Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Toxicol Appl Pharmacol ; 284(2): 142-51, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25732252

RESUMEN

DNA methylation is one of the most important epigenetic modifications involved in the regulation of gene expression. The DNA methylation reaction is catalyzed by DNA methyltransferases (DNMTs). Recent studies have demonstrated that toxicants can affect normal development by altering DNA methylation patterns, but the mechanisms of action are poorly understood. Hence, we tested the hypothesis that developmental exposure to TCDD affects dnmt gene expression patterns. Zebrafish embryos were exposed to 5nM TCDD for 1h from 4 to 5h post-fertilization (hpf) and sampled at 12, 24, 48, 72, and 96 hpf to determine dnmt gene expression and DNA methylation patterns. We performed a detailed analysis of zebrafish dnmt gene expression during development and in adult tissues. Our results demonstrate that dnmt3b genes are highly expressed in early stages of development, and dnmt3a genes are more abundant in later stages. TCDD exposure upregulated dnmt1 and dnmt3b2 expression, whereas dnmt3a1, 3b1, and 3b4 are downregulated following exposure. We did not observe any TCDD-induced differences in global methylation or hydroxymethylation levels, but the promoter methylation of aryl hydrocarbon receptor (AHR) target genes was altered. In TCDD-exposed embryos, AHR repressor a (ahrra) and c-fos promoters were differentially methylated. To characterize the TCDD effects on DNMTs, we cloned the dnmt promoters with xenobiotic response elements and conducted AHR transactivation assays using a luciferase reporter system. Our results suggest that ahr2 can regulate dnmt3a1, dnmt3a2, and dnmt3b2 expression. Overall, we demonstrate that developmental exposure to TCDD alters dnmt expression and DNA methylation patterns.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Animales , ADN/genética , Regulación hacia Abajo/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Metiltransferasas/genética , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/genética , Elementos de Respuesta , Regulación hacia Arriba/efectos de los fármacos , Pez Cebra , Proteínas de Pez Cebra/genética
2.
BMC Biotechnol ; 7: 40, 2007 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-17623073

RESUMEN

BACKGROUND: The zebrafish Danio rerio is an important model system for drug discovery and to study cardiovascular development. Using a laser-scanning confocal microscope, we have developed a non-invasive method of measuring cardiac performance in zebrafish embryos and larvae that obtains cardiovascular parameters similar to those obtained using Doppler echocardiography in mammals. A laser scan line placed parallel to the path of blood in the dorsal aorta measures blood cell velocity, from which cardiac output and indices of vascular resistance and contractility are calculated. RESULTS: This technique, called laser-scanning velocimetry, was used to quantify the effects of pharmacological, developmental, and genetic modifiers of cardiac function. Laser-scanning velocimetry was applied to analyze the cardiovascular effects of morpholino knockdown of osmosensing scaffold for MEKK3 (OSM), which when mutated causes the human vascular disease cerebral cavernous malformations. OSM-deficient embryos had a constricted aortic arch and markedly increased peak cell velocity, a characteristic indicator of aortic stenosis. CONCLUSION: These data validate laser-scanning velocimetry as a quantitative tool to measure cardiovascular performance for pharmacological and genetic analysis in zebrafish, which requires no specialized equipment other than a laser-scanning confocal microscope.


Asunto(s)
Corazón/fisiología , Microscopía Confocal/métodos , Pez Cebra/fisiología , Aminobenzoatos/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/embriología , Aorta/fisiología , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Western Blotting , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/fisiología , Regulación del Desarrollo de la Expresión Génica , Prueba de Complementación Genética , Corazón/embriología , Larva/citología , Larva/genética , Larva/fisiología , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/fisiología , Microinyecciones , Morfolinas/farmacología , Mutación , Miocardio/citología , Miocardio/metabolismo , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Pez Cebra/embriología
3.
Toxicol Sci ; 99(1): 326-37, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17562736

RESUMEN

Gene expression profiling is a widely used technique with data from the majority of published microarray studies being publicly available. These data are being used for meta-analyses and in silico discovery; however, the comparability of toxicogenomic data generated in multiple laboratories has not been critically evaluated. Using the power of prospective multilaboratory investigations, seven centers individually conducted a common toxicogenomics experiment designed to advance understanding of molecular pathways perturbed in liver by an acute toxic dose of N-acetyl-p-aminophenol (APAP) and to uncover reproducible genomic signatures of APAP-induced toxicity. The nonhepatotoxic APAP isomer N-acetyl-m-aminophenol was used to identify gene expression changes unique to APAP. Our data show that c-Myc is induced by APAP and that c-Myc-centered interactomes are the most significant networks of proteins associated with liver injury. Furthermore, sources of error and data variability among Centers and methods to accommodate this variability were identified by coupling gene expression with extensive toxicological evaluation of the toxic responses. We show that phenotypic anchoring of gene expression data is required for biologically meaningful analysis of toxicogenomic experiments.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Perfilación de la Expresión Génica/métodos , Expresión Génica/efectos de los fármacos , Genómica/métodos , Hígado/efectos de los fármacos , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Determinación de Punto Final , Islas Genómicas , Isomerismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Reproducibilidad de los Resultados , alfa-Amilasas Salivales , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
4.
Gene Expr Patterns ; 5(3): 363-9, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15661642

RESUMEN

We have cloned the zebrafish ortholog of the mammalian cyp26b1 gene. The predicted zebrafish cyp26b1 protein shares greater than 73% identity with mammalian homologues. cDNA transfection assays showed that like human cyp26b1, zebrafish cyp26b1 is involved in limiting the activity of retinoic acid. Reverse transcription-polymerase chain reaction (RT-PCR) analysis of embryonic RNAs suggested that no maternal cyp26b1 message is detectable. Zygotic cyp26b1 message could be detected at 75% epiboly by RT-PCR and localized to presumptive rhombomere 3 and rhombomere 4 at the early two-somite (2S) stage (10.5 hpf: hour post fertilization) by whole mount in situ hybridization. As development proceeds expression expands anteriorly to include rhombomere 2 at the 10S stage (14hpf). By 14S (16hpf) expression in the hindbrain has also expanded posteriorly and encompasses rhombomere 2 through rhombomere 6. At later stages, 24 through 48 hpf, additional expression was found in the eyes, diencephalon, midbrain-hindbrain boundary, cerebellum, pectoral fin and the pharyngeal arch primordia.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/crecimiento & desarrollo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cerebelo/enzimología , Clonación Molecular , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Diencéfalo/enzimología , Expresión Génica , Hibridación in Situ , Datos de Secuencia Molecular , Ácido Retinoico 4-Hidroxilasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rombencéfalo/enzimología , Transcripción Genética/efectos de los fármacos , Tretinoina/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Mech Dev ; 121(4): 339-50, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15110044

RESUMEN

Retinoic acid (RA) signaling in vertebrate embryos occurs in a distinct physical and temporal pattern. Regulating this spatial distribution is crucial to the development of the embryo, as RA in excess or in inappropriate tissues is teratogenic. In order to understand how RA availability is determined in zebrafish we have investigated the expression of cyp26a1, an enzyme that inactivates RA, and its relationship to raldh2, one of the enzymes that produce RA from retinal. cyp26a1 expression follows three phases: in presumptive anterior neurectoderm and in a circumblastoporal ring during gastrulation, in the tailbud throughout somitogenesis, and in multiple specific tissue types beginning at mid-somitogenesis and continuing through 48 h postfertilization (hpf). This expression was either adjacent or opposite to those tissues expressing raldh2. We then investigated how RA production might regulate these relationships. Endogenous RA produced by raldhs did not play a role in setting cyp26a1 expression in most tissues. However, exogenous RA regulates expression of both enzymes. cyp26a1 is up regulated in the embryo in a time, concentration, and tissue-dependent manner. Conversely, raldh2 expression is reduced with RA treatment. Tests of the raldh2 promoter in cell transfections proved that RA directly represses its activity. These data demonstrate that the feedback mechanisms regulating production and degradation of RA must be considered in any experiments altering levels of RA in the developing vertebrate embryo.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Tretinoina/metabolismo , Pez Cebra/embriología , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Expresión Génica , Retinal-Deshidrogenasa , Ácido Retinoico 4-Hidroxilasa , Pez Cebra/metabolismo , Proteínas de Pez Cebra
6.
Gene ; 299(1-2): 141-51, 2002 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-12459262

RESUMEN

Estrogenic steroid hormones mediate complex actions important in both embryonic and adult life. The hormones signal through ligand-inducible transcription factors known as estrogen receptors (ERs). In this study, we have isolated a zebrafish estrogen receptor with homology to human estrogen receptor beta (ERbeta). This zebrafish ERbeta (ERbetaa) has a conserved genomic structure of eight coding exons with boundaries similar to those of human ERbeta. The coding exon structures of two other zebrafish estrogen receptors (ERalpha and ERbetab) are presented as well. We also analyzed 3.3 kb of the promoter region and identified numerous putative transcription factor binding sites, including SP1 and ER half sites. Zebrafish ERbetaa message RNA is maternally loaded, but quickly degraded after fertilization, as detected by reverse transcriptase polymerase chain reaction. ERbetaa transcripts are detected again between 24 and 48 h post fertilization. These results indicate that ERbeta has been highly conserved during evolution and is likely used during later embryogenesis in zebrafish. Future identification of the expression levels and patterns of this and other estrogen receptors in zebrafish will allow a better understanding of estrogen signaling during embryogenesis.


Asunto(s)
Embrión no Mamífero/metabolismo , Genes/genética , Receptores de Estrógenos/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , ADN/química , ADN/genética , ADN Complementario/química , ADN Complementario/genética , Desarrollo Embrionario , Receptor beta de Estrógeno , Exones , Femenino , Regulación del Desarrollo de la Expresión Génica , Intrones , Luciferasas/genética , Luciferasas/metabolismo , Datos de Secuencia Molecular , Filogenia , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Secuencia de ADN , Pez Cebra/embriología
7.
Neurotoxicol Teratol ; 26(6): 709-18, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15451034

RESUMEN

At a time when common regulatory pathways are being identified in several different species and genomics is beginning to allow comparisons of genes, how they are arranged on chromosomes and how they are regulated, zebrafish has emerged as a valuable and complementary vertebrate model. Some of the characteristics that prove of value are described and illustrated. Fluorescent transgenic lines of zebrafish embryos are presented for time-line studies with neurotoxicants. While genetic knockout technology has yet to be developed for the model, the anti-sense, morpholino approach allows for knockdown of expression of genes for the 3 day, embryonic period. This can provide for phenocopies of mutant genes for those genes essential to embryonic development or it can provide for a limited inhibition of gene expression that allows subsequent development of the fish. With the zebrafish genomic sequencing effort, microarray technology is now developing for the model system. These resources and technologies allow one to challenge the system with toxicants, and to view the immediate effects of the toxicants with transgenic embryos that fluoresce in part or all of the nervous system. Behavioral and learning protocols have been developed for the organism so that early exposures can be assayed for effects upon adult fish. Microarray technology should allow for one to identify specific genes and pathways affected by a neurotoxicant. In the future, these approaches should provide a working protocol for exploring molecular mechanisms of neurotoxicants. This type of complementary approach should then allow for more efficient examination and testing of mechanisms in mammalian models.


Asunto(s)
Animales Modificados Genéticamente/genética , Modelos Animales de Enfermedad , Embrión no Mamífero/efectos de los fármacos , Neurotoxinas/toxicidad , Pez Cebra/genética , Animales , Animales Modificados Genéticamente/embriología , Elementos sin Sentido (Genética)/genética , Proteínas Bacterianas , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Cloropirifos/toxicidad , Embrión no Mamífero/embriología , Embrión no Mamífero/fisiología , Exposición a Riesgos Ambientales , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Biblioteca Genómica , Proteínas Fluorescentes Verdes , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Proteínas Luminiscentes , Mutación/efectos de los fármacos , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Factores de Tiempo , Pez Cebra/embriología
8.
Neurotoxicol Teratol ; 26(6): 719-23, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15451035

RESUMEN

Chlorpyrifos (CPF), a widely used organophosphate insecticide and potent acetylcholinesterase inhibitor, interferes with neurobehavioral development. Rat models have been key in demonstrating that developmental CPF exposure causes learning deficits and locomotor activity alterations, which persist into adulthood. Complementary nonmammalian models can be useful in determining the neurodevelopmental mechanisms underlying these persisting behavioral effects. Zebrafish (Danio rerio) with their clear chorion and extensive developmental information base provide an excellent model for assessment of molecular processes of toxicant-impacted neurodevelopment. We have developed methods for assessing spatial discrimination learning in adult zebrafish and have documented persisting effects of developmental CPF exposure on swimming activity and learning after low and high doses of CPF (10 and 100 ng/ml) administered to zebrafish embryos on Days 1-5 postfertilization (pf). In the current study, we developed methods for behavioral assessment of CPF exposure on swimming activity in newly hatched zebrafish. An equal area segmented annular grid (concentric circles divided into quadrants through the diameter) was made in a 16-mm diameter cylinder. The test area was placed on a heating device secured to an Olympus SZH10 dissecting scope stage. Zebrafish embryos were exposed to 10 ng/ml CPF, 100 ng/ml CPF, or vehicle control (25 microl/ml DMSO) (n=8-10/treatment group). Each treatment group was kept in a total volume of 25 ml of egg water (60 mg/ml Instant Ocean) including DMSO with or without CPF mixed to above dilutions in an incubator set at 28.5 degrees C. CPF dilutions or vehicle were changed daily with exposure ending on Day 5 pf. Testing of larval zebrafish was performed on Days 6 and 9 pf. The fish were placed in the test cylinder with 1.5 ml of egg H(2)O (28.5 degrees C). After a 2-min acclimation period, the swimming activity of the fish was measured for a 3-min testing session. The 100 ng/ml CPF dose caused significant slowing of swimming activity on Days 6 and 9 pf and had persisting effects of impairing spatial discrimination and decreasing response latency in adulthood. Developmental exposure to 10 ng/ml of CPF did not cause a significant change in locomotor activity during the period soon after hatching. CPF exposure during early development caused clear behavioral impairments detectable during the posthatching period. In a previous study, we found that early developmental CPF exposure caused behavioral alterations in zebrafish, which lasted throughout adulthood. The molecular mechanisms by which early developmental CPF exposure produces these behavioral impairments expressed in adulthood can now be studied in the zebrafish model.


Asunto(s)
Cloropirifos/toxicidad , Embrión no Mamífero/efectos de los fármacos , Larva/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Pez Cebra/fisiología , Animales , Conducta Animal/efectos de los fármacos , Inhibidores de la Colinesterasa/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/embriología , Embrión no Mamífero/fisiología , Habituación Psicofisiológica/efectos de los fármacos , Habituación Psicofisiológica/fisiología , Larva/crecimiento & desarrollo , Actividad Motora/fisiología , Estimulación Física/métodos , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Natación/fisiología , Pez Cebra/embriología
9.
Neurotoxicol Teratol ; 25(1): 51-7, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12633736

RESUMEN

Chlorpyrifos (CPF) is a widely used insecticide, which has been shown to interfere with neurobehavioral development. Rat models have been key in demonstrating that prenatal CPF exposure causes choice accuracy deficits and motor alterations, which persist into adulthood. Complementary nonmammalian models can be useful in determining the molecular mechanisms underlying the persisting behavioral effects of developmental CPF exposure. Zebrafish with their clear chorion and extensive developmental information base provide an excellent model for assessment of molecular processes of toxicant impacted neurodevelopment. To facilitate the use of the zebrafish model and to compare it to the more typical rodent models, the behavioral phenotype of CPF toxicity in zebrafish must be well characterized. Our laboratory has developed methods for assessing spatial discrimination learning in zebrafish, which can differentiate response latency from choice accuracy in a three chambered fish tank. Low and high doses of CPF (10 and 100 ng/ml on days 1-5 postfertilization) both had significant persisting effects on both spatial discrimination and response latency over 18 weeks of testing. The high, but not the low dose, significantly accelerated mortality rates of the fish during the study from 20-38 weeks of age. Developmental exposure to either 10 or 100 ng/ml of CPF caused significant spatial discrimination impairments in zebrafish when they were adults. The impairment caused by 10 ng/ml was seen during early but not later testing, while the impairment caused by 100 ng/ml became more pronounced with continued testing. The higher dose caused a more pervasive impairment. The 10 and 100 ng/ml doses had opposite effects on response latency. The low 10 ng/ml dose significantly slowed response latency, while the high 100 ng/ml dose significant increased response latency. Both of these effects diminished with continued testing. CPF exposure during early development caused clear behavioral impairments, which lasted throughout adulthood in zebrafish. The molecular mechanisms by which early developmental CPF exposure produces these behavioral impairments expressed in adulthood can now be studied in the zebrafish model.


Asunto(s)
Cloropirifos/toxicidad , Embrión no Mamífero/efectos de los fármacos , Insecticidas/toxicidad , Larva/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/fisiopatología , Aprendizaje Discriminativo/efectos de los fármacos , Aprendizaje Discriminativo/fisiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/embriología , Desarrollo Embrionario , Femenino , Humanos , Larva/crecimiento & desarrollo , Embarazo , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Percepción Espacial/efectos de los fármacos , Percepción Espacial/fisiología , Tasa de Supervivencia
10.
PLoS One ; 9(11): e113158, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25402455

RESUMEN

Oxidative stress is an important mechanism of chemical toxicity, contributing to teratogenesis and to cardiovascular and neurodegenerative diseases. Developing animals may be especially sensitive to chemicals causing oxidative stress. The developmental expression and inducibility of anti-oxidant defenses through activation of NF-E2-related factor 2 (NRF2) affect susceptibility to oxidants, but the embryonic response to oxidants is not well understood. To assess the response to chemically mediated oxidative stress and how it may vary during development, zebrafish embryos, eleutheroembryos, or larvae at 1, 2, 3, 4, 5, and 6 days post fertilization (dpf) were exposed to DMSO (0.1%), tert-butylhydroquinone (tBHQ; 10 µM) or 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 2 nM) for 6 hr. Transcript abundance was assessed by real-time qRT-PCR and microarray. qRT-PCR showed strong (4- to 5-fold) induction of gstp1 by tBHQ as early as 1 dpf. tBHQ also induced gclc (2 dpf), but not sod1, nqo1, or cyp1a. TCDD induced cyp1a but none of the other genes. Microarray analysis showed that 1477 probes were significantly different among the DMSO-, tBHQ-, and TCDD-treated eleutheroembryos at 4 dpf. There was substantial overlap between genes induced in developing zebrafish and a set of marker genes induced by oxidative stress in mammals. Genes induced by tBHQ in 4-dpf zebrafish included those involved in glutathione synthesis and utilization, signal transduction, and DNA damage/stress response. The strong induction of hsp70 determined by microarray was confirmed by qRT-PCR and by use of transgenic zebrafish expressing enhanced green fluorescent protein (EGFP) under control of the hsp70 promoter. Genes strongly down-regulated by tBHQ included mitfa, providing a molecular explanation for the loss of pigmentation in tBHQ-exposed embryos. These data show that zebrafish embryos are responsive to oxidative stress as early as 1 dpf, that responsiveness varies with development in a gene-specific manner, and that the oxidative stress response is substantially conserved in vertebrate animals.


Asunto(s)
Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hidroquinonas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Proteínas de Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Antioxidantes/toxicidad , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/patología , Perfilación de la Expresión Génica , Oxidación-Reducción , Teratógenos/toxicidad , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
Neurotoxicol Teratol ; 34(6): 587-91, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23022260

RESUMEN

Zebrafish provide a powerful model of the impacts of embryonic toxicant exposure on neural development that may result in long-term behavioral dysfunction. In this study, zebrafish embryos were treated with 1.5mM strychnine for short embryonic time windows to induce transient changes in inhibitory neural signaling, and were subsequently raised in untreated water until adulthood. PCR analysis showed indications that strychnine exposure altered expression of some genes related to glycinergic, GABAergic and glutamatergic neuronal synapses during embryonic development. In adulthood, treated fish showed significant changes in swimming speed and tank diving behavior compared to controls. Taken together, these data show that a short embryonic exposure to a neurotoxicant can alter development of neural synapses and lead to changes in adult behavior.


Asunto(s)
Conducta Animal/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Estricnina/toxicidad , Sinapsis/efectos de los fármacos , Pez Cebra/embriología , Alternativas al Uso de Animales , Animales , Embrión no Mamífero/metabolismo , Neurotransmisores/metabolismo , Reacción en Cadena de la Polimerasa , Natación , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo , Pruebas de Toxicidad , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
12.
Gene ; 486(1-2): 31-6, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21767619

RESUMEN

The retinoic acid receptors (RARs or rars) and the thyroid hormone receptors are members of the steroid receptor superfamily that interact with their DNA response elements (for RARs: retinoic acid response elements or RAREs) in the regulatory regions of promoters in the absence of their ligand. In this ligand minus configuration, it has been suggested that the RAR provides a binding site for a corepressor (SMRT or N-CoR) that also brings in other proteins to repress the gene. In the presence of the ligand, the receptor goes through an allosteric change eliminating the corepressor binding site and providing a coactivator binding site. In this manuscript we describe the isolation of the zebrafish corepressor, smrt. We show that its association with the zebrafish rar aa is sensitive to retinoic acid and that the corepressor mRNA is present in 8 cell zebrafish embryos - a time at which the embryonic genome is not active. We suggest that this rar-corepressor complex may be part of an embryonic, epigenetic switch that keeps retinoic acid responsive genes off before retinoic becomes available to the embryo.


Asunto(s)
Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Epigénesis Genética , Exones , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Modelos Biológicos , Datos de Secuencia Molecular , Complejos Multiproteicos , Co-Represor 2 de Receptor Nuclear/química , Estructura Terciaria de Proteína , Receptores de Ácido Retinoico/metabolismo , Homología de Secuencia de Aminoácido , Técnicas del Sistema de Dos Híbridos , Pez Cebra/embriología , Proteínas de Pez Cebra/química
13.
Neurotoxicol Teratol ; 33(6): 631-40, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21767634

RESUMEN

This is a review of research that supports a hypothesis regarding early restriction of gene expression in the vertebrate embryo. We hypothesize that vertebrate retinoic acid receptors (RARs for several vertebrates but rars for zebrafish) are part of an embryonic, epigenetic switch whose default position, at the time of fertilization is "OFF". This is due to the assemblage of a rar-corepressor-histone deacetylase complex on retinoic acid response elements (RAREs) in regulatory regions of a subset of genes. In addition, selective and precise allocation of retinoic acid during early development through the interaction of Phase I enzymes throws the switch "ON" in a predictable, developmental manner. We are proposing that this is a basic, early embryonic switch that can cause the initiation of cascades of gene expression that are responsible for at least some early, diversification of cell phenotypes. Dehydrogenases and a subset of cytochrome p450 genes (cyp26a1, cyp26b1, and cyp26c1) play the major role in providing the retinoic acid and limiting its access. We also suggest that this mechanism may be playing a significant role in the repression of genes in undifferentiated stem cells.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Receptores de Ácido Retinoico/genética , Tretinoina/toxicidad , Pez Cebra/embriología , Animales , Embrión no Mamífero/enzimología , Embrión no Mamífero/metabolismo , Epigénesis Genética/efectos de los fármacos , Elementos de Respuesta/efectos de los fármacos , Elementos de Respuesta/genética , Pez Cebra/genética
14.
Neurotoxicol Teratol ; 33(6): 668-73, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21741476

RESUMEN

As more adults take the stimulant medication methylphenidate to treat attention deficit hyperactivity disorder (ADHD) residual type, the risk arises with regard to exposure during early development if people taking the medication become pregnant. We studied the neurobehavioral effects of methylphenidate in zebrafish. Zebrafish offer cellular reporter systems, continuous visual access and molecular interventions such as morpholinos to help determine critical mechanisms underlying neurobehavioral teratogenicity. Previously, we had seen that persisting neurobehavioral impairment in zebrafish with developmental chlorpyrifos exposure was associated with disturbed dopamine systems. Because methylphenidate is an indirect dopamine agonist, it was thought that it might also cause persistent behavioral impairment after developmental exposure. Zebrafish embryos were exposed to the ADHD stimulant medication methylphenidate 0-5 days post fertilization (12.5-50mg/l). They were tested for long-term behavioral effects as adults. Methylphenidate exposure (50mg/l) caused significant increases in dopamine, norepinepherine and serotonin on day 6 but not day 30 after fertilization. In the novel tank diving test of predatory avoidance developmental methylphenidate (50mg/l) caused a significant reduction in the normal diving response. In the three-chamber spatial learning task early developmental methylphenidate (50mg/l) caused a significant impairment in choice accuracy. These data show that early developmental exposure of zebrafish to methylphenidate causes a long-term impairment in neurobehavioral plasticity. The identification of these functional deficits in zebrafish enables further studies with this model to determine how molecular and cellular mechanisms are disturbed to arrive at this compromised state.


Asunto(s)
Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/toxicidad , Embrión no Mamífero/efectos de los fármacos , Metilfenidato/toxicidad , Pez Cebra/embriología , Animales , Monoaminas Biogénicas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Química Encefálica/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Conducta Espacial/efectos de los fármacos , Pez Cebra/crecimiento & desarrollo
15.
Neurotoxicol Teratol ; 33(6): 735-41, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22036888

RESUMEN

Zebrafish are increasingly used for developmental neurotoxicity testing because early embryonic events are easy to visualize, exposures are done without affecting the mother and the rapid development of zebrafish allows for high throughput testing. We used zebrafish to examine how exposures to three different organophosphorus pesticides (chlorpyrifos, diazinon and parathion) over the first five days of embryonic and larval development of zebrafish affected their survival, acetylcholinesterase (AChE) activity and behavior. We show that at non-lethal, equimolar concentrations, chlorpyrifos (CPF) is more effective at equimolar concentrations than diazinon (DZN) and parathion (PA) in producing AChE inhibition. As concentrations of DZN and PA are raised, lethality occurs before they can produce the degree of AChE inhibition observed with CPF at 300 nM. Because of its availability outside the mother at the time of fertilization, zebrafish provides a complementary model for studying the neurotoxicity of very early developmental exposures.


Asunto(s)
Acetilcolinesterasa/metabolismo , Cloropirifos/toxicidad , Inhibidores de la Colinesterasa/toxicidad , Diazinón/toxicidad , Síndromes de Neurotoxicidad/embriología , Paratión/toxicidad , Pez Cebra/embriología , Animales , Conducta Animal/efectos de los fármacos , Cloropirifos/química , Inhibidores de la Colinesterasa/química , Diazinón/química , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/enzimología , Desarrollo Embrionario/efectos de los fármacos , Larva , Síndromes de Neurotoxicidad/enzimología , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/fisiopatología , Paratión/química , Relación Estructura-Actividad
16.
Neurotoxicol Teratol ; 33(6): 742-51, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21745564

RESUMEN

Developmental exposure of rats to the pesticide chlorpyrifos (CPF) causes persistent neurobehavioral impairment. In a parallel series of studies with zebrafish, we have also found persisting behavioral dysfunction after developmental CPF exposure. We have developed a battery of measures of zebrafish behavior, which are reliable and sensitive to toxicant-induced damage. This study determined the critical duration of developmental CPF exposure for causing persisting neurobehavioral effects. Tests of sensorimotor response (tap startle response and habituation), stress response (novel tank diving test) and learning (3-chamber tank spatial discrimination) were conducted with adult zebrafish after early developmental CPF exposure. The CPF exposure level was 100 ng/ml with durations of 0-1, 0-2, 0-3, 0-4 and 0-5 days after fertilization. Developmental CPF exposure had persisting behavioral effects in zebrafish tested as adults. In the tactile startle test, CPF exposed fish showed decreased habituation to startle and a trend toward increased overall startle response. In the novel tank exploration test, exposed fish showed decreased escape diving response and increased swimming activity. In the 3-chamber learning test, the 0-5 day CPF exposure group had a significantly lower learning rate. There was evidence for persisting declines in brain dopamine and norepinepherine levels after developmental CPF exposure. In all of the measures the clearest persistent effects were seen in fish exposed for the full duration of five days after fertilization. In a follow-up experiment there were some indications for persisting behavioral effects after exposure during only the later phase of this developmental window. This study demonstrated the selective long-term neurobehavioral alterations caused by exposure to CPF in zebrafish. The zebrafish model can facilitate the determination of the molecular mechanisms underlying long-term neurobehavioral impairment after developmental toxicant exposure.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cloropirifos/toxicidad , Modelos Animales de Enfermedad , Desarrollo Embrionario/efectos de los fármacos , Síndromes de Neurotoxicidad/embriología , Pez Cebra/embriología , Animales , Química Encefálica/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Exposición a Riesgos Ambientales/efectos adversos , Exposición a Riesgos Ambientales/análisis , Aprendizaje/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Reflejo de Sobresalto/efectos de los fármacos , Factores de Tiempo , Pez Cebra/metabolismo
17.
Neurotoxicol Teratol ; 32(1): 99-108, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19268529

RESUMEN

Chlorpyrifos (CPF) an organophosphate pesticide causes persisting behavioral dysfunction in rat models when exposure is during early development. In earlier work zebrafish were used as a complementary model to study mechanisms of CPF-induced neurotoxicity induced during early development. We found that developmental (first five days after fertilization) chlorpyrifos exposure significantly impaired learning in zebrafish. However, this testing was time and labor intensive. In the current study we tested the hypothesis that persisting effects of developmental chlorpyrifos could be detected with a brief automated assessment of startle response and that this behavioral index could be used to help determine the neurobehavioral mechanisms for persisting CPF effects. The swimming activity of adult zebrafish was assessed by a computerized video-tracking device after a sudden tap to the test arena. Ten consecutive trials (1/min) were run to determine startle response and its habituation. Additionally, habituation recovery trials were run at 8, 32 and 128 min after the end of the initial trial set. CPF-exposed fish showed a significantly (p<0.025) greater overall startle response during the 10-trial session compared to controls (group sizes: Control N=40, CPF N=24). During the initial recovery period (8 min) CPF-exposed fish showed a significantly (p<0.01) greater startle response compared to controls. To elucidate the contributions of nicotinic and muscarinic acetylcholine receptors to developmental CPF-mediated effects, the effects of developmental nicotine and pilocarpine exposure throughout the first five days after fertilization were determined. Developmental nicotine and pilocarpine exposure significantly increased startle response, though nicotine (group sizes: Control N=32, 15 mM N=12, 25 mM N=20) was much more potent than pilocarpine (group sizes: Control N=20, 100 microM N=16, 1000 microM N=12). Neither was as potent as CPF for developmental exposure increasing startle response in adulthood. Lastly, developmental CPF exposure decreased dopamine and serotonin levels and increased transmitter turnover in developing zebrafish larvae (N=4 batches of 50 embryos/treatment). Only the decline in dopamine concentrations persisted into adulthood (group sizes: Control N=14, CPF N=13). This study shows that a quick automated test of startle can detect persisting neurobehavioral impairments caused by developmental exposure to CPF. This may be helpful in screening for persisting neurobehavioral defects from a variety of toxicants.


Asunto(s)
Cloropirifos/toxicidad , Larva/efectos de los fármacos , Modelos Animales , Agonistas Muscarínicos/toxicidad , Nicotina/toxicidad , Pilocarpina/toxicidad , Reflejo de Sobresalto/efectos de los fármacos , Pez Cebra/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Femenino , Habituación Psicofisiológica/efectos de los fármacos , Insecticidas/toxicidad , Larva/metabolismo , Exposición Materna , Agonistas Nicotínicos/toxicidad , Serotonina/metabolismo , Factores de Tiempo , Pruebas de Toxicidad/métodos
18.
PLoS One ; 5(5): e10465, 2010 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-20463971

RESUMEN

BACKGROUND: In order to generate hypotheses regarding the mechanisms by which 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin) causes toxicity, we analyzed global gene expression changes in developing zebrafish embryos exposed to this potent toxicant in the context of a dynamic gene network. For this purpose, we also computationally inferred a zebrafish (Danio rerio) interactome based on orthologs and interaction data from other eukaryotes. METHODOLOGY/PRINCIPAL FINDINGS: Using novel computational tools to analyze this interactome, we distinguished between dioxin-dependent and dioxin-independent interactions between proteins, and tracked the temporal propagation of dioxin-dependent transcriptional changes from a few genes that were altered initially, to large groups of biologically coherent genes at later times. The most notable processes altered at later developmental stages were calcium and iron metabolism, embryonic morphogenesis including neuronal and retinal development, a variety of mitochondria-related functions, and generalized stress response (not including induction of antioxidant genes). Within the interactome, many of these responses were connected to cytochrome P4501A (cyp1a) as well as other genes that were dioxin-regulated one day after exposure. This suggests that cyp1a may play a key role initiating the toxic dysregulation of those processes, rather than serving simply as a passive marker of dioxin exposure, as suggested by earlier research. CONCLUSIONS/SIGNIFICANCE: Thus, a powerful microarray experiment coupled with a flexible interactome and multi-pronged interactome tools (which are now made publicly available for microarray analysis and related work) suggest the hypothesis that dioxin, best known in fish as a potent cardioteratogen, has many other targets. Many of these types of toxicity have been observed in mammalian species and are potentially caused by alterations to cyp1a.


Asunto(s)
Dioxinas/toxicidad , Transcripción Genética/efectos de los fármacos , Pez Cebra/genética , Animales , Bases de Datos Genéticas , Eucariontes/efectos de los fármacos , Eucariontes/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/genética , Glucólisis/efectos de los fármacos , Glucólisis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Teratógenos/toxicidad
19.
Neurotoxicol Teratol ; 32(3): 391-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20116428

RESUMEN

The increased use of silver nanoparticles in consumer and medical products has led to elevated human and environmental exposures. Silver nanoparticles act as antibacterial/antifungal agents by releasing Ag(+) and recent studies show that Ag(+) impairs neural cell replication and differentiation in culture, suggesting that in vivo exposures could compromise neurodevelopment. To determine whether Ag(+) impairs development in vivo, we examined the effects of exposure on survival, morphological, and behavioral parameters in zebrafish embryos and larvae. We exposed zebrafish from 0 to 5days post-fertilization to concentrations of Ag(+) ranging from 10nM to 100microM in order to assess effects on survival and early embryonic development. We then tested whether concentrations below the threshold for dysmorphology altered larval behavior and subsequent survival. Ag(+) concentrations >or=3microM significantly reduced embryonic survival, whereas 1microM delayed hatching with no effect on survival. Reducing the concentration to as low as 0.1microM delayed the inflation of the swim bladder without causing gross dysmorphology or affecting hatching. At this concentration, swimming activity was impaired, an effect that persisted past the point where swim bladder inflation became normal; in contrast, general motor function was unaffected. The early behavioral impairment was then predictive of subsequent decreases in survival. Ag(+) is a developmental toxicant at concentrations only slightly above allowable levels. At low concentrations, Ag(+) acts as a neurobehavioral toxicant even in the absence of dysmorphology.


Asunto(s)
Conducta Animal/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Nanopartículas del Metal/toxicidad , Plata/toxicidad , Pez Cebra/embriología , Animales , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/efectos de los fármacos , Larva , Análisis de Supervivencia , Pez Cebra/crecimiento & desarrollo
20.
Neurotoxicology ; 31(5): 582-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20359493

RESUMEN

The understanding of how environmental exposures interact with genetics in central nervous system dysfunction has gained great momentum in the last decade. Seminal findings have been uncovered in both mammalian and non-mammalian model in large result of the extraordinary conservation of both genetic elements and differentiation processes between mammals and non-mammalians. Emerging model organisms, such as the nematode and zebrafish have made it possible to assess the effects of small molecules rapidly, inexpensively, and on a miniaturized scale. By combining the scale and throughput of in vitro screens with the physiological complexity and traditional animal studies, these models are providing relevant information on molecular events in the etiology of neurodegenerative disorders. The utility of these models is largely driven by the functional conservation seen between them and higher organisms, including humans so that knowledge obtained using non-mammalian model systems can often provide a better understanding of equivalent processes, pathways, and mechanisms in man. Understanding the molecular events that trigger neurodegeneration has also greatly relied upon the use of tissue culture models. The purpose of this summary is to provide-state-of-the-art review of recent developments of non-mammalian experimental models and their utility in addressing issues pertinent to neurotoxicity (Caenorhabditis elegans and Danio rerio). The synopses by Aschner and Levin summarize how genetic mutants of these species can be used to complement the understanding of molecular and cellular mechanisms associated with neurobehavioral toxicity and neurodegeneration. Next, studies by Suñol and Olopade detail the predictive value of cultures in assessing neurotoxicity. Suñol and colleagues summarize present novel information strategies based on in vitro toxicity assays that are predictive of cellular effects that can be extrapolated to effects on individuals. Olopade and colleagues describe cellular changes caused by sodium metavanadate (SMV) and demonstrate how rat primary astrocyte cultures can be used as predicitive tools to assess the neuroprotective effects of antidotes on vanadium-induced astrogliosis and demyelination.


Asunto(s)
Exposición a Riesgos Ambientales , Degeneración Nerviosa/etiología , Degeneración Nerviosa/genética , Síndromes de Neurotoxicidad , Neurotoxinas/toxicidad , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Síndromes de Neurotoxicidad/complicaciones , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/genética , Literatura de Revisión como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA