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










Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
Aquat Toxicol ; 164: 52-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25914093

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD or dioxin) disrupts craniofacial development in zebrafish larvae. However, the cellular changes responsible for the decreased jaw size remain poorly understood. We show that smaller jaw size is due to a decrease in both the size and number of chondrocytes in the developing craniofacial cartilages. TCDD was found to decrease ossification of osteoblasts in the perichondrium of craniofacial cartilages. We also discovered that TCDD caused clefting of the parasphenoid, an effect with similarity to TCDD-induced cleft palate in mice. Thus, dermal and perichondrial bone development of the craniofacial skeleton are clearly disrupted by TCDD exposure in the zebrafish larvae. This dysmorphic response of the zebrafish craniofacial skeleton after exposure to TCDD is consistent with findings demonstrating disruption of axial bone development in medaka and repression of sox9b in zebrafish.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Cartílago/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Huesos Faciales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Maxilares/efectos de los fármacos , Ratones , Receptores de Hidrocarburo de Aril/genética , Factor de Transcripción SOX9/genética , Contaminantes Químicos del Agua/toxicidad , Proteínas de Pez Cebra/genética
5.
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
6.
Mol Cell Endocrinol ; 398(1-2): 36-41, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25194296

RESUMEN

Dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin; TCDD) is an aryl hydrocarbon receptor (AHR) agonist, an endocrine disruptor, and a potent global pollutant. TCDD exposure is associated with diseases of almost every organ system, and its toxicity is highly conserved across vertebrates. While the acute developmental effects of dioxin exposure have been extensively studied, the ability of early sublethal exposure to produce toxicity in adulthood or subsequent generations is poorly understood. This type of question is difficult to study because of the time frame of the effects. With human subjects, such a study could span more than a lifetime. We have chosen zebrafish (Danio rerio) as a model because they are vertebrates with short generation times and consistent genetic backgrounds. Zebrafish have very modest housing needs, facilitating single and multigenerational studies with minimal time and expense. We have used this model to identify transgenerational effects of TCDD on skeletal development, sex ratio, and male-mediated decreases in reproductive capacity. Here we compare these findings with transgenerational effects described in laboratory rodent species. We propose that the zebrafish is a cost-effective model system for evaluating the transgenerational effects of toxic chemicals and their role in the fetal basis of adult disease.


Asunto(s)
Dioxinas/toxicidad , Disruptores Endocrinos/toxicidad , Predisposición Genética a la Enfermedad , Efectos Tardíos de la Exposición Prenatal , Pez Cebra/genética , Animales , Exposición a Riesgos Ambientales , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Mutágenos/toxicidad , Embarazo , Receptores de Hidrocarburo de Aril/agonistas , Reproducción/genética
7.
Toxicol Sci ; 141(1): 141-54, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25037585

RESUMEN

Exposure of zebrafish embryos to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) activates the zebrafish aryl hydrocarbon receptor 2 (AHR) to produce developmental and cardiovascular toxicity. AHR is found in the heart; however, AHR activation by TCDD is not confined to the heart and occurs throughout the organism. In order to understand the cause of cardiotoxicity, we constructed a constitutively active AHR (caAHR) based on the zebrafish AHR2 and expressed it specifically in cardiomyocytes. We show that AHR activation within the cardiomyocytes can account for the heart failure induced by TCDD. Expression of the caAHR within the heart produced cardiac malformations, loss of circulation, and pericardial edema. The heart-specific activation of AHR reproduced several other well-characterized endpoints of TCDD toxicity outside of the cardiovascular system, including defects in swim bladder and craniofacial development. This work identifies a single cellular site of TCDD action, the myocardial cell, that can account for the severe cardiovascular collapse observed following early life stage exposure to TCDD, and contributes to other forms of toxicity.


Asunto(s)
Miocitos Cardíacos/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra , Animales , Cardiotoxicidad , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/metabolismo , Miocitos Cardíacos/metabolismo , Plásmidos , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/genética , Flujo Sanguíneo Regional/efectos de los fármacos , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
8.
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
9.
Toxicol Sci ; 138(2): 403-11, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24470537

RESUMEN

2,3,7,8 Tetrachlorodibenzo-p-dioxin (TCDD) has been associated with many disease states in humans. A rising concern is that exposure early in life can lead to adult toxicity and toxicity in subsequent generations. Juvenile zebrafish exposed to TCDD (50 pg/ml in water; 1 h exposure) at 3 and 7 weeks post fertilization showed toxicity only later in adulthood. We have maintained the offspring of these exposed F0 fish to determine whether we could find adverse affects in the next two generations of F1 and F2 offspring. TCDD exposure produced a significantly higher female:male ratio in all three generations. Scoliosis-like axial skeleton abnormalities, not normally observed in controls, were present in the F1 and F2 generations descended from the treated F0 founders. Egg release and fertilization success were reduced in the TCDD lineage F1 and F2 generations. This reduction in fertility in the TCDD lineage F2 generation could be attributed to alterations in the F2 males. Using zebrafish as a model allowed the simultaneous maintenance of different generations with relatively small space and costs. The zebrafish showed clear signs of transgenerational responses persisting into generations never directly exposed to TCDD.


Asunto(s)
Modelos Animales de Enfermedad , Disruptores Endocrinos/toxicidad , Anomalías Musculoesqueléticas/inducido químicamente , Dibenzodioxinas Policloradas/toxicidad , Reproducción/efectos de los fármacos , Pez Cebra/crecimiento & desarrollo , Animales , Femenino , Masculino , Anomalías Musculoesqueléticas/embriología , Ovario/efectos de los fármacos , Ovario/embriología , Ovario/crecimiento & desarrollo , Razón de Masculinidad , Pez Cebra/anomalías , Pez Cebra/embriología
10.
Artículo en Inglés | MEDLINE | ID: mdl-26180821

RESUMEN

There is strong evidence indicating that disease in adult humans stems from a combination of genetic and environmental factors. A problem in identifying environmental factors is that subacute exposures during early life are often unnoticed, or exposures are variable among a diverse population. This leads to a confusing pattern in adulthood. An additional problem in following exposure effects in humans is the length of time needed to study outcomes spanning a human generation. We have recently developed a zebrafish model for studying the effects of sublethal juvenile exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin). Although the initial exposure produces no effect at the time, we find skeletal and reproductive defects in adulthood and into subsequent generations. The short generation time of zebrafish along with the ability to maintain large cohorts of exposed individuals and their offspring allows us to overcome variation in exposure and genetic background. Here we describe progress in studying TCDD as an endocrine and developmental disruptor, and our results showing adult consequences of early exposure.

11.
Int J Dev Biol ; 58(9): 693-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25896205

RESUMEN

The transcription factor SOX9 is a member of the SRY-related high-mobility-group box (SOX) superfamily of genes. In mammals, Sox9 plays important roles in many developmental processes including craniofacial, skeletal and heart morphogenesis, retinal and brain development, and gonad differentiation. Human mutations in SOX9 or the SOX9 promoter result in campomelic dysplasia, a severe genetic disorder, which disrupts skeletal, craniofacial, cardiac, neural and reproductive development. Due to the duplication of the teleost fish genome, zebrafish (Danio rerio) have two Sox9 genes: sox9a and sox9b. Loss of sox9b in zebrafish results in loss of function phenotypes that are similar to those observed in humans and mice. In order to generate a transgenic sox9b:EGFP reporter line, we cloned a 2450 bp fragment of the sox9b promoter and fused it to an EGFP reporter. Consistent with reported sox9b expression and function, we observed sox9b:EGFP in the developing heart, skeletal and craniofacial structures, brain, retina, and ovaries. Our resulting transgenic line is a useful tool for identifying and studying sox9b function in development and visualizing a number of zebrafish organs and tissues in which sox9b is normally expressed.


Asunto(s)
Animales Modificados Genéticamente/crecimiento & desarrollo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Factor de Transcripción SOX9/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Corazón/embriología , Corazón/fisiología , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Ratones , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Ovario/embriología , Ovario/metabolismo , Retina/embriología , Retina/metabolismo , Factor de Transcripción SOX9/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
12.
Toxicol Sci ; 135(1): 241-50, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23811824

RESUMEN

The acute effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure have been well documented in many vertebrate species. However, less is known about the consequences in adulthood from sublethal exposure during development. To address this, we exposed zebrafish to sublethal levels of TCDD (1h; 50 pg/ml), either in early embryogenesis (day 0) or during sexual determination (3 and 7 weeks), and assessed the effects later in adulthood. We found that exposure during embryogenesis produced few effects on the adults themselves but did affect the offspring of these fish: Malformations and increased mortality were observed in the subsequent generation. Zebrafish exposed during sexual development showed defects in the cranial and axial skeleton as adults. This was most clearly manifested as scoliosis caused by malformation of individual vertebrae. These fish also showed defects in reproduction, producing fewer eggs with lower fertilization success. Both males and females were affected, with males contributing to the decrease in egg release from the females and exposed females contributing to fertilization failure. TCDD exposure at 3 and 7 weeks produced feminization of the population. Surprisingly, part of this was due to the appearance of fish with clearly female bodies, yet carrying testes in place of ovaries. Our results show that exposures that produce little if any impact during development can cause severe consequences during adulthood and present a model for studying this process.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Desarrollo Sexual/efectos de los fármacos , Anomalías Inducidas por Medicamentos , Animales , Huesos/anomalías , Relación Dosis-Respuesta a Droga , Femenino , Fertilidad/efectos de los fármacos , Masculino , Ovario/anomalías , Ovario/efectos de los fármacos , Razón de Masculinidad , Pez Cebra
13.
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
14.
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
15.
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
16.
Methods Mol Biol ; 1005: 119-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23606253

RESUMEN

Two-dimensional HPLC coupled with tandem MS (MS/MS) has become a mainstream technique in the shotgun proteomics for large-scale identification of proteins from biological samples. This powerful technology provides speed, sensitivity, and dynamic range which are essential to probe complex peptide mixtures from proteomic samples. Herein we present a pH gradient SCX-RP 2D HPLC-MS/MS method designed to improve the peptide resolution and protein identification from complex proteomic samples. The comparison between the pH gradient SCX-RP 2D HPLC method and traditional salt gradient SCX-RP method was presented. A two-step sample prefractionation method utilizing microwave-assisted tryptic digestion to improve the identification of insoluble proteins was also introduced. This novel 2D HPLC-MS/MS method was applied to the heart proteomic sample of the zebrafish, Danio rerio, to provide comprehensive cardiac proteomic profiling of this important model organism for cardiovascular and environmental toxicology studies.


Asunto(s)
Miocardio/metabolismo , Mapeo Peptídico/métodos , Proteoma/metabolismo , Animales , Cromatografía por Intercambio Iónico/métodos , Cromatografía de Fase Inversa/métodos , Expresión Génica , Microondas , Miocardio/química , Proteolisis , Proteoma/genética , Fuerza Protón-Motriz , Espectrometría de Masas en Tándem/métodos , Tripsina/metabolismo , Pez Cebra
17.
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
18.
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
19.
Toxicol Sci ; 131(2): 558-67, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23135548

RESUMEN

Embryonic exposure to the environmental contaminant and aryl hydrocarbon receptor agonist, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin), disrupts cardiac development and function in fish, birds, and mammals. In zebrafish, the temporal window of sensitivity to the cardiotoxic effects of TCDD coincides with epicardium formation. We hypothesized that this TCDD-induced heart failure results from disruption of epicardial development. To determine whether embryonic TCDD exposure inhibits epicardium and proepicardium (PE) development in zebrafish, we used histology and fluorescence immunocytochemistry to examine the epicardium formation in fish exposed to TCDD. TCDD exposure prevented epicardium formation. Using live imaging and in situ hybridization, we found that TCDD exposure blocked the formation of the PE cluster. In situ hybridization experiments showed that TCDD exposure also prevented the expression of the PE marker tcf21 at the site where the PE normally forms. TCDD also inhibited expansion of the epicardial layer across the developing heart: Exposure after PE formation was completed prevented further expansion of the epicardium. However, TCDD exposure did not affect epicardial cells already present. Because TCDD blocks epicardium formation, but is not directly toxic to the epicardium once complete, we propose that inhibition of epicardium formation can account for the window of sensitivity to TCDD cardiotoxicity in developing zebrafish. Epicardium development is crucial to heart development. Loss of this layer during development may account for most if not all of the TCDD-induced cardiotoxicity in zebrafish.


Asunto(s)
Pericardio/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Animales , Pericardio/embriología , Pez Cebra/embriología
20.
Toxicol Sci ; 132(1): 211-21, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23204111

RESUMEN

Normal adult zebrafish can completely regenerate lost myocardium following partial amputation of the ventricle apex. We report that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) significantly impairs this regeneration. Adult male zebrafish were injected with vehicle (control) or TCDD (70ng/g, ip) 1 day prior to partial amputation of the ventricle apex. Gross observation and histological analysis of the amputated heart at 21 days postamputation revealed that TCDD-exposed fish had not progressed beyond the initial clot formation stage, whereas the vehicle control fish showed substantial recovery and almost complete resolution of the formed clot. In contrast, hearts that were not surgically wounded showed no signs of TCDD toxicity. Striking features in the TCDD-exposed hearts were the absence of the normal sheath of new tissue enveloping the wound and the absence of intense cell proliferation at the site of the wound. In addition, the patterns of collagen deposition at the wound site were different between the TCDD and vehicle groups. Because the receptor for TCDD is the aryl hydrocarbon receptor ligand-activated transcriptional regulator, we examined the effects of TCDD exposure on gene expression in the ventricle using DNA microarrays. Samples were collected just prior to amputation and at 6h and 7 days postamputation. TCDD-pretreated hearts had dysregulated expression of genes involved in heart function, tissue regeneration, cell growth, and extracellular matrix. Because embryonic, but not adult, hearts are major targets for TCDD-induced cardiotoxicity, we speculate that the need for embryonic-like cells in regeneration is connected with the effects of TCDD in inhibiting the response to wounding.


Asunto(s)
Corazón/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Regeneración/efectos de los fármacos , Aldehído Oxidorreductasas/genética , Animales , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Corazón/fisiología , Hibridación in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Regulación hacia Arriba , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...