulation of residual apoptotic cell bodies in the optic tectum, which could not be achieved by miR-124 overexpression in differentiated neurons. Conversely, expression of the miR-124 sponge caused an increase in the motility of microglia and transiently rescued motility and phagocytosis functions when activated simultaneously with miR-124 overexpression. This study provides in vivo evidence that miR-124 activity has a key role in the development of functionally mature microglia.


Asunto(s)
MicroARNs/metabolismo , Microglía/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Técnicas de Silenciamiento del Gen , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/genética , Neuronas/metabolismo , Fagocitosis/fisiología , Homología de Secuencia , Colículos Superiores/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
13.
Nat Commun ; 6: 7378, 2015 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-26051838

RESUMEN

Although zebrafish is used to model human diseases through mutational and morpholino-based knockdown approaches, there are currently no robust transgenic knockdown tools. Here we investigate the knockdown efficiency of three synthetic miRNA-expressing backbones and show that these constructs can downregulate a sensor transgene with different degrees of potency. Using this approach, we reproduce spinal muscular atrophy (SMA) in zebrafish by targeting the smn1 gene. We also generate different transgenic lines, with severity and age of onset correlated to the level of smn1 inhibition, recapitulating for the first time the different forms of SMA in zebrafish. These lines are proof-of-concept that miRNA-based approaches can be used to generate potent heritable gene knockdown in zebrafish.


Asunto(s)
Técnicas de Silenciamiento del Gen , MicroARNs/genética , Pez Cebra/genética , Regiones no Traducidas 3' , Animales , Animales Modificados Genéticamente , Proteínas de Pez Cebra/genética
14.
Nat Commun ; 6: 6904, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25908307

RESUMEN

Enhancers can regulate the transcription of genes over long genomic distances. This is thought to lead to selection against genomic rearrangements within such regions that may disrupt this functional linkage. Here we test this concept experimentally using the human X chromosome. We describe a scoring method to identify evolutionary maintenance of linkage between conserved noncoding elements and neighbouring genes. Chromatin marks associated with enhancer function are strongly correlated with this linkage score. We test >1,000 putative enhancers by transgenesis assays in zebrafish to ascertain the identity of the target gene. The majority of active enhancers drive a transgenic expression in a pattern consistent with the known expression of a linked gene. These results show that evolutionary maintenance of linkage is a reliable predictor of an enhancer's function, and provide new information to discover the genetic basis of diseases caused by the mis-regulation of gene expression.


Asunto(s)
Cromosomas Humanos X/genética , Elementos de Facilitación Genéticos/genética , Expresión Génica/genética , Ligamiento Genético/genética , Selección Genética/genética , Animales , Animales Modificados Genéticamente , Evolución Molecular , Reordenamiento Génico/genética , Humanos , Pez Cebra
15.
Small GTPases ; 5(3): 1-15, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25425145

RESUMEN

The formation of the vascular network requires a tightly controlled balance of pro-angiogenic and stabilizing signals. Perturbation of this balance can result in dysregulated blood vessel morphogenesis and drive pathologies including cancer. Here, we have identified a novel gene, ARHGAP18, as an endogenous negative regulator of angiogenesis, limiting pro-angiogenic signaling and promoting vascular stability. Loss of ARHGAP18 promotes EC hypersprouting during zebrafish and murine retinal vessel development and enhances tumor vascularization and growth. Endogenous ARHGAP18 acts specifically on RhoC and relocalizes to the angiogenic and destabilized EC junctions in a ROCK dependent manner, where it is important in reaffirming stable EC junctions and suppressing tip cell behavior, at least partially through regulation of tip cell genes, Dll4, Flk-1 and Flt-4. These findings highlight ARHGAP18 as a specific RhoGAP to fine tune vascular morphogenesis, limiting tip cell formation and promoting junctional integrity to stabilize the angiogenic architecture.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Uniones Intercelulares/metabolismo , Melanoma Experimental/irrigación sanguínea , Neovascularización Fisiológica , Proteínas de Unión al GTP rho/metabolismo , Animales , Línea Celular Tumoral , Células Endoteliales/metabolismo , Proteínas Activadoras de GTPasa/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Retina/citología , Retina/metabolismo , Retina/patología , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
16.
Zebrafish ; 10(3): 447-50, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23808385

RESUMEN

Abstract Our zebrafish colony experienced a period of increased mortality rate of 6.5 times more deaths per month in a colony of over 13,000 zebrafish (Danio rerio), which developed over 3 months. We observed that before death, affected fish appeared emaciated, often with an abdominal bulge. We performed dissection on 18 fish that had this appearance and found in 15 that their gut was infected with a nematode that closely resembled Pseudocapillaria tomentosa. We devised a treatment protocol for this nematode infection, which involved addition of fenbendazole, a drug used to treat nematode infections in cattle and sheep, to the fish feed. Fenbendazole produced no severe side effects in the fish and several treatments have effectively eradicated the parasite from our colony. The mortality rate of our fish has decreased to a value of 0.7%/month (p<0.001, equal to that before the infection). We propose this protocol as an inexpensive alternative to having to rederive an entire colony from bleached eggs, and as a prophylactic measure used in quarantine facilities on a regular basis.


Asunto(s)
Antinematodos/uso terapéutico , Infecciones por Enoplida/veterinaria , Fenbendazol/uso terapéutico , Parasitosis Intestinales/veterinaria , Pez Cebra/parasitología , Crianza de Animales Domésticos , Animales , Infecciones por Enoplida/tratamiento farmacológico , Femenino , Parasitosis Intestinales/tratamiento farmacológico , Masculino , Trichuroidea/aislamiento & purificación
17.
Dev Neurobiol ; 73(1): 60-71, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22648905

RESUMEN

Microglia, the resident macrophage precursors of the brain, are necessary for the maintenance of tissue homeostasis and activated by a wide range of pathological stimuli. They have a key role in immune and inflammatory responses. Early microglia stem from primitive macrophages, however the transition from early motile forms to the ramified mature resident microglia has not been assayed in real time. In order to provide such an assay, we used zebrafish transgenic lines in which fluorescent reporter expression is driven by the promoter of macrophage expressed gene 1 (mpeg1; Ellet et al. [2011]: Blood 117(4): e49-e56,). This enabled the investigation of the development of these cells in live, intact larvae. We show that microglia develop from highly motile amoeboid cells that are engaged in phagocytosis of apoptotic cell bodies into a microglial cell type that rapidly morphs back and forth between amoeboid and ramified morphologies. These morphing microglia eventually settle into a typical mature ramified morphology. Developing microglia frequently come into contact with blood capillaries in the brain, and also frequently contact each other. Up to 10 days postfertilization, microglia were observed to undergo symmetric division. In the adult optic tectum, the microglia are highly branched, resembling mammalian microglia. In addition, the mpeg1 transgene also labeled highly branched cells in the skin overlying the optic tectum from 8-9 days postfertilization, which likely represent Langerhans cells. Thus, the development of zebrafish microglia and their cellular interactions was studied in the intact developing brain in real time and at cellular resolution.


Asunto(s)
Macrófagos/fisiología , Microglía/fisiología , Colículos Superiores , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Anexina A5/metabolismo , Apoptosis/genética , Encéfalo/citología , Encéfalo/embriología , Recuento de Células , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Luminiscentes/genética , Proteínas de la Membrana/genética , Microscopía Confocal , Fagocitos/fisiología , Colículos Superiores/citología , Colículos Superiores/embriología , Colículos Superiores/crecimiento & desarrollo , Factores de Tiempo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteína Fluorescente Roja
18.
Dev Neurobiol ; 72(3): 415-28, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21465670

RESUMEN

Whole exome sequencing and, to a lesser extent, genome-wide association studies, have provided unprecedented advances in identifying genes and candidate genomic regions involved in the development of human disease. Further progress will come from sequencing the entire genome of multiple patients and normal controls to evaluate overall mutational burden and disease risk. A major challenge will be the interpretation of the resulting data and distinguishing true pathogenic mutations from rare benign variants.While in model organisms such as the zebrafish,mutants are sought that disrupt the function of individual genes, human mutations that cause, or are associated with, the development of disease, are often not acting in a Mendelian fashion, are frequently of small effect size, are late onset, and may reside in noncoding parts of the genome. The zebrafish model is uniquely poised for understanding human coding- and noncoding variants because of its sequenced genome, a large body of knowledge on gene expression and function, rapid generation time, and easy access to embryos. A critical advantage is the ease of zebrafish transgenesis, both for the testing of human regulatory DNA driving expression of fluorescent reporter proteins, and the expression of mutated disease-associated human proteins in specific neurons to rapidly model aspects of neurological disorders. The zebrafish affords progress both through its model genome and it is rapidly developing transparent model vertebrate embryo.


Asunto(s)
Modelos Animales de Enfermedad , Genómica/métodos , Herencia Multifactorial/genética , Enfermedades del Sistema Nervioso/genética , Pez Cebra/genética , Animales , Estudio de Asociación del Genoma Completo/métodos , Humanos , Enfermedades del Sistema Nervioso/fisiopatología
19.
Pigment Cell Melanoma Res ; 25(1): 28-36, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21883983

RESUMEN

Greying with age in horses is an autosomal dominant trait, characterized by hair greying, high incidence of melanoma and vitiligo-like depigmentation. Previous studies have revealed that the causative mutation for this phenotype is a 4.6-kb intronic duplication in STX17 (Syntaxin 17). By using reporter constructs in transgenic zebrafish, we show that a construct containing two copies of the duplicated sequence acts as a strong enhancer in neural crest cells and has subsequent melanophore-specific activity during zebrafish embryonic development whereas a single copy of the duplicated sequence acts as a weak enhancer, consistent with the phenotypic manifestation of the mutation in horses. We further used luciferase assays to investigate regulatory regions in the duplication, to reveal tissue-specific activities of these elements. One region upregulated the reporter gene expression in a melanocyte-specific manner and contained two microphthalmia-associated transcription factor (MITF) binding sites, essential for the activity. Microphthalmia-associated transcription factor regulates melanocyte development, and these binding sites are outstanding candidates for mediating the melanocyte-specific activity of the element. These results provide strong support for the causative nature of the duplication and constitute an explanation for the melanocyte-specific effects of the Grey allele.


Asunto(s)
Elementos de Facilitación Genéticos , Duplicación de Gen , Color del Cabello/genética , Enfermedades de los Caballos/genética , Caballos/genética , Intrones/genética , Melanocitos/metabolismo , Melanoma/veterinaria , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Qa-SNARE/genética , Neoplasias Cutáneas/veterinaria , Envejecimiento/genética , Animales , Animales Modificados Genéticamente , Sitios de Unión , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Genes Dominantes , Genes Reporteros , Humanos , Mamíferos , Melanoma/genética , Melanóforos/metabolismo , Cresta Neural/citología , Fenotipo , Proteínas Qa-SNARE/fisiología , Neoplasias Cutáneas/genética , Especificidad de la Especie , Pez Cebra
20.
Prog Neurobiol ; 93(2): 231-43, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21130139

RESUMEN

Rapid technological advances over the past decade have moved us closer to a high throughput molecular approach to neurobiology, where we see the merging of neurogenetics, genomics, physiology, imaging and pharmacology. This is the case more in zebrafish than in any other model organism commonly used. Recent improvements in the generation of transgenic zebrafish now allow genetic manipulation and live imaging of neuronal development and function in early embryonic, larval, and adult animals. The sequenced zebrafish genome and comparative genomics give unprecedented insights into genome evolution and its relation to genome structure and function. There is now information on embryonic and larval expression of over 12,000 genes and just under 1000 mutant phenotypes. We review the remarkable similarity of the zebrafish genetic blueprint for the nervous system to that of mammals and assess recent technological advances that make the zebrafish a model of choice for elucidating the development and function of neuronal circuitry, transgene-based neuroanatomy, and small molecule neuropharmacology.


Asunto(s)
Genómica , Neurobiología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Expresión Génica , Humanos , Red Nerviosa/anatomía & histología , Red Nerviosa/fisiología , Sistema Nervioso/anatomía & histología , Sistema Nervioso/embriología , Sistema Nervioso/crecimiento & desarrollo , Neurotransmisores/metabolismo , Transgenes , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA