Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 474(7353): 645-8, 2011 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-21623370

RESUMEN

The four-chambered mammalian heart develops from two fields of cardiac progenitor cells distinguished by their spatiotemporal patterns of differentiation and contributions to the definitive heart. The first heart field differentiates earlier in lateral plate mesoderm, generates the linear heart tube and ultimately gives rise to the left ventricle. The second heart field (SHF) differentiates later in pharyngeal mesoderm, elongates the heart tube, and gives rise to the outflow tract and much of the right ventricle. Because hearts in lower vertebrates contain a rudimentary outflow tract but not a right ventricle, the existence and function of SHF-like cells in these species has remained a topic of speculation. Here we provide direct evidence from Cre/Lox-mediated lineage tracing and loss-of-function studies in zebrafish, a lower vertebrate with a single ventricle, that latent TGF-ß binding protein 3 (ltbp3) transcripts mark a field of cardiac progenitor cells with defining characteristics of the anterior SHF in mammals. Specifically, ltbp3(+) cells differentiate in pharyngeal mesoderm after formation of the heart tube, elongate the heart tube at the outflow pole, and give rise to three cardiovascular lineages in the outflow tract and myocardium in the distal ventricle. In addition to expressing Ltbp3, a protein that regulates the bioavailability of TGF-ß ligands, zebrafish SHF cells co-express nkx2.5, an evolutionarily conserved marker of cardiac progenitor cells in both fields. Embryos devoid of ltbp3 lack the same cardiac structures derived from ltbp3(+) cells due to compromised progenitor proliferation. Furthermore, small-molecule inhibition of TGF-ß signalling phenocopies the ltbp3-morphant phenotype whereas expression of a constitutively active TGF-ß type I receptor rescues it. Taken together, our findings uncover a requirement for ltbp3-TGF-ß signalling during zebrafish SHF development, a process that serves to enlarge the single ventricular chamber in this species.


Asunto(s)
Corazón/embriología , Proteínas de Unión a TGF-beta Latente/metabolismo , Miocardio/metabolismo , Pez Cebra/embriología , Animales , Anomalías Cardiovasculares/embriología , Linaje de la Célula , Técnicas de Silenciamiento del Gen , Proteína Homeótica Nkx-2.5 , Datos de Secuencia Molecular , Miocardio/citología , Fenotipo , Transducción de Señal , Factores de Transcripción/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
BMC Dev Biol ; 15: 50, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26715205

RESUMEN

BACKGROUND: The vertebrate heart consists of three cell layers: the innermost endothelium, the contractile myocardium and the outermost epicardium. The epicardium is vital for heart development and function, and forms from epicardial progenitor cells (EPCs), which migrate to the myocardium during early development. Disruptions in EPC migration and epicardium formation result in a number of cardiac malformations, many of which resemble congenital heart diseases in humans. Hence, it is important to understand the mechanisms that influence EPC migration and spreading in the developing heart. In vitro approaches heretofore have been limited to monolayer epicardial cell cultures, which may not fully capture the complex interactions that can occur between epicardial and myocardial cells in vivo. RESULTS: Here we describe a novel in vitro co-culture assay for assessing epicardial cell migration using embryonic zebrafish hearts. We isolated donor hearts from embryonic zebrafish carrying an epicardial-specific fluorescent reporter after epicardial cells were present on the heart. These were co-cultured with recipient hearts expressing a myocardial-specific fluorescent reporter, isolated prior to EPC migration. Using this method, we can clearly visualize the movement of epicardial cells from the donor heart onto the myocardium of the recipient heart. We demonstrate the utility of this method by showing that epicardial cell migration is significantly delayed or absent when myocardial cells lack contractility and when myocardial cells are deficient in tbx5 expression. CONCLUSIONS: We present a method to assess the migration of epicardial cells in an in vitro assay, wherein the migration of epicardial cells from a donor heart onto the myocardium of a recipient heart in co-culture is monitored and scored. The donor and recipient hearts can be independently manipulated, using either genetic tools or pharmacological agents. This allows flexibility in experimental design for determining the role that target genes/signaling pathways in specific cell types may have on epicardial cell migration.


Asunto(s)
Movimiento Celular/fisiología , Corazón/embriología , Organogénesis/fisiología , Pericardio/fisiología , Pez Cebra/embriología , Animales , Proliferación Celular , Técnicas de Cocultivo , Embrión no Mamífero/embriología , Cardiopatías Congénitas/embriología , Trasplante de Corazón/métodos , Miocardio/metabolismo , Técnicas de Cultivo de Órganos , Pericardio/citología , Proteínas de Dominio T Box/genética
3.
BMC Dev Biol ; 14: 18, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24885804

RESUMEN

BACKGROUND: The outermost layer of the vertebrate heart, the epicardium, forms from a cluster of progenitor cells termed the proepicardium (PE). PE cells migrate onto the myocardium to give rise to the epicardium. Impaired epicardial development has been associated with defects in valve development, cardiomyocyte proliferation and alignment, cardiac conduction system maturation and adult heart regeneration. Zebrafish are an excellent model for studying cardiac development and regeneration; however, little is known about how the zebrafish epicardium forms. RESULTS: We report that PE migration occurs through multiple mechanisms and that the zebrafish epicardium is composed of a heterogeneous population of cells. Heterogeneity is first observed within the PE and persists through epicardium formation. Using in vivo imaging, histology and confocal microscopy, we show that PE cells migrate through a cellular bridge that forms between the pericardial mesothelium and the heart. We also observed the formation of PE aggregates on the pericardial surface, which were released into the pericardial cavity. It was previously reported that heartbeat-induced pericardiac fluid advections are necessary for PE cluster formation and subsequent epicardium development. We manipulated heartbeat genetically and pharmacologically and found that PE clusters clearly form in the absence of heartbeat. However, when heartbeat was inhibited the PE failed to migrate to the myocardium and the epicardium did not form. We isolated and cultured hearts with only a few epicardial progenitor cells and found a complete epicardial layer formed. However, pharmacologically inhibiting contraction in culture prevented epicardium formation. Furthermore, we isolated control and silent heart (sih) morpholino (MO) injected hearts prior to epicardium formation (60 hpf) and co-cultured these hearts with "donor" hearts that had an epicardium forming (108 hpf). Epicardial cells from donor hearts migrated on to control but not sih MO injected hearts. CONCLUSIONS: Epicardial cells stem from a heterogeneous population of progenitors, suggesting that the progenitors in the PE have distinct identities. PE cells attach to the heart via a cellular bridge and free-floating cell clusters. Pericardiac fluid advections are not necessary for the development of the PE cluster, however heartbeat is required for epicardium formation. Epicardium formation can occur in culture without normal hydrodynamic and hemodynamic forces, but not without contraction.


Asunto(s)
Movimiento Celular , Modelos Biológicos , Pericardio/citología , Células Madre/citología , Animales , Animales Modificados Genéticamente , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Frecuencia Cardíaca/fisiología , Inmunohistoquímica , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Organogénesis , Pericardio/embriología , Pericardio/fisiología , Células Madre/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Pez Cebra/embriología , Pez Cebra/metabolismo , Pez Cebra/fisiología
4.
J Proteome Res ; 12(7): 3093-103, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23682714

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental pollutant and teratogen that produces cardiac toxicity in the developing zebrafish. Here we adopted a label free quantitative proteomic approach based on normalized spectral abundance factor (NSAF) to investigate the disturbance of the cardiac proteome induced by TCDD in the adult zebrafish heart. The protein expression level changes between heart samples from TCDD-treated and control zebrafish were systematically evaluated by a large scale MudPIT analysis, which incorporated triplicate analyses for both control and TCDD-exposed heart proteomic samples to overcome the data-dependent variation in shotgun proteomic experiments and obtain a statistically significant protein data set with improved quantification confidence. A total of 519 and 443 proteins were identified in hearts collected from control and TCDD-treated zebrafish, respectively, among which 106 proteins showed statistically significant expression changes. After correcting for the experimental variation between replicate analyses by statistical evaluation, 55 proteins exhibited NSAF ratios above 2 and 43 proteins displayed NSAF ratios smaller than 0.5, with statistical significance by t test (p < 0.05). The proteins identified as altered by TCDD encompass a wide range of biological functions including calcium handling, myocardium cell architecture, energy production and metabolism, mitochondrial homeostasis, and stress response. Collectively, our results indicate that TCDD exposure alters the adult zebrafish heart in a way that could result in cardiac hypertrophy and heart failure and suggests a potential mechanism for the diastolic dysfunction observed in TCDD-exposed embryos.


Asunto(s)
Corazón/crecimiento & desarrollo , Dibenzodioxinas Policloradas/toxicidad , Proteínas/aislamiento & purificación , Pez Cebra/crecimiento & desarrollo , Animales , Embrión no Mamífero , Contaminantes Ambientales/toxicidad , Corazón/efectos de los fármacos , Proteómica
5.
Mol Pharmacol ; 84(3): 353-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23775563

RESUMEN

Activation of the transcription factor aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) prevents the formation of the epicardium and leads to severe heart malformations in developing zebrafish (Danio rerio). The downstream genes that cause heart malformation are not known. Because TCDD causes craniofacial malformations in zebrafish by downregulating the sox9b gene, we hypothesized that cardiotoxicity might also result from sox9b downregulation. We found that sox9b is expressed in the developing zebrafish heart ventricle and that TCDD exposure markedly reduces this expression. Furthermore, we found that manipulation of sox9b expression could phenocopy many but not all of the effects of TCDD at the heart. Loss of sox9b prevented the formation of epicardium progenitors comprising the proepicardium on the pericardial wall, and prevented the formation and migration of the epicardial layer around the heart. Zebrafish lacking sox9b showed pericardial edema, an elongated heart, and reduced blood circulation. Fish lacking sox9b failed to form valve cushions and leaflets. Sox9b is one of two mammalian Sox9 homologs, sox9b and sox9a. Knock down of sox9a expression did not cause cardiac malformations, or defects in epicardium development. We conclude that the decrease in sox9b expression in the heart caused by TCDD plays a role in many of the observed signs of cardiotoxicity. We find that while sox9b is expressed in myocardial cells, it is not normally expressed in the affected epicardial cells or progenitors. We therefore speculate that sox9b is involved in signals between the cardiomyocytes and the nascent epicardial cells.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Cardiopatías Congénitas/inducido químicamente , Pericardio/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Factor de Transcripción SOX9/metabolismo , Proteínas de Pez Cebra/metabolismo , Anomalías Inducidas por Medicamentos/fisiopatología , Animales , Circulación Coronaria , Regulación hacia Abajo , Edema/inducido químicamente , Edema/metabolismo , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/fisiopatología , Válvulas Cardíacas/anomalías , Válvulas Cardíacas/efectos de los fármacos , Válvulas Cardíacas/embriología , Válvulas Cardíacas/crecimiento & desarrollo , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pericardio/embriología , Pericardio/crecimiento & desarrollo , Pericardio/metabolismo , Pez Cebra
6.
Environ Sci Technol ; 47(9): 4726-33, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23510150

RESUMEN

Photoactivation of titanium dioxide nanoparticles (TiO2NPs) can produce reactive oxygen species (ROS). Over time, this has the potential to produce cumulative cellular damage. To test this, we exposed zebrafish (Danio rerio) to two commercial TiO2NP preparations at concentrations ranging from 0.01 to 10,000 ng/mL over a 23 day period spanning embryogenesis, larval development, and juvenile metamorphosis. Fish were illuminated with a lamp that mimics solar irradiation. TiO2NP exposure produced significant mortality at 1 ng/mL. Toxicity included stunted growth, delayed metamorphosis, malformations, organ pathology, and DNA damage. TiO2NPs were found in the gills and gut and elsewhere. The two preparations differed in nominal particle diameter (12.1 ± 3.7 and 23.3 ± 9.8 nm) but produced aggregates in the 1 µm range. Both were taken up in a dose-dependent manner. Illuminated particles produced a time- and dose-dependent increase in 8-hydroxy-2'-deoxyguanosine DNA adducts consistent with cumulative ROS damage. Zebrafish take up TiO2NPs from the aqueous environment even at low ng/mL concentrations, and these particles when illuminated in the violet-near UV range produce cumulative toxicity.


Asunto(s)
Nanopartículas del Metal , Titanio/toxicidad , Pez Cebra/embriología , Animales , Microscopía Electrónica de Transmisión , Titanio/análisis , Pez Cebra/genética
7.
Environ Sci Technol ; 47(9): 4718-25, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23347333

RESUMEN

Titanium dioxide nanoparticle (TiO2NP) suspension stability can be altered by adsorption of dissolved organic matter (DOM). This is expected to impact their environmental fate and bioavailability. To date, the influence of DOM on the toxicity of TiO2NPs to aquatic vertebrates has not been reported. We examined the impact of Suwannee River humic acid (HA) on the toxicity of TiO2NPs to developing zebrafish (Danio rerio) in the dark and under simulated sunlight illumination. Adsorption of HA increased suspension stability and decreased TiO2NP exposure. TiO2NPs were more toxic in the presence of HA. In the absence of simulated sunlight, a small but significant increase in lethality was observed in fish exposed to TiO2NPs in the presence of HA. Under simulated sunlight illumination, photocatalytic degradation of HA reduced suspension stability. Despite the lower concentrations of Ti associated with fish in the treatments containing HA, under simulated sunlight illumination, median lethal concentrations were lower and oxidative DNA damage was elevated relative to fish exposed to TiO2NPs in the absence of HA. This study demonstrates the importance of considering environmental factors (i.e., exposure to sunlight, adsorption of DOM) when assessing the potential risks posed by engineered nanomaterials in the environment.


Asunto(s)
Sustancias Húmicas , Nanopartículas del Metal/toxicidad , Titanio/toxicidad , Animales , Ambiente , Pez Cebra
8.
Environ Sci Technol ; 47(16): 9132-9, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23815598

RESUMEN

Once released into the environment, engineered nanoparticles (eNPs) are subjected to processes that may alter their physical or chemical properties, potentially altering their toxicity vis-à-vis the as-synthesized materials. We examined the toxicity to zebrafish ( Danio rerio ) embryos of CdSecore/ZnSshell quantum dots (QDs) before and after exposure to an in vitro chemical model designed to simulate oxidative weathering in soil environments based on a reductant-driven Fenton's reaction. Exposure to these oxidative conditions resulted in severe degradation of the QDs: the Zn shell eroded, Cd(2+) and selenium were released, and amorphous Se-containing aggregates were formed. Products of QD weathering exhibited higher potency than did as-synthesized QDs. Morphological endpoints of toxicity included pericardial, ocular and yolk sac edema, nondepleted yolk, spinal curvature, tail malformations, and craniofacial malformations. To better understand the selenium-like toxicity observed in QD exposures, we examined the toxicity of selenite, selenate, and amorphous selenium nanoparticles (SeNPs). Selenite exposures resulted in high mortality to embryos/larvae while selenate and SeNPs were nontoxic. Co-exposures to SeNPs + CdCl2 resulted in dramatic increase in mortality and recapitulated the morphological endpoints of toxicity observed with exposure to products of QD weathering. Cadmium body burden was increased in larvae exposed to weathered QDs or SeNP + CdCl2 suggesting the increased potency of products of QD weathering was due to selenium modulation of cadmium toxicity. Our findings highlight the need to examine the toxicity of eNPs after they have undergone environmental weathering processes.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Puntos Cuánticos/toxicidad , Animales , Cadmio/toxicidad , Oxidación-Reducción , Compuestos de Selenio/toxicidad , Pruebas de Toxicidad , Pez Cebra
9.
J Sep Sci ; 33(10): 1462-71, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20235133

RESUMEN

2D HPLC separations by coupling strong cation exchange (SCX) and RP fractionation have been widely used in large-scale proteomic studies. Traditionally this method is performed by salt gradient SCX separation followed by RP and MS/MS analysis. The salt gradient SCX method has been known to have low peptide and protein resolution. In this study, we implemented a pH gradient SCX-RP HPLC platform to separate proteome digests from adult zebrafish hearts, followed by ESI quadrupole-TOF MS/MS analysis. This pH gradient SCX method has improved peptide separation, as demonstrated by a greater number of peptides and proteins identified from individual SCX fractions. This pH gradient method also has better MS compatibility owing to lower salt usage. This setup allows fast microflow fractionation in SCX dimension and nanoflow RP separation in the second dimension, and can be easily implemented on conventional capillary LC ESI MS/MS systems. Using this setup, we identified 1375 proteins from adult zebrafish hearts, establishing the first reported experimental data set for the heart proteome of zebrafish. This work laid the foundation for further studies of environmental cardiac toxicology using zebrafish as a model organism.


Asunto(s)
Envejecimiento/metabolismo , Miocardio/química , Proteoma/análisis , Proteínas de Pez Cebra/análisis , Pez Cebra/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Cromatografía de Fase Inversa , Concentración de Iones de Hidrógeno , Miocardio/metabolismo , Espectrometría de Masa por Ionización de Electrospray
10.
Eukaryot Cell ; 7(2): 358-67, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18156291

RESUMEN

Nutrient repletion leads to substantial restructuring of the transcriptome in Saccharomyces cerevisiae. The expression levels of approximately one-third of all S. cerevisiae genes are altered at least twofold when a nutrient-depleted culture is transferred to fresh medium. Several nutrient-sensing pathways are known to play a role in this process, but the relative contribution that each pathway makes to the total response has not been determined. To better understand this, we used a chemical-genetic approach to block the protein kinase A (PKA), TOR (target of rapamycin), and glucose transport pathways, alone and in combination. Of the three pathways, we found that loss of PKA produced the largest effect on the transcriptional response; however, many genes required both PKA and TOR for proper nutrient regulation. Those genes that did not require PKA or TOR for nutrient regulation were dependent on glucose transport for either nutrient induction or repression. Therefore, loss of these three pathways is sufficient to prevent virtually the entire transcriptional response to fresh medium. In the absence of fresh medium, activation of the cyclic AMP/PKA pathway does not induce cellular growth; nevertheless, PKA activation induced a substantial fraction of the PKA-dependent genes. In contrast, the absence of fresh medium strongly limited gene repression by PKA. These results account for the signals needed to generate the transcriptional responses to glucose, including induction of growth genes required for protein synthesis and repression of stress genes, as well as the classical glucose repression and hexose transporter responses.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Fúngica de la Expresión Génica , Glucosa/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transporte Biológico , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Biosíntesis de Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal , Transcripción Genética
11.
Mol Pharmacol ; 74(6): 1544-53, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18784347

RESUMEN

Exposure to environmental contaminants can disrupt normal development of the early vertebrate skeleton. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) impairs craniofacial skeletal development across many vertebrate species, and its effects are especially prominent in early life stages of fish. TCDD activates the aryl hydrocarbon receptor, a transcription factor that mediates most if not all TCDD responses. We investigated the transcriptional response in the developing zebrafish jaw after TCDD exposure using DNA microarrays. Zebrafish larvae were exposed to TCDD at 96 h after fertilization, and jaw cartilage tissue was harvested for microarray analysis at 1, 2, 4, and 12 h after exposure. Numerous chondrogenic transcripts were misregulated by TCDD in the jaw. Comparison of transcripts altered by TCDD in jaw with transcripts altered in embryonic heart showed that the transcriptional responses in the jaw and the heart were strikingly different. Sox9b, a critical chondrogenic transcription factor, was the most significantly reduced transcript in the jaw. We hypothesized that the TCDD reduction of sox9b expression plays an integral role in affecting the formation of the embryonic jaw. Morpholino knockdown of sox9b expression demonstrated that partial reduction of sox9b expression alone was sufficient to produce a TCDD-like jaw phenotype. Loss of a single copy of the sox9b gene in sox9b(+/-) heterozygotes increased sensitivity to jaw malformation by TCDD. Finally, embryos injected with sox9b mRNA and then exposed to TCDD blocked TCDD-induced jaw toxicity in approximately 14% of sox9b-injected embryos. These results suggest that reduced sox9b expression in TCDD-exposed zebrafish embryos contributes to jaw malformation.


Asunto(s)
Anomalías Maxilomandibulares/embriología , Maxilares/embriología , Receptores de Hidrocarburo de Aril/fisiología , Factor de Transcripción SOX9/biosíntesis , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/embriología , Animales , Dioxinas/toxicidad , Regulación hacia Abajo , Embrión no Mamífero , Contaminantes Ambientales/toxicidad , Anomalías Maxilomandibulares/inducido químicamente , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/farmacología , Factor de Transcripción SOX9/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
12.
Physiol Genomics ; 33(2): 148-58, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18230668

RESUMEN

Retinoic acid (RA) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) activate distinct ligand-dependent transcription factors, and both cause cardiac malformation and heart failure in zebrafish embryos. We hypothesized that they cause this response by hyperactivating a common set of genes critical for heart development. To test this, we used microarrays to measure transcript changes in hearts isolated from zebrafish embryos 1, 2, 4, and 12 h after exposure to 1 muM RA. We used hierarchical clustering to compare the transcriptional responses produced in the embryonic heart by RA and TCDD. We could identify no early responses in common between the two agents. However, at 12 h both treatments produced a dramatic downregulation of a common cluster of cell cycle progression genes, which we term the cell cycle gene cluster. This was associated with a halt in heart growth. These results suggest that RA and TCDD ultimately trigger a common transcriptional response associated with heart failure, but not through the direct activation of a common set of genes. Among the genes rapidly induced by RA was Nr2F5, a member of the COUP-TF family of transcriptional repressors. We found that induction of Nr2F5 was both necessary and sufficient for the cardiotoxic response to RA.


Asunto(s)
Cardiotoxinas/toxicidad , Embrión no Mamífero/efectos de los fármacos , Genómica , Corazón/efectos de los fármacos , Corazón/embriología , Pez Cebra/embriología , Pez Cebra/genética , Animales , Recuento de Células , Edema/genética , Embrión no Mamífero/metabolismo , Fertilización/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos Antisentido/farmacología , Dibenzodioxinas Policloradas/toxicidad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Factores de Tiempo , Tretinoina/toxicidad , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
13.
Sci Rep ; 8(1): 13906, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30224706

RESUMEN

The high mobility group transcription factor SOX9 is expressed in stem cells, progenitor cells, and differentiated cell-types in developing and mature organs. Exposure to a variety of toxicants including dioxin, di(2-ethylhexyl) phthalate, 6:2 chlorinated polyfluorinated ether sulfonate, and chlorpyrifos results in the downregulation of tetrapod Sox9 and/or zebrafish sox9b. Disruption of Sox9/sox9b function through environmental exposures or genetic mutations produce a wide range of phenotypes and adversely affect organ development and health. We generated a dominant-negative sox9b (dnsox9b) to inhibit sox9b target gene expression and used the Gal4/UAS system to drive dnsox9b specifically in cardiomyocytes. Cardiomyocyte-specific inhibition of sox9b function resulted in a decrease in ventricular cardiomyocytes, an increase in atrial cardiomyocytes, hypoplastic endothelial cushions, and impaired epicardial development, ultimately culminating in heart failure. Cardiomyocyte-specific dnsox9b expression significantly reduced end diastolic volume, which corresponded with a decrease in stroke volume, ejection fraction, and cardiac output. Further analysis of isolated cardiac tissue by RT-qPCR revealed cardiomyocyte-specific inhibition of sox9b function significantly decreased the expression of the critical cardiac development genes nkx2.5, nkx2.7, and myl7, as well as c-fos, an immediate early gene necessary for cardiomyocyte progenitor differentiation. Together our studies indicate sox9b transcriptional regulation is necessary for cardiomyocyte development and function.


Asunto(s)
Corazón/embriología , Morfogénesis , Miocitos Cardíacos/metabolismo , Factor de Transcripción SOX9/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Genes Dominantes , Células HEK293 , Humanos , Ratones , Factor de Transcripción SOX9/metabolismo , Transcripción Genética , Pez Cebra
14.
Mol Cell Biol ; 22(5): 1607-14, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11839825

RESUMEN

Transcription of the CLN3 G(1) cyclin in Saccharomyces cerevisiae is positively regulated by glucose in a process that involves a set of DNA elements with the sequence AAGAAAAA (A(2)GA(5)). To identify proteins that interact with these elements, we used a 1-hybrid approach, which yielded a nuclear zinc finger protein previously identified as Azf1. Gel shift and chromatin immunoprecipitation experiments show that Azf1 binds to the A(2)GA(5) CLN3 regulatory sequences in vitro and in vivo, thus identifying a transcriptional regulatory protein for CLN3 and a DNA sequence target for Azf1. We show that glucose-induced expression of a reporter gene driven by the A(2)GA(5) CLN3 regulatory sequences is dependent upon the presence of AZF1. Furthermore, deletion of AZF1 markedly reduces the transcriptional induction of CLN3 by glucose. In addition, Azf1 can induce reporter expression in a glucose-specific manner when artificially tethered to a promoter via the DNA-binding domain from Gal4. We conclude that AZF1 is a glucose-dependent transcription factor that interacts with the CLN3 A(2)GA(5) repeats to play a positive role in the regulation of CLN3 mRNA expression by glucose.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclinas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas/genética , Glucosa/metabolismo , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Factores de Transcripción , Secuencia de Bases , Sitios de Unión , Ciclinas/biosíntesis , Proteínas Fúngicas/biosíntesis , Regulación Fúngica de la Expresión Génica , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Secuencias Repetitivas de Ácidos Nucleicos , Transcripción Genética
15.
Toxicol Sci ; 94(1): 175-82, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16936225

RESUMEN

The zebrafish (Danio rerio) has become an attractive vertebrate model for studying developmental processes, and is emerging as a model system for studying the mechanisms by which xenobiotic compounds perturb normal development. Embryos treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) shortly after fertilization exhibit a range of adverse effects on the heart: an early reduction in cardiac myocyte number, followed by a change in heart looping and morphology, with an apparent compaction of the ventricle and overall decrease in heart size. These changes are accompanied by impaired cardiac function including a decrease in cardiac output and eventually irreversible ventricular standstill. The mechanisms involved in mediating effects of TCDD on the heart remain unknown. However, it is widely accepted that aryl hydrocarbon receptor (AHR) activation mediates endpoints of TCDD toxicity in vertebrates. In zebrafish, there are multiple forms of AHR and AHR nuclear translocator protein (ARNT) raising the question about whether different endpoints of TCDD toxicity are mediated by different components of the AHR/ARNT pathway. To address this question we used morpholino oligonucleotide technology to specifically block the expression of zfAHR2, zfARNT1, zfARNT2, and zfCYP1A, and assessed the previously described effects of TCDD on heart morphology, size, and function in the developing morphants. We report that blocking zfAHR2 and zfARNT1 expression provided protection against the TCDD-mediated alteration in heart morphology, reduced cardiac myocyte number, decreased cardiac output and ventricular standstill in zebrafish larvae, while the zfarnt2 and zfcyp1a morpholinos did not block the TCDD-induced cardiac toxicity.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Cardiopatías Congénitas/prevención & control , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/antagonistas & inhibidores , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Bloqueo Cardíaco/inducido químicamente , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/prevención & control , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/genética , Larva/efectos de los fármacos , Larva/genética , Miocitos Cardíacos/efectos de los fármacos , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/metabolismo , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Función Ventricular/efectos de los fármacos , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo
16.
Gene ; 578(2): 281-9, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26721460

RESUMEN

The SRY-related high-mobility box 9 (SOX9) gene is expressed in many different tissues. To better understand the DNA elements that control tissue-specific expression, we cloned and sequenced a 2.5 kb fragment lying 5' to the zebrafish sox9b gene transcriptional start site. Three regions of this clone contained stable secondary structures that hindered cloning, sequencing, and amplification. This segment and smaller fragmentswere inserted 5' of an EGFP reporter and transgenic fish were raised with the different reporters. Reporter expression was also observed in embryos directly injected with the constructs to transiently express the reporter. Heart expression required only a very short 5' sequence, as a 0.6 kb sox9b fragment produced reporter expression in heart in transgenic zebrafish, and transient experiments showed heart expression from a minimal sox9b promoter region containing a conserved TATA box and an EGR2 element (-74/+29 bp). Reporter expression in transgenic skeletal muscle was consistently lower than in other tissues. Jaw, brain, and notochord expression was strong with the full-length clone, but was dramatically reduced as the size of the fragment driving the reporter decreased from approximately 1.8 to 0.9 kb. The 2.5 kb region 5' of the sox9b contained 7 conserved non-coding elements (CNEs) that included putative hypoxia inducible factor 1α (HIF1α), CAAT box (CCAAT), early growth response protein 2 (EGR2), and core promoter elements. While a synthetic fragment containing all 7 CNEs produced some degree of reporter expression in muscle, jaw, heart and brain, the degree of reporter expression was considerably lower than that produced by the full length clone. These results can account for the tissue-specific expression of sox9b in the developing zebrafish.


Asunto(s)
Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Factor de Transcripción SOX9/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Maxilares/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Notocorda/crecimiento & desarrollo , Notocorda/metabolismo , Factor de Transcripción SOX9/biosíntesis , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/biosíntesis
17.
Toxicol Sci ; 86(1): 6-19, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15703261

RESUMEN

Zebrafish (Danio rerio) has been a prominent model vertebrate in a variety of biological disciplines. Substantial information gathered from developmental and genetic research, together with near-completion of the zebrafish genome project, has placed zebrafish in an attractive position for use as a toxicological model. Although still in its infancy, there is a clear potential for zebrafish to provide valuable new insights into chemical toxicity, drug discovery, and human disease using recent advances in forward and reverse genetic techniques coupled with large-scale, high-throughput screening. Here we present an overview of the rapidly increasing use of zebrafish in toxicology. Advantages of the zebrafish both in identifying endpoints of toxicity and in elucidating mechanisms of toxicity are highlighted.


Asunto(s)
Modelos Animales , Pruebas de Toxicidad , Animales , Células Cultivadas , Análisis de Secuencia por Matrices de Oligonucleótidos , Pez Cebra/embriología , Pez Cebra/genética
18.
Toxicol Sci ; 84(2): 368-77, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15635151

RESUMEN

The zebrafish (Danio rerio) has become an attractive vertebrate model for studying developmental processes, and is emerging as a model system for studying the mechanisms by which toxic compounds perturb normal development. When exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) shortly after fertilization, zebrafish embryos exhibit pericardial edema and reduced blood flow by 72 h post fertilization (hpf). To better understand the progression of dioxin toxicity in zebrafish, we have examined the effects of TCDD on heart development. At 72 hpf, TCDD-treated embryos exhibited altered looping, with the atria positioned distinctly posterior to the ventricles, contrary to the looping of control hearts, where the two chambers lied side by side. Moreover, the ventricles in dioxin-exposed hearts became more compact, and the atria elongated in comparison to controls. These defects are not secondary to pericardial edema because they were observed when edema formation was suppressed with osmotic support. In addition to morphological changes, TCDD produced functional deficits in the developing hearts, including blood regurgitation and a striking ventricular standstill that became prevalent by 120 hpf. We also assessed the effect of TCDD on the heart size using stereological measurements, which demonstrated significant reduction in heart tissue volume at 72 hpf. Perhaps our most significant finding was a decrease in the total number of cardiomyocytes in TCDD-exposed embryos by 48 hpf, one day prior to observable effects on peripheral blood flow. We conclude that the developing heart is an important target for TCDD in zebrafish.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Cardiopatías Congénitas/inducido químicamente , Dibenzodioxinas Policloradas/toxicidad , Teratógenos/toxicidad , Pez Cebra/fisiología , Anomalías Inducidas por Medicamentos/patología , Animales , Miosinas Cardíacas/metabolismo , Recuento de Células , Modelos Animales de Enfermedad , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/fisiología , Técnica del Anticuerpo Fluorescente Indirecta , Cardiopatías Congénitas/patología , Frecuencia Cardíaca/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/metabolismo
19.
Cardiovasc Toxicol ; 5(2): 203-14, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16046794

RESUMEN

Model systems are a mainstay in toxicological research. Zebrafish are rapidly becoming an important model organism for studying vertebrate development. The advantages of zebrafish: short reproductive cycle, production of numerous transparent, synchronously developing embryos, low cost, and standardization make zebrafish an attractive model for toxicologists as well. The use of these fish to study heart development has moved forward very rapidly, laying the groundwork for studying the effects of chemicals on cardiac development and function. Here we describe approaches that can be used to study cardiac toxicity in developing zebrafish, focusing on examples where zebrafish embryos have been especially useful in understanding the impact of specific toxicants on heart development and function.


Asunto(s)
Cardiopatías/inducido químicamente , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/genética , Pez Cebra/fisiología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Cardiopatías/genética , Cardiopatías/patología , Cardiopatías/fisiopatología
20.
Aquat Toxicol ; 162: 10-17, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25766903

RESUMEN

The swim bladder is a gas-filled organ that is used for regulating buoyancy and is essential for survival in most teleost species. In zebrafish, swim bladder development begins during embryogenesis and inflation occurs within 5 days post fertilization (dpf). Embryos exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) before 96 h post fertilization (hpf) developed swim bladders normally until the growth/elongation phase, at which point growth was arrested. It is known that TCDD exposure causes heart malformations that lead to heart failure in zebrafish larvae, and that blood circulation is a key factor in normal development of the swim bladder. The adverse effects of TCDD exposure on the heart occur during the same period of time that swim bladder development and growth occurs. Based on this coincident timing, and the dependence of swim bladder development on proper circulatory development, we hypothesized that the adverse effects of TCDD on swim bladder development were secondary to heart failure. We compared swim bladder development in TCDD-exposed embryos to: (1) silent heart morphants, which lack cardiac contractility, and (2) transiently transgenic cmlc2:caAHR-2AtRFP embryos, which mimic TCDD-induced heart failure via heart-specific, constitutive activation of AHR signaling. Both of these treatment groups, which were not exposed to TCDD, developed hypoplastic swim bladders of comparable size and morphology to those found in TCDD-exposed embryos. Furthermore, in all treatment groups swim bladder development was arrested during the growth/elongation phase. Together, these findings support a potential role for heart failure in the inhibition of swim bladder development caused by TCDD.


Asunto(s)
Sacos Aéreos/efectos de los fármacos , Insuficiencia Cardíaca/inducido químicamente , Corazón/efectos de los fármacos , Organogénesis/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/embriología , Sacos Aéreos/embriología , Animales , Embrión no Mamífero/efectos de los fármacos , Corazón/embriología , Insuficiencia Cardíaca/embriología , Pez Cebra/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA