Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 827686, 2022.
Article in English | MEDLINE | ID: mdl-35548346

ABSTRACT

The clinical heterogeneity of heart failure has challenged our understanding of the underlying genetic mechanisms of this disease. In this respect, large-scale patient DNA sequencing studies have become an invaluable strategy for identifying potential genetic contributing factors. The complex aetiology of heart failure, however, also means that in vivo models are vital to understand the links between genetic perturbations and functional impacts as part of the process for validating potential new drug targets. Traditional approaches (e.g., genetically-modified mice) are optimal for assessing small numbers of genes, but less practical when multiple genes are identified. The zebrafish, in contrast, offers great potential for higher throughput in vivo gene functional assessment to aid target prioritisation, by providing more confidence in target relevance and facilitating gene selection for definitive loss of function studies undertaken in mice. Here we used whole-exome sequencing and bioinformatics on human patient data to identify 3 genes (API5, HSPB7, and LMO2) suggestively associated with heart failure that were also predicted to play a broader role in disease aetiology. The role of these genes in cardiovascular system development and function was then further investigated using in vivo CRISPR/Cas9-mediated gene mutation analysis in zebrafish. We observed multiple impacts in F0 knockout zebrafish embryos (crispants) following effective somatic mutation, including changes in ventricle size, pericardial oedema, and chamber malformation. In the case of lmo2, there was also a significant impact on cardiovascular function as well as an expected reduction in erythropoiesis. The data generated from both the human in silico and zebrafish in vivo assessments undertaken supports further investigation of the potential roles of API5, HSPB7, and LMO2 in human cardiovascular disease. The data presented also supports the use of human in silico genetic variant analysis, in combination with zebrafish crispant phenotyping, as a powerful approach for assessing gene function as part of an integrated multi-level drug target validation strategy.

2.
Br J Pharmacol ; 178(13): 2671-2689, 2021 07.
Article in English | MEDLINE | ID: mdl-33768524

ABSTRACT

BACKGROUND AND PURPOSE: Functional brain imaging using genetically encoded Ca2+ sensors in larval zebrafish is being developed for studying seizures and epilepsy as a more ethical alternative to rodent models. Despite this, few data have been generated on pharmacological mechanisms of action other than GABAA antagonism. Assessing larval responsiveness across multiple mechanisms is vital to test the translational power of this approach, as well as assessing its validity for detecting unwanted drug-induced seizures and testing antiepileptic drug efficacy. EXPERIMENTAL APPROACH: Using light-sheet imaging, we systematically analysed the responsiveness of 4 days post fertilisation (dpf; which are not considered protected under European animal experiment legislation) transgenic larval zebrafish to treatment with 57 compounds spanning more than 12 drug classes with a link to seizure generation in mammals, alongside eight compounds with no such link. KEY RESULTS: We show 4dpf zebrafish are responsive to a wide range of mechanisms implicated in seizure generation, with cerebellar circuitry activated regardless of the initiating pharmacology. Analysis of functional connectivity revealed compounds targeting cholinergic and monoaminergic reuptake, in particular, showed phenotypic consistency broadly mapping onto what is known about neurotransmitter-specific circuitry in the larval zebrafish brain. Many seizure-associated compounds also exhibited altered whole brain functional connectivity compared with controls. CONCLUSIONS AND IMPLICATIONS: This work represents a significant step forward in understanding the translational power of 4dpf larval zebrafish for use in neuropharmacological studies and for studying the events driving transition from small-scale pharmacological activation of local circuits, to the large network-wide abnormal synchronous activity associated with seizures.


Subject(s)
Brain , Zebrafish , Animals , Brain/diagnostic imaging , Functional Neuroimaging , Larva , Seizures/chemically induced , Seizures/drug therapy
3.
Environ Sci Technol ; 53(1): 463-474, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30520632

ABSTRACT

The plastic monomer bisphenol A (BPA) is one of the highest production volume chemicals in the world and is frequently detected in wildlife and humans, particularly children. BPA has been associated with numerous adverse health outcomes relating to its estrogenic and other hormonal properties, but direct causal links are unclear in humans and animal models. Here we simulated measured (1×) and predicted worst-case (10× ) maximum fetal exposures for BPA, or equivalent concentrations of its metabolite MBP, using fluorescent reporter embryo-larval zebrafish, capable of quantifying Estrogen Response Element (ERE) activation throughout the body. Heart valves were primary sites for ERE activation by BPA and MBP, and transcriptomic analysis of microdissected heart tissues showed that both chemicals targeted several molecular pathways constituting biomarkers for calcific aortic valve disease (CAVD), including extra-cellular matrix (ECM) alteration. ECM collagen deficiency and impact on heart valve structural integrity were confirmed by histopathology for high-level MBP exposure, and structural defects (abnormal curvature) of the atrio-ventricular valves corresponded with impaired cardiovascular function (reduced ventricular beat rate and blood flow). Our results are the first to demonstrate plausible mechanistic links between ERE activation in the heart valves by BPA's reactive metabolite MBP and the development of valvular-cardiovascular disease states.


Subject(s)
Benzhydryl Compounds , Zebrafish , Animals , Child , Estrogens , Humans , Phenols
4.
Sci Rep ; 8(1): 15903, 2018 Oct 23.
Article in English | MEDLINE | ID: mdl-30349014

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

5.
Sci Rep ; 7(1): 6581, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747660

ABSTRACT

Functional neuroimaging, using genetically-encoded Ca2+ sensors in larval zebrafish, offers a powerful combination of high spatiotemporal resolution and higher vertebrate relevance for quantitative neuropharmacological profiling. Here we use zebrafish larvae with pan-neuronal expression of GCaMP6s, combined with light sheet microscopy and a novel image processing pipeline, for the 4D profiling of chemoconvulsant action in multiple brain regions. In untreated larvae, regions associated with autonomic functionality, sensory processing and stress-responsiveness, consistently exhibited elevated spontaneous activity. The application of drugs targeting different convulsant mechanisms (4-Aminopyridine, Pentylenetetrazole, Pilocarpine and Strychnine) resulted in distinct spatiotemporal patterns of activity. These activity patterns showed some interesting parallels with what is known of the distribution of their respective molecular targets, but crucially also revealed system-wide neural circuit responses to stimulation or suppression. Drug concentration-response curves of neural activity were identified in a number of anatomically-defined zebrafish brain regions, and in vivo larval electrophysiology, also conducted in 4dpf larvae, provided additional measures of neural activity. Our quantification of network-wide chemoconvulsant drug activity in the whole zebrafish brain illustrates the power of this approach for neuropharmacological profiling in applications ranging from accelerating studies of drug safety and efficacy, to identifying pharmacologically-altered networks in zebrafish models of human neurological disorders.


Subject(s)
Brain/drug effects , Brain/diagnostic imaging , Convulsants/administration & dosage , Functional Neuroimaging/methods , Animals , Spatio-Temporal Analysis , Zebrafish
6.
PLoS One ; 12(1): e0168837, 2017.
Article in English | MEDLINE | ID: mdl-28045944

ABSTRACT

At high internal doses, pharmaceuticals have the potential for inducing biological/pharmacological effects in fish. One particular concern for the environment is their potential to bioaccumulate and reach pharmacological levels; the study of these implications for environmental risk assessment has therefore gained increasing attention. To avoid unnecessary testing on animals, in vitro methods for assessment of xenobiotic metabolism could aid in the ecotoxicological evaluation. Here we report the use of a 3-D in vitro liver organoid culture system (spheroids) derived from rainbow trout to measure the metabolism of seven pharmaceuticals using a substrate depletion assay. Of the pharmaceuticals tested, propranolol, diclofenac and phenylbutazone were metabolised by trout liver spheroids; atenolol, metoprolol, diazepam and carbamazepine were not. Substrate depletion kinetics data was used to estimate intrinsic hepatic clearance by this spheroid model, which was similar for diclofenac and approximately 5 fold higher for propranolol when compared to trout liver microsomal fraction (S9) data. These results suggest that liver spheroids could be used as a relevant and metabolically competent in vitro model with which to measure the biotransformation of pharmaceuticals in fish; and propranolol acts as a reproducible positive control.


Subject(s)
Drug Evaluation, Preclinical , Liver/drug effects , Oncorhynchus mykiss/metabolism , Water Pollutants, Chemical/analysis , Animals , Atenolol/pharmacology , Biotransformation , Carbamazepine/pharmacology , Diazepam/pharmacology , Diclofenac/pharmacology , Female , Kinetics , Liver/metabolism , Metoprolol/pharmacology , Models, Animal , Phenylbutazone/pharmacology , Propranolol/pharmacology , Tandem Mass Spectrometry , Xenobiotics/pharmacology
7.
J Pharmacol Toxicol Methods ; 69(1): 30-8, 2014.
Article in English | MEDLINE | ID: mdl-24140389

ABSTRACT

INTRODUCTION: Despite effective in vitro preclinical strategies to identify cardiovascular (CV) liabilities, there remains a need for early functional assessment prior to complex in vivo mammalian models. The larval zebrafish (Danio rerio, Zf) has been suggested for this role: previous data suggest that cardiac electrophysiology and vascular ultrastructure are comparable with mammals, and also indicate responsiveness of individual Zf CV system endpoints to some functional modulators. Little information is, however, available regarding integrated functional CV responses to drug treatment. Consequently, we developed a novel larval Zf model capable of simultaneous quantification of chronotropic, inotropic and arrhythmic effects, alongside measures of blood flow and vessel diameter. METHODS: Non-invasive video analysis of the heart and dorsal aorta of anaesthetized and agarose-embedded larval ZF was used to measure multiple cardiovascular endpoints, simultaneously, following treatment with a range of functional modulators of CV physiology. RESULTS: Changes in atrial and ventricular beat frequencies were detected in response to acute treatment with cardio-stimulants (adrenaline and theophylline), and negative chrono/inotropes (cisapride, haloperidol, terfenadine and verapamil). Arrhythmias were also observed including terfenadine-induced 2:1 atrial-ventricular (A-V) block, a previously proposed hERG surrogate measure. Significant increases in blood flow were detected in response to adrenaline and theophylline exposure; and decreases after cisapride, haloperidol, terfenadine, and verapamil treatment. Using dorsal aorta (DA) blood flow and ventricular beat rate, surrogate stoke volumes were also calculated for all compounds. DISCUSSION: These data support the use of this approach for CV function studies. Moreover the throughput and compound requirements (approximately 3 compounds/person effort/week and <10 mg) make our approach potentially suitable for higher throughput drug safety and efficacy applications, pending further assessment of ZF-mammalian pharmacological comparability.


Subject(s)
Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacology , Cardiovascular Physiological Phenomena/drug effects , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Zebrafish/physiology , Animals , Larva/drug effects , Models, Animal , Regional Blood Flow/drug effects , Regional Blood Flow/physiology
8.
Environ Toxicol ; 27(10): 573-82, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21384489

ABSTRACT

The conservation of common physiological systems across vertebrate classes suggests the potential for certain pharmaceuticals, which have been detected in surface waters, to produce biological effects in nontarget vertebrates such as fish. However, previous studies assessing the effects of such compounds in fish have not taken into account the potential for metabolism and elimination. This study aimed to assess if propranolol, a ß-adrenergic receptor antagonist or ß-blocker, could modulate EROD activity (indicative of CYP1A activity) in rainbow trout (Oncorhynchus mykiss) gills and liver. For this, an in vivo time course exposure with 1 mg/L was conducted. Additionally, using measured in vivo plasma concentrations, an in vitro exposure at human therapeutic levels was undertaken. This allowed comparison of in vitro and in vivo rates of EROD activity, thus investigating the applicability of cell preparations as surrogates for whole animal enzyme activity analysis. In vitro exposure of suspended liver and gill cells at concentrations similar to in vivo levels resulted in EROD activity in both tissues, but with significantly higher rates (up to six times in vivo levels). These results show that propranolol exposure elevated EROD activity in the liver and gill of rainbow trout, and that this is demonstrable both in vivo (albeit nonsignificantly in the liver) and in vitro, thus supporting the use of the latter as a surrogate of the former. These data also provide an insight into the potential role of the gill as a site of metabolism of pharmaceuticals in trout, suggesting that propranolol (and feasibly other pharmaceuticals) may undergo "first pass" metabolism in this organ.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Gills/enzymology , Liver/enzymology , Oncorhynchus mykiss/metabolism , Propranolol/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cells, Cultured , Cytochrome P-450 CYP1A1/drug effects , Female , Gills/cytology , Gills/drug effects , Hepatocytes/drug effects , Hepatocytes/enzymology , Liver/drug effects , Propranolol/blood
9.
Environ Toxicol Chem ; 29(12): 2831-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20836071

ABSTRACT

Two studies to examine the effect of waterborne clofibric acid (CA) on growth-rate and condition of rainbow trout were conducted using accepted regulatory tests (Organisation for Economic Co-operation and Development [OECD] 215). The first study (in 2005) showed significant reductions after 21 d of exposure (21-d growth lowest-observed-effect concentration [LOEC] = 0.1 µg/L, 21-d condition LOEC = 0.1 µg/L) that continued to 28 d. Growth rate was reduced by approximately 50% (from 5.27 to 2.67% per day), while the condition of the fish reduced in a concentration-dependant manner. Additionally, in a concentration-dependent manner, significant changes in relative liver size were observed, such that increasing concentrations of CA resulted in smaller livers after 28-d exposure. A no-observed-effect concentration (NOEC) was not achieved in the 2005 study. An expanded second study (in 2006) that included a robust bridge to the 2005 study, with four replicate tanks of eight individual fish per concentration, did not repeat the 2005 findings. In the 2006 study, no significant effect on growth rate, condition, or liver biometry was observed after 21 or 28 d (28-d growth NOEC = 10 µg/L, 28-d condition NOEC = 10 µg/L), contrary to the 2005 findings. We do not dismiss either of these findings and suggest both are relevant and stand for comparison. However, the larger 2006 study carries more statistical power and multiple-tank replication, so probably produced the more robust findings. Despite sufficient statistical power in each study, interpretation of these and similar studies should be conducted with caution, because much significance is placed on the role of limited numbers of individual and tank replicates and the influence of control animals.


Subject(s)
Clofibric Acid/toxicity , Oncorhynchus mykiss/growth & development , Toxicity Tests, Chronic/methods , Water Pollutants, Chemical/toxicity , Animals , Anticholesteremic Agents/blood , Anticholesteremic Agents/toxicity , Body Composition/drug effects , Body Weight/drug effects , Clofibric Acid/blood , Dose-Response Relationship, Drug , Female , Oncorhynchus mykiss/blood , Organ Size/drug effects , Water Pollutants, Chemical/blood
10.
Integr Environ Assess Manag ; 6(1): 38-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19558206

ABSTRACT

Over recent years, human pharmaceuticals have been detected in the aquatic environment. This, combined with the fact that many are (by design) biologically active compounds, has raised concern about potential impacts in wildlife species. This concern was realized with two high-profile cases of unforeseen environmental impact (i.e., estrogens and diclofenac), which have led to a flurry of work addressing how best to predict such effects in the future. One area in which considerable research effort has been made, partially in response to regulatory requirements, has been on the potential use of preclinical and clinical pharmacological and toxicological data (generated during drug development from nonhuman mammals and humans) to predict possible effects in nontarget, environmentally relevant species: so-called read across. This approach is strengthened by the fact that many physiological systems are conserved between mammals and certain environmentally relevant species. Consequently, knowledge of how a pharmaceutical works (the "mode-of-action," or MoA) in nonclinical species and humans could assist in the selection of appropriate test species, study designs, and endpoints, in an approach referred to as "intelligent testing." Here we outline the data available from the human drug development process and suggest how this might be used to design a testing strategy best suited to the specific characteristics of the drug in question. In addition, we review published data that support this type of approach, discuss the potential pitfalls associated with read across, and identify knowledge gaps that require filling to ensure accuracy in the extrapolation of data from preclinical and clinical studies, for use in the environmental risk assessment of human pharmaceuticals.


Subject(s)
Drug Discovery , Environmental Monitoring/methods , Risk Assessment/methods , Humans , Pharmaceutical Preparations/analysis
11.
Philos Trans R Soc Lond B Biol Sci ; 364(1534): 3377-90, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19833649

ABSTRACT

Exposure to environmental chemicals can have negative consequences for wildlife and even cause localized population extinctions. Resistance to chemical stress, however, can evolve and the mechanisms include desensitized target sites, reduced chemical uptake and increased metabolic detoxification and sequestration. Chemical resistance in wildlife populations can also arise independently of exposure and may be spread by gene flow between populations. Inbreeding-matings between closely related individuals-can have negative fitness consequences for natural populations, and there is evidence of inbreeding depression in many wildlife populations. In some cases, reduced fitness in inbred populations has been shown to be exacerbated under chemical stress. In chemical testing, both inbred and outbred laboratory animals are used and for human safety assessments, iso-genic strains (virtual clones) of mice and rats are often employed that reduce response variation, the number of animals used and associated costs. In contrast, for environmental risk assessment, strains of animals are often used that have been selectively bred to maintain heterozygosity, with the assumption that they are better able to predict adverse effects in wild, genetically variable, animals. This may not necessarily be the case however, as one outbred strain may not be representative of another or of a wild population. In this paper, we critically discuss relationships between genetic variation, inbreeding and chemical effects with the intention of seeking to support more effective chemical testing for the protection of wildlife.


Subject(s)
Animals, Wild/genetics , Ecotoxicology/methods , Environmental Pollutants/toxicity , Genetic Variation , Inbreeding , Animals
12.
Environ Toxicol Chem ; 28(12): 2622-31, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19642832

ABSTRACT

Transformation products of pharmaceuticals formed by human metabolism within sewage treatment plant or receiving waters are predicted, in most cases, to be less toxic than the parent compound to common aquatic species. However, there is little available data to demonstrate whether this is generally the case. In the present study, a framework was developed to guide testing of transformation products using phototransformation of the beta-blocker propranolol to test the hypothesis for this particular transformation route. Phototransformation is an important depletion mechanism of some pharmaceuticals in surface waters with fast reaction rate constants at environmentally relevant conditions. Samples of propranolol in deionized water (DIW) and river water (RW) were exposed to a solar simulator (lambda: 295-800 nm) and comparative toxicity of propranolol and its degraded mixtures measured using algal (Pseudokirchneriella subcapitata) and rotifer (Brachionus calyciflorus) screening tests. Results suggested a reduction of toxicity in photodegraded mixtures compared to the parent active pharmaceutical ingredient in all samples tested. Chemical analysis of effect test solutions supported the hypothesis that propranolol was transformed into compounds that appear to be less toxic to the organisms tested under the study conditions. Although the reactions were much faster in RW than in DIW, profiles of transformation products were similar in both matrices at two starting concentrations (1 and 10 mg/L). Results for propranolol implied that the reduction of toxicity using algal and rotifer screening tests was probably due to the production of more hydrophilic and more polar transformation products. Such results will provide useful insights into the environmental risk assessment of pharmaceuticals by taking into account their transformation products.


Subject(s)
Eukaryota/drug effects , Propranolol/toxicity , Rotifera/drug effects , Water Pollutants, Chemical/toxicity , Animals , Female , Photolysis , Propranolol/chemistry , Rivers
13.
Aquat Toxicol ; 93(4): 217-24, 2009 Jul 26.
Article in English | MEDLINE | ID: mdl-19515433

ABSTRACT

Pharmaceuticals in the environment (PIE) are of importance since these compounds are designed to affect biological receptors/enzymes that are often conserved across vertebrate families. Across-species extrapolation of these therapeutic targets suggests potential for impacting amphibia and fish in the aquatic environment. Due to the scarcity of relevant ecotoxicological data, the long-tem impact of PIE remains a research question. Efficient use of mammalian data has been proposed to better understand and predict the potential for a given pharmaceutical to impact the environment. Using a model cardiovascular pharmaceutical (propranolol, a non-specific beta(1)/beta(2)-adrenergic antagonist), the hypothesis that mammalian data can be used to predict toxicity in fish was tested. Rainbow trout (Oncorhynchus mykiss (Walbaum)) have beta-adrenergic signalling mechanisms analogous to human cardiovascular receptors that respond to pharmacological doses of agonists and antagonists. Trout absorbed propranolol from water such that after 40 days of exposure, the linear relationship was [plasma] - 0.59[water] (n - 31, r - 0.96). Growth rate was affected only at very high aqueous concentrations (10-day (growth)NOEC - 1.0 and (growth)LOEC - 10 mg/l). Growth recovered with time (40-day (growth)NOEC - 10 mg/l), suggesting possible adaptation to the pharmaceutical, although the internal plasma concentration in trout exposed to 10mg propranolol/l of water was higher than the mammalian therapeutic plasma concentration. Additional endpoints suggested subtle changes of liver and heart size at much lower concentrations may have occurred, although these were not concentration-related. There was, however, a dose-dependent effect upon overall body condition. The trout plasma concentrations at these effective aqueous concentrations fell within the range of mammalian effective plasma concentrations, supporting the potential for developing 'read-across' from mammalian pharmacology safety data to fish ecotoxicology. Despite these effects at relatively high concentrations, propranolol is not expected to pose a risk to fish at the concentrations considered to be present in the aquatic environment.


Subject(s)
Adrenergic beta-Antagonists/toxicity , Oncorhynchus mykiss/blood , Oncorhynchus mykiss/growth & development , Propranolol/toxicity , Water Pollutants, Chemical/toxicity , Adrenergic beta-Antagonists/blood , Adrenergic beta-Antagonists/pharmacokinetics , Animals , Biometry , Body Composition/drug effects , Body Weight/drug effects , Fresh Water/chemistry , Heart/drug effects , Heart/growth & development , Liver/drug effects , Liver/growth & development , Organ Size/drug effects , Propranolol/blood , Propranolol/pharmacokinetics , Water Pollutants, Chemical/blood , Water Pollutants, Chemical/pharmacokinetics
14.
Chemosphere ; 53(3): 223-35, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12919782

ABSTRACT

A number of currently used industrial chemicals are estrogenic, and therefore have potential to disrupt sexual differentiation in vertebrate wildlife during critical developmental windows. We assessed the effect of larval exposure to bisphenol A (BPA) on growth, development and sexual differentiation of the gonad in the African Clawed frog, Xenopus laevis. Larvae were maintained in flow-through conditions at 22 +/- 1 degrees C and exposed to BPA at mean measured concentrations of 0.83, 2.1, 9.5, 23.8, 100, and 497 microg/l, from developmental stages 43/45-66 (completion of metamorphosis). Each test concentration, plus dilution water control (DWC) and positive control (17beta-estradiol (E2), 2.7 microg/l) employed four replicate test vessels with 40 larvae per tank. Individual froglets were removed from test vessels upon reaching stage 66, and the study was terminated at 90 days. Froglets were dissected and sex was determined by inspection of gross gonadal morphology. Test concentrations of BPA had no effect on survival, growth, developmental stage distributions at exposure days 32 and 62, or mean time to completion of metamorphosis, compared to DWC. Analysis of post-metamorphic sex ratio, determined by gross gonadal morphology, indicated no significant deviations from expected (50:50) sex ratio, in DWC or any BPA test concentration. In contrast, exposure of larvae to (E2) resulted in feminisation, with sex ratio deviating significantly (31% male, replicates pooled). Exposure to BPA in the concentration range 0.83-497 microg/l in flow-through conditions had no observable effect on larval growth, development or sexual differentiation (as determined by gross gonadal morphology) in this study.


Subject(s)
Estrogens, Non-Steroidal/toxicity , Phenols/toxicity , Xenopus laevis/physiology , Animals , Benzhydryl Compounds , Gonads/growth & development , Larva/drug effects , Larva/growth & development , Metamorphosis, Biological/drug effects , Sex Differentiation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...