Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 284(2): 142-51, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25732252

ABSTRACT

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.


Subject(s)
DNA Methylation/drug effects , Gene Expression Regulation, Developmental/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , DNA/genetics , Down-Regulation/drug effects , Embryo, Nonmammalian/drug effects , Methyltransferases/genetics , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/genetics , Response Elements , Up-Regulation/drug effects , Zebrafish , Zebrafish Proteins/genetics
2.
PLoS One ; 9(11): e113158, 2014.
Article in English | MEDLINE | ID: mdl-25402455

ABSTRACT

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.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Hydroquinones/toxicity , Oxidative Stress/drug effects , Polychlorinated Dibenzodioxins/toxicity , Zebrafish Proteins/genetics , Zebrafish/growth & development , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Antioxidants/toxicity , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Gene Expression Profiling , Oxidation-Reduction , Teratogens/toxicity , Zebrafish/genetics , Zebrafish Proteins/metabolism
3.
Neurotoxicol Teratol ; 34(6): 587-91, 2012.
Article in English | MEDLINE | ID: mdl-23022260

ABSTRACT

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.


Subject(s)
Behavior, Animal/drug effects , Embryo, Nonmammalian/drug effects , Environmental Pollutants/toxicity , Strychnine/toxicity , Synapses/drug effects , Zebrafish/embryology , Animal Use Alternatives , Animals , Embryo, Nonmammalian/metabolism , Neurotransmitter Agents/metabolism , Polymerase Chain Reaction , Swimming , Synaptic Transmission/drug effects , Time Factors , Toxicity Tests , Zebrafish/growth & development , Zebrafish/metabolism
4.
Neurotoxicol Teratol ; 33(6): 735-41, 2011.
Article in English | MEDLINE | ID: mdl-22036888

ABSTRACT

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.


Subject(s)
Acetylcholinesterase/metabolism , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Diazinon/toxicity , Neurotoxicity Syndromes/embryology , Parathion/toxicity , Zebrafish/embryology , Animals , Behavior, Animal/drug effects , Chlorpyrifos/chemistry , Cholinesterase Inhibitors/chemistry , Diazinon/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Embryonic Development/drug effects , Larva , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/physiopathology , Parathion/chemistry , Structure-Activity Relationship
5.
Neurotoxicol Teratol ; 33(6): 668-73, 2011.
Article in English | MEDLINE | ID: mdl-21741476

ABSTRACT

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.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/toxicity , Embryo, Nonmammalian/drug effects , Methylphenidate/toxicity , Zebrafish/embryology , Animals , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/embryology , Brain Chemistry/drug effects , Choice Behavior/drug effects , Dose-Response Relationship, Drug , Learning/drug effects , Memory/drug effects , Reflex, Startle/drug effects , Spatial Behavior/drug effects , Zebrafish/growth & development
6.
Neurotoxicol Teratol ; 33(6): 742-51, 2011.
Article in English | MEDLINE | ID: mdl-21745564

ABSTRACT

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.


Subject(s)
Behavior, Animal/drug effects , Chlorpyrifos/toxicity , Disease Models, Animal , Embryonic Development/drug effects , Neurotoxicity Syndromes/embryology , Zebrafish/embryology , Animals , Brain Chemistry/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Learning/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Reflex, Startle/drug effects , Time Factors , Zebrafish/metabolism
7.
Gene ; 486(1-2): 31-6, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21767619

ABSTRACT

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.


Subject(s)
Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Amino Acid Sequence , Animals , Epigenesis, Genetic , Exons , Gene Expression Regulation, Developmental , In Situ Hybridization , Models, Biological , Molecular Sequence Data , Multiprotein Complexes , Nuclear Receptor Co-Repressor 2/chemistry , Protein Structure, Tertiary , Receptors, Retinoic Acid/metabolism , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , Zebrafish/embryology , Zebrafish Proteins/chemistry
8.
Neurotoxicol Teratol ; 33(6): 631-40, 2011.
Article in English | MEDLINE | ID: mdl-21767634

ABSTRACT

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.


Subject(s)
Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Receptors, Retinoic Acid/genetics , Tretinoin/toxicity , Zebrafish/embryology , Animals , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Epigenesis, Genetic/drug effects , Response Elements/drug effects , Response Elements/genetics , Zebrafish/genetics
9.
PLoS One ; 5(5): e10465, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20463971

ABSTRACT

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.


Subject(s)
Dioxins/toxicity , Transcription, Genetic/drug effects , Zebrafish/genetics , Animals , Databases, Genetic , Eukaryota/drug effects , Eukaryota/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks/genetics , Glycolysis/drug effects , Glycolysis/genetics , Oligonucleotide Array Sequence Analysis , Teratogens/toxicity
10.
Neurotoxicology ; 31(5): 582-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20359493

ABSTRACT

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.


Subject(s)
Environmental Exposure , Nerve Degeneration/etiology , Nerve Degeneration/genetics , Neurotoxicity Syndromes , Neurotoxins/toxicity , Animals , Disease Models, Animal , Gene Expression Regulation , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/genetics , Review Literature as Topic
11.
Neurotoxicol Teratol ; 32(3): 391-7, 2010.
Article in English | MEDLINE | ID: mdl-20116428

ABSTRACT

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.


Subject(s)
Behavior, Animal/drug effects , Embryonic Development/drug effects , Environmental Pollutants/toxicity , Metal Nanoparticles/toxicity , Silver/toxicity , Zebrafish/embryology , Animals , Dose-Response Relationship, Drug , Embryo, Nonmammalian/drug effects , Larva , Survival Analysis , Zebrafish/growth & development
12.
Neurotoxicol Teratol ; 32(1): 99-108, 2010.
Article in English | MEDLINE | ID: mdl-19268529

ABSTRACT

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.


Subject(s)
Chlorpyrifos/toxicity , Larva/drug effects , Models, Animal , Muscarinic Agonists/toxicity , Nicotine/toxicity , Pilocarpine/toxicity , Reflex, Startle/drug effects , Zebrafish/metabolism , Animals , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Female , Habituation, Psychophysiologic/drug effects , Insecticides/toxicity , Larva/metabolism , Maternal Exposure , Nicotinic Agonists/toxicity , Serotonin/metabolism , Time Factors , Toxicity Tests/methods
13.
Neurotoxicology ; 30(5): 741-53, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19647018

ABSTRACT

Considerable progress has been made over the past couple of decades concerning the molecular bases of neurobehavioral function and dysfunction. The field of neurobehavioral genetics is becoming mature. Genetic factors contributing to neurologic diseases such as Alzheimer's disease have been found and evidence for genetic factors contributing to other diseases such as schizophrenia and autism are likely. This genetic approach can also benefit the field of behavioral neurotoxicology. It is clear that there is substantial heterogeneity of response with behavioral impairments resulting from neurotoxicants. Many factors contribute to differential sensitivity, but it is likely that genetic variability plays a prominent role. Important discoveries concerning genetics and behavioral neurotoxicity are being made on a broad front from work with invertebrate and piscine mutant models to classic mouse knockout models and human epidemiologic studies of polymorphisms. Discovering genetic factors of susceptibility to neurobehavioral toxicity not only helps identify those at special risk, it also advances our understanding of the mechanisms by which toxicants impair neurobehavioral function in the larger population. This symposium organized by Edward Levin and Annette Kirshner, brought together researchers from the laboratories of Michael Aschner, Douglas Ruden, Ulrike Heberlein, Edward Levin and Kathleen Welsh-Bohmer conducting studies with Caenorhabditis elegans, Drosophila, fish, rodents and humans studies to determine the role of genetic factors in susceptibility to behavioral impairment from neurotoxic exposure.


Subject(s)
Behavior/drug effects , Genetics , Neurotoxicity Syndromes , Neurotoxins/toxicity , Toxicology , Animals , Disease Models, Animal , Humans , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Phylogeny
14.
Toxicol Sci ; 109(2): 217-27, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19233942

ABSTRACT

Early piscine life stages are sensitive to polycyclic aromatic hydrocarbon (PAH) exposure, which can cause pericardial effusion and craniofacial malformations. We previously reported that certain combinations of PAHs cause synergistic developmental toxicity, as observed with coexposure to the aryl hydrocarbon receptor agonist beta-naphthoflavone (BNF) and cytochrome P4501A inhibitor alpha-naphthoflavone (ANF). Herein, we hypothesized that oxidative stress is a component of this toxicity. We examined induction of antioxidant genes in zebrafish embryos (Danio rerio) exposed to BNF or ANF individually, a BNF + ANF combination, and a prooxidant positive control, tert-butylhydroperoxide (tBOOH). We measured total glutathione (GSH) and attempted to modulate deformities using the GSH synthesis inhibitor L-buthionine (S,R)-sulfoximine (BSO) and increase GSH pools with N-acetyl cysteine (NAC). In addition, we used a morpholino to knockdown expression of the antioxidant response element transcription factor NRF2 to determine if this would alter gene expression or increase deformity severity. BNF + ANF coexposure significantly increased expressions of superoxide dismutase 1 and 2, glutathione peroxidase 1, pi class glutathione-s-transferase, and glutamate cysteine-ligase to a greater extent than tBOOH, BNF, or ANF alone. BSO pretreatment decreased some GSH levels, but did not worsen deformities, nor did NAC diminish toxicity. Knockdown of NRF2 increased mortality following tBOOH challenge, prevented significant upregulation of antioxidant genes following both tBOOH and BNF + ANF exposures, and exacerbated BNF + ANF-related deformities. Collectively, these findings demonstrate that antioxidant responses are a component of PAH synergistic developmental toxicity and that NRF2 is protective against prooxidant and PAH challenges during development.


Subject(s)
Antioxidants/metabolism , Embryo, Nonmammalian/drug effects , Gene Expression Regulation/drug effects , NF-E2-Related Factor 2/metabolism , Polycyclic Aromatic Hydrocarbons/toxicity , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Benzoflavones/toxicity , Buthionine Sulfoximine/toxicity , Drug Synergism , Embryo, Nonmammalian/abnormalities , Embryonic Development/drug effects , Enzyme Inhibitors/toxicity , Gene Knockdown Techniques , Glutathione/analysis , Oxidation-Reduction , Oxidative Stress , Pericardial Effusion , Up-Regulation , beta-Naphthoflavone/toxicity , tert-Butylhydroperoxide/pharmacology
15.
Dev Dyn ; 237(12): 3798-808, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19035346

ABSTRACT

Cyp26A1 is a major enzyme that controls retinoic acid (RA) homeostasis by metabolizing RA into bio-inactive metabolites. Previous research revealed that the mouse Cyp26A1 promoter has two canonical RA response elements (RAREs) that underlie the regulation of the gene by RA. Analyzing the 2,533-base pairs (2.5 k) genomic sequence upstream of zebrafish cyp26a1 start codon, we report that the two RAREs are conserved in zebrafish cyp26a1 promoter. Mutagenesis demonstrated that the two RAREs work synergistically in RA inducibility of cyp26a1. Fusing the 2.5 k (kilobase pairs) fragment to the enhanced yellow fluorescent protein (eYFP) reporter gene, we have generated two transgenic lines of zebrafish [Tg(cyp26a1:eYFP)]. The transgenic zebrafish display expression patterns similar to that of cyp26a1 gene in vivo. Consistent with the in vitro results, the reporter activity is RA inducible in embryos. Taken together, our results demonstrate that the 2.5 k fragment underlies the regulation of the zebrafish cyp26a1 gene by RA.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Promoter Regions, Genetic/genetics , Tretinoin/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Cell Line , Gene Expression Regulation, Developmental/genetics , Genes, Reporter/genetics , Humans , Molecular Mimicry , Molecular Sequence Data , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Retinoic Acid 4-Hydroxylase , Zebrafish/genetics , Zebrafish Proteins
16.
Zebrafish ; 4(1): 49-57, 2007.
Article in English | MEDLINE | ID: mdl-18041942

ABSTRACT

Estradiol is produced from testosterone by the aromatase gene, cyp19. In the zebrafish Danio rerio, brain aromatase, cyp19a1b, is highly expressed during development. We report the developmental expression pattern of cyp19a1b using whole mount in situ hybridization and describe hormonal effects on the gene using RT-PCR. Expression is up-regulated between 24 and 48 hours postfertilization (hpf). Localized expression of cyp19a1b is first detected at 48 hpf in the preoptic area, hypothalamus, terminal nerve, and olfactory bulb. The gene is itself induced by estradiol in a positive feedback loop. Testosterone exposure also induces the cyp19a1b gene in zebrafish; however, a majority of this induction is blocked by an estrogen receptor antagonist. The expression pattern of aromatase in the brain and its control by steroid hormones is well conserved among the vertebrate lineage.


Subject(s)
Aromatase/biosynthesis , Brain/enzymology , Embryo, Nonmammalian/enzymology , Gene Expression Regulation, Developmental/physiology , Steroids/physiology , Zebrafish Proteins/biosynthesis , Zebrafish/genetics , Animals , Aromatase/genetics , Brain/embryology , Dihydrotestosterone/pharmacology , Embryo, Nonmammalian/embryology , Enzyme Induction/physiology , Estradiol/physiology , Gene Expression Regulation, Enzymologic , Green Fluorescent Proteins/metabolism , In Situ Hybridization , Luminescent Agents/metabolism , Molecular Sequence Data , RNA, Messenger/metabolism , Receptors, Estrogen/physiology , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/physiology , Time Factors , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
17.
BMC Biotechnol ; 7: 40, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17623073

ABSTRACT

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.


Subject(s)
Heart/physiology , Microscopy, Confocal/methods , Zebrafish/physiology , Aminobenzoates/pharmacology , Animals , Aorta/drug effects , Aorta/embryology , Aorta/physiology , Blood Flow Velocity/drug effects , Blotting, Western , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Genetic Complementation Test , Heart/embryology , Larva/cytology , Larva/genetics , Larva/physiology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Microinjections , Morpholines/pharmacology , Mutation , Myocardium/cytology , Myocardium/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Zebrafish/embryology
18.
Toxicol Sci ; 99(1): 326-37, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17562736

ABSTRACT

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.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Gene Expression Profiling/methods , Gene Expression/drug effects , Genomics/methods , Liver/drug effects , Animals , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Endpoint Determination , Genomic Islands , Isomerism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Reproducibility of Results , Salivary alpha-Amylases , Transcription Factors/biosynthesis , Transcription Factors/genetics
19.
Glycobiology ; 17(2): 231-47, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17110391

ABSTRACT

Although recent studies have extended our understanding of agrin's function during development, its function in the central nervous system (CNS) is not clearly understood. To address this question, zebrafish agrin was identified and characterized. Zebrafish agrin is expressed in the developing CNS and in nonneural structures such as somites and notochord. In agrin morphant embryos, acetylcholine receptor (AChR) cluster number and size on muscle fibers at the choice point were unaffected, whereas AChR clusters on muscle fibers in the dorsal and ventral regions of the myotome were reduced or absent. Defects in the axon outgrowth by primary motor neurons, subpopulations of branchiomotor neurons, and Rohon-Beard sensory neurons were also observed, which included truncation of axons and increased branching of motor axons. Moreover, agrin morphants exhibit significantly inhibited tail development in a dose-dependent manner, as well as defects in the formation of the midbrain-hindbrain boundary and reduced size of eyes and otic vesicles. Together these results show that agrin plays an important role in both peripheral and CNS development and also modulates posterior development in zebrafish.


Subject(s)
Agrin/physiology , Motor Neurons/physiology , Nervous System/embryology , Zebrafish Proteins/physiology , Zebrafish/embryology , Agrin/analysis , Agrin/genetics , Animals , Axons/chemistry , Axons/physiology , Cell Differentiation , Embryo, Nonmammalian , Embryonic Development/genetics , Motor Neurons/chemistry , Motor Neurons/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Nervous System/chemistry , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Cholinergic/analysis , Receptors, Cholinergic/metabolism , Zebrafish/abnormalities , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics
20.
Birth Defects Res A Clin Mol Teratol ; 73(11): 868-75, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16237707

ABSTRACT

BACKGROUND: Vitamin A (retinol), in the form of retinoic acid (RA), is essential for normal development of the human embryo. Studies in the mouse and zebrafish have shown that retinol is metabolized in the developing spinal cord and must be maintained in a precise balance along the anteroposterior axis. Both excess and deficiency of RA can affect morphogenesis, including failures of neural tube closure. METHODS: We chose to investigate 5 genes involved in the metabolism or synthesis of RA, ALDH1A2, CYP26A1, CYP26B1, CRABP1, and CRABP2, for their role in the development of human neural tube defects, such as spina bifida. RESULTS: An association analysis using both allelic and genotypic single-locus tests revealed a significant association between the risk for spina bifida and 3 polymorphisms in the gene ALDH1A2; however, we found no evidence of a significant multilocus association. CONCLUSIONS: These results may suggest that polymorphisms in ALDH1A2 may influence the risk for lumbosacral myelomeningocele in humans.


Subject(s)
Alleles , Meningomyelocele/enzymology , Oxidoreductases/genetics , Polymorphism, Genetic , Receptors, Retinoic Acid/genetics , Animals , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , Linkage Disequilibrium , Male , Meningomyelocele/genetics , Meningomyelocele/metabolism , Mice , Organogenesis/genetics , Quantitative Trait Loci , Vitamin A/genetics , Vitamin A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...