Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Apr 16.
Published in final edited form as: Chem Res Toxicol. 2019 Nov 16;33(1):95–118. doi: 10.1021/acs.chemrestox.9b00335

Use of Zebrafish in Drug Discovery Toxicology

Steven Cassar , Isaac Adatto , Jennifer L Freeman §, Joshua T Gamse , Iñaki Iturria , Christian Lawrence #, Arantza Muriana , Randall T Peterson , Steven Van Cruchten , Leonard I Zon
PMCID: PMC7162671  NIHMSID: NIHMS1579617  PMID: 31625720

Abstract

Unpredicted human safety events in clinical trials for new drugs are costly in terms of human health and money. The drug discovery industry attempts to minimize those events with diligent preclinical safety testing. Current standard practices are good at preventing toxic compounds from being tested in the clinic; however, false negative preclinical toxicity results are still a reality. Continual improvement must be pursued in the preclinical realm. Higher-quality therapies can be brought forward with more information about potential toxicities and associated mechanisms. The zebrafish model is a bridge between in vitro assays and mammalian in vivo studies. This model is powerful in its breadth of application and tractability for research. In the past two decades, our understanding of disease biology and drug toxicity has grown significantly owing to thousands of studies on this tiny vertebrate. This Review summarizes challenges and strengths of the model, discusses the 3Rs value that it can deliver, highlights translatable and untranslatable biology, and brings together reports from recent studies with zebrafish focusing on new drug discovery toxicology.

Graphical Abstract

graphic file with name nihms-1579617-f0001.jpg

INTRODUCTION

In pharmaceutical drug discovery, the number of compounds discovered in early target- or phenotypic-based screens for drug candidates far outweighs the number that advances as clinical candidates, of which relatively few to none progress to clinical trials.13 This nonclinical attrition is the result of diligent studies conducted in animal models designed to demonstrate bioavailability, efficacy, and safety. During this early phase, depending on the reporting source, between 40 and 80% of the compounds are halted in development due to safety concerns.4,5 The current practice serves well to provide many safe molecules to clinical studies,6 yet surprising clinical toxicities are still a reality. An analysis of a pharmaceutical-industry-wide preclinical-to-clinical database7 showed that overall, the specificity of preclinical toxicology studies is good (>80%), reflecting a low level of false positive predictions of toxicity. However, the sensitivity is low (<50%), reflecting a high false negative rate meaning unpredicted human safety events.

The later they occur during drug development, the more costly unpredicted safety events are in terms of human health and money. Improving preclinical testing practices could mitigate such losses. Advances in investigative toxicology, which aims to derisk safety and reveal toxic mechanisms of discovery candidates, have provided paths toward such improvement.8,9 Many of these include platforms that are considered alternatives to animal models in line with the 3Rs philosophy toward animal research.10,11 These include stem cells and other cell cultures,12,13 3D tissue models,1416 organs on chips,17 in silico prediction,18 and machine learning19 as well as humanized chimeric mouse models for more translatable data20 and nonmammalian animal testing in animals like the round worm Caenorhabditis elegans21 and teleost fish Danio rerio (zebrafish)2227 (Figure 1).

Figure 1.

Figure 1.

General body plans of a larval (top) and an adult (bottom) zebrafish indicating organ locations. Note the relative lengths of the two stages—approximately 5 days post fertilization for the larva and 3 months for the adult. In body volume, the larval fish is hundreds of times smaller than the adult. Cartoon used with permission from authors of ref 46 and The Journal of Clinical Investigation, copyright 2012.

Low sensitivity for clinical safety issues is the primary focus of efforts to improve preclinical toxicology testing, but mammalian models are also laborious, costly, and fraught with ethical questions concerning animal testing,28,29 affording other motivations for developing and implementing alternative assays. Regulatory requirements for new drug discoveries are rigorous; thus, acceptance of alternative toxicology assays by government agencies will rely on well-established predictivity of clinical toxicity.30 Predictivity of an alternative assay can be evaluated independently for each human biological system, organ, tissue, or cell type. This Review brings together literature reporting on potential and actual applications of zebrafish in drug discovery toxicology, sorted into sections based on organ or biological system.

ZEBRAFISH PROVIDE 3RS VALUE TO DRUG DISCOVERY TOXICOLOGY

The 3Rs, replacement, reduction, and refinement of animal studies in research,10,31,32 has evolved, over time, along with philosophies underlying the use of animals, regulatory directives, and new technologies, the latter of which can be beneficial to science as well as animal welfare. Among new approaches, the zebrafish has arisen as a popular alternative animal model. According to the European Commission Directive from 2010, experiments with the earliest life-stages of some animals are not regulated as animal studies; for zebrafish, independent feeding, which begins around 5 days post fertilization (dpf), is considered the first stage subject to regulation for animal experimentation. Therefore, work with zebrafish embryos/larvae under 5 dpf can be considered an alternative to animal testing. Some controversy exists around this; there are different interpretations of criteria by national and regional authorities, especially around 120–144 h postfertilization (hpf). Supporting the 5 dpf transition, according to a review of the literature and available data,33 where they include criteria such as yolk consumption, feeding, and swimming behavior, independent feeding becomes evident at 120 hpf.

HOW THE 3 RS APPLY TO ZEBRAFISH

  • Replacement: Zebrafish assays using larval zebrafish could be used to replace some animal toxicity studies, first establishing that the larval zebrafish is a relevant model for the system (target, gene, pathway, mechanism, tissue, organ, etc.) with validation studies.

  • Reduction: As a first-tier model for toxicity, zebrafish larvae could be used to identify toxic drug candidates, allowing safer molecules to be tested in mammalian models. In the end, this would ultimately reduce the number of animals used in testing.

  • Refinement: The embryonic and larval zebrafish model offers refinement to animal study design, because the embryos are fertilized externally and are transparent through the early days of life. This allows for noninvasive observation of toxicities and perhaps recovery.

CHALLENGES OF ZEBRAFISH AS AN ANIMAL MODEL

There are many reasons zebrafish have become a popular laboratory animal, some of which that make them easy to work with are described below. However, this model has several deficiencies compared to mammalian models, making certain aspects of toxicology cumbersome to interrogate in zebrafish and raising yet unanswered questions about the translatability of toxic potencies and affected tissues. For example, the most popular method for dosing embryonic and larval zebrafish is through solubilizing chemicals in water (in-water dosing). This poses two issues; one is that chemicals with poor solubility and/or absorption need to be injected into fish to gain sufficient exposure, limiting the pool of chemicals that can be tested in high-throughput screening to those that have good water solubility; the second is that in-water dosing may yield unique exposures compared to typical mammalian routes, as the fish are literally immersed and/or swimming in the treatment solution. How this second issue is manifested, and impacts the translatability of results, needs interrogation and likely varies depending on the biological system and chemicals being investigated.

One of the main strengths of zebrafish as a model organism, their small size, also poses challenges. Nonclinical toxicology studies are relied upon to predict a therapeutic window between efficacious and toxic exposures, guide clinical dosing upper limits based on those exposures, and deliver absorption, distribution, metabolism, and excretion (ADME) data. Toxic in-water dosing levels generated from larval zebrafish studies currently cannot be decidedly related to mammalian plasma levels; more work is needed in this area and, like the questions around unique exposures driven by in-water dosing, will likely depend on the system being interrogated. Methods for measuring plasma levels and ADME from larval zebrafish are currently being pursued but are in their early stages.34,35

Finally, because of phylogenetic distance manifested in anatomy and physiology differences, zebrafish will, in general, generate less translatable data to clinical toxicities compared to data from mammalian models. Strategic studies that employ zebrafish to interrogate systems and chemicals that have the highest likelihood of translatability will make the most of this model. Beyond considering gene similarity, congruence of cellular mechanisms, and comparable tissue biology, toxicology studies with zebrafish need to consider the regenerative capacity of the model. Zebrafish can regenerate multiple tissues including the fin, brain, retina, spinal cord, and heart.36,37 This regenerative capacity may impact translational toxicity end points as has been demonstrated for the retina.38

ENVIRONMENTAL PARAMETERS AND HUSBANDRY PRACTICES

An inherent advantage of zebrafish is their broad tolerance of environmental conditions.39 This trait makes it possible to rear and breed these animals for research using a wide array of protocols. Accordingly, there are very few standards for husbandry and management of the fish and their environment, and conventional approaches have long centered around the simple and straightforward goal of producing fish that are free from visible disease, breed well, and survive at rates high enough to complete experiments.40

While this flexibility has been a major driver in the growth of the zebrafish model overall, it somewhat paradoxically limits its adoption by the pharmaceutical industry. This is because other animal models (mice, rats, rabbits, dogs) already accepted and widely employed by drug companies for preclinical toxicology studies are, by comparison, well-established models, with commercially available standardized breeds/strains accompanied by robust historical control data. This allows researchers employing these models to exert a degree of control over a number of variables (e.g., diet, genetic background, pathogen status) that they are not able to achieve with fish. Without this level of standardization, preclinical studies using fish may be less reproducible and more challenging to undertake, especially when they involve collaborations among multiple groups at multiple sites.

The problem for the zebrafish model with respect to experimental variability is twofold; the inaccurate and/or incomplete reporting of husbandry and environmental conditions is compounded by the overall lack of available standards for researchers to follow. Reporting can be improved now by using tools like ARRIVE guidelines,41 protocols.io,42 or benchmark toxicity concentrations,43 but the overall lack of standards for diet, health monitoring, and genetic maintenance is a larger, more complicated puzzle to solve.44 Standards can be developed through improving scientific understanding of these factors to the point where broad recommendations for standards can be made with confidence. At the same time, commercial platforms are needed to support these efforts, for example, in the production of specialized diets and genetically defined and specific-pathogen-free (SPF) fish. At present, the lack of this infrastructure is limiting.

VALUE OF ZEBRAFISH AS AN ANIMAL MODEL

Conserved vertebrate biology, ease of husbandry, high fecundity, small size, rapid development, and transparent young are some of the main attractions of zebrafish as an alternative to mammals for toxicology studies. Although there are some major differences related to anatomy and physiology associated with an aquatic species, most zebrafish organs perform the same functions as their human counterparts and exhibit well-conserved physiology.45,46 Hundreds of adult zebrafish, each only about 4 cm long, can be easily housed in a standard aquarium, and there are several commercial options for automated water quality control and feeding. Successful mating of a single pair results in hundreds of externally fertilized embryos that develop rapidly. During early development, between 48 and 72 hpf, the development of most organs is nearly complete, except for organs in the gastrointestinal (GI) tract. After 76 hpf, the liver, pancreas, and gut are fully developed, and at 96 hpf, the GI tract is completely developed.47 As at this stage they are small enough to fit into individual wells of multiwell plates, toxicology can be conducted on hundreds of whole organisms in a hand-held platform using an amount of test article that would be required to dose only one or two mice.

The transparency of young zebrafish allows for noninvasive examination of organ development and toxic end points. This, along with the ease of generating transgenic models, lends itself to gene expression and cell-specific reporter assays, offering powerful options for real-time in vivo studies of toxic mechanisms.48 Biological effects of chemicals have been studied extensively in the zebrafish.49,50 Exposure to xenobiotics is typically accomplished by simply dissolving a chemical in the water after which phenotypic changes and/or toxic effects can be monitored. Eight small molecules discovered in zebrafish have been advanced into clinical trials,5158 illustrating the ability to move fundamental discoveries from zebrafish to humans. The zebrafish genome has been sequenced, and comparison to that of humans reveals that 70% of human genes have a zebrafish homologue, and 82% of human genes associated with disease have a zebrafish homologue.59 This allows for strategic application of this model when homology is high, and molecular players can be expected to interact with xenobiotics in translatable ways. For the reasons above, there is a long history of zebrafish being studied for genetics, cell biology, embryology, and environmental toxicology. Most toxicological studies using zebrafish have focused on environmental contaminants, but an increasing number are emerging in the field of pharmaceutical toxicology.48,60

LITERATURE SURVEY

The number of scientific publications mentioning zebrafish has grown steadily (Figure 2); in 2018, there were well over 3000, roughly 3 times the amount from 10 years ago. A similar trend is observed in those publications around zebrafish toxicity, with latest yearly reports being 4 times those from 10 years ago (Figure 2).

Figure 2.

Figure 2.

Publications on zebrafish for toxicology have grown over 4-fold in the last 10 years.

These publications originate not only from universities, hospital research centers, and regulatory or governmental entities but also pharma, biotech, chemical, cosmetic, tobacco, and nutraceutical companies. In recent years, the developmental, hepatic, and nervous systems are the main systems being interrogated for toxicity using zebrafish (Figure 3).

Figure 3.

Figure 3.

Numerous systems can be interrogated for toxic end points using zebrafish, and the number of publications is trending upward, as indicated by these examples.

SYSTEMS PHARMACOLOGY

Given that it is important to understand the capabilities and limitations of mammalian models, the need is even greater for animal models that are phylogenetically further removed from humans like zebrafish.45 Interspecies translation will improve if toxicologists move from an empirical to a mechanistic approach, in which multiple components of a biological system and their interactions can be monitored. Systems pharmacology is such an approach, combining the strengths of systems biology and pharmacometrics.47,61,62 It integrates modeling and simulations with preclinical and clinical data. Combining omics data with mechanistic and computational models can improve prediction of toxicity. Data that contribute to basic understanding of a toxicity can be gathered from in vitro experiments; however, as organ toxicities are complex, in vivo whole organism experiments, such as those using zebrafish larvae, deliver more detail to the understanding of the system. Combining all approaches will improve interspecies translation by understanding biological processes in a model organism and understanding how these systems differ among species.

EMBRYO TOXICITY

Zebrafish embryos are increasingly used for developmental toxicity screening of candidate drugs and chemicals (Figure 3). The zebrafish embryo model is already accepted as a validated alternative assay to assess fish acute toxicity (OECD, No. 236),63 and currently, the zebrafish embryo is also being explored as a potential replacement for one of the regulatory in vivo mammalian embryofetal developmental toxicity studies in view of the upcoming third revision of the ICH S5 guideline on detection of the toxicity to reproduction for human pharmaceuticals.64 In the 2015 final concept paper of this guideline, it is stated that in vitro, ex vivo, and nonmammalian in vivo assays are considered not to be the default approach for developmental toxicity testing but might be considered for regulatory purposes under limited circumstances.65 The 2017 draft of the revised guideline also includes a section on the qualification of alternative test systems for regulatory acceptance.64 The zebrafish embryo is of particular interest, as effects are assessed in a whole vertebrate organism during the entire period of organogenesis, in contrast to other alternative assays such as the whole embryo culture and embryonic stem cell test. Additionally, zebrafish embryo assays are less time-consuming and costly than the embryofetal development studies in rats and rabbits. This increases throughput for pharmaceutical and chemical companies, and if accepted as a regulatory test, this could also lead to replacement and/or reduction of animal studies in line with the 3Rs principle in animal research.

In general, concordance with the findings in mammalian developmental toxicity studies is high, reaching up to 8566 or 87%67 in some laboratories, but false negative and positive results are reported, and the results for the same compound can vary substantially between laboratories.6674 The latter can be explained by the large diversity in protocols, such as study design70 and calculation of the teratogenic index,66,67,7174 indicating a clear need for harmonization of this alternative assay when considering it for more than internal decision making. There may be several other reasons for discordance between laboratories and false negative/positive results in the zebrafish embryo developmental toxicity assay, such as internal concentration (compound uptake), metabolism, and species differences in mode of action. Compound uptake has already been addressed as a challenge earlier in this manuscript. Although the chorion is removed by some groups to ensure adequate exposure of the embryos,67 the former does not appear to be the major cause of impaired uptake for most small molecules.66 Solubility can be an issue for some compounds, especially at higher concentrations,73 and the actual exposure of the zebrafish embryos may be overestimated when assessing the medium concentrations without analysis of the internal concentrations.68 Recently, efforts have been undertaken to better predict embryotoxic medium concentrations in the zebrafish embryo assay and link them with embryotoxic rat plasma concentrations.75,76 As such, the assay can be refined and its predictivity enhanced. Regarding metabolism, it is generally accepted that the intrinsic biotransformation capacity of zebrafish embryos is low up to 72 hpf, especially for cytochrome P450 mediated reactions,77,78 and that exogenous metabolic activation, e.g., by rat liver microsomes, increases the sensitivity of the assay for some compounds.79 The latter should be kept in mind when negative results are obtained in the zebrafish embryo assay and the compound appears to be highly metabolized in man. Also, species differences in the mode of action of compounds need to be considered when using the zebrafish embryo assay. For instance, ribavirin has been reported as negative for zebrafish embryotoxicity but causes malformations in mammals by accumulation of ribavirin triphosphate in the erythrocytes.74 As zebrafish have nucleated erythrocytes and no accumulation of ribavirin triphosphate can occur, this may be a reason for the lack of malformations in this species. Finally, although organogenesis in zebrafish is well-characterized,80 the number of morphological end points evaluated in the zebrafish embryo may need to be extended to increase the sensitivity of the assay. The number of end points that are evaluated in the currently used zebrafish embryo protocols is much more limited compared to the exhaustive list of external, visceral, and skeletal end points in the in vivo mammalian embryofetal development studies.81

NEUROTOXICITY AND BEHAVIORAL ANALYSES

The zebrafish is being widely applied to study the mechanisms and pathogenesis of neurological disorders and diseases,8284 with great promise for drug discovery and toxicity testing in this realm.8587 The central nervous system of the zebrafish is similarly organized to that of other vertebrates and is well-described at multiple life-stages.88 The main structures, as well as many principal subdivisions of the brain, are found in the zebrafish,89 and behavioral studies have identified strong associations between the functions of zebrafish and human brain regions.90 One brain region that zebrafish do not have is a neocortex, so they cannot be used to model cognitive processes that rely on that region.91 Neurotransmitter systems, such as dopamine, GABA, glutamate, noradrenaline, serotonin, histamine, and acetylcholine, are present in zebrafish9296 and can serve as pharmacological and toxicological targets.

Locomotion is a complex behavior that requires an integrated response of the brain function, nervous system, and visual pathway. Given so, this behavioral pattern, in conjunction with automatic tracking methods, is increasingly gaining attention for its use in high-throughput screening of neurotoxic compounds.97 Embryos show a basic swimming capacity right after hatching, which is then refined to a beat and glide mode of swimming after 4 dpf.98 The startle reflex in response to tactile, visual, or auditory stimuli appears around 5 dpf.99 Adult zebrafish are able to display a varied repertoire of complex behaviors including memory and learning, social interactions, or prey hunting.89

The photomotor response assay, consisting of the automatic tracking of larval movement in response to alternative illumination conditions, is extensively used for the screening of neurotoxic effects.85,100 The principle of this assay relies on the specific patterns in response to the illumination transition. The light–dark transition increases the locomotor activity, while the dark–light transition decreases locomotor activity. Convulsant drugs, such as the GABA receptor antagonists pentylenetetrazol and picrotoxin cause a dose-dependent locomotor activity increase,93,101,102 and antiepileptic drugs such as diphenylhydantoin and valproic acid produce a decrease in locomotor activity103 (for examples of data from other neuronally active drugs, see Figure 4). The locomotor effects induced by these drugs are similar to those observed in rodents. In the case of some environmental hazards such as flame retardants, this assay has shown a similar sensitivity and value for predicting human neurotoxicity.104 However, non-translatable results have also been reported; for instance, exposure to venlafaxine showed an opposite effect to that observed in rodents.105

Figure 4.

Figure 4.

Example results from a zebrafish locomotor assay for seizure liability. Drugs which interact through different mechanisms with the nervous system evoke activity responses with different potencies. Each dot represents the maximum activity for a single larva. Red dots indicate that the group average was significantly higher than that of the vehicle (0 mM) group; p-values are given in red. Copied with permission from ref 92 and Elsevier, copyright 2019.

The touch-evoked response test, which tracks behavior of zebrafish larvae in response to a tactile stimulus given to the head or tail, is an indicator of the integration of sensory and motor function.99 For instance, larvae exposed to endosulfan sulfate and fipronil showed a decreased reactiveness in this assay.106,107 Assays based on the startle response to other stimuli such as visual and acoustic stimuli have also been useful to screen for neurotoxic effects.85,108,109

Likewise, adult zebrafish can be used to identify the anxiety-like effects of new pharmaceuticals in preclinical screenings. The most common models are the novel tank test (NTT) and the light–dark test (LDT). The NTT principle is similar to the rodent open field test. It relies on instinctive diving behavior in response to an unfamiliar environment, which diminishes with time.110 The LDT is based on fish scototaxis (innate preference for dark vs light areas). This behavior is associated with the natural tendency of wild zebrafish to prefer dark environments to avoid detection by potential predators.111

Recent advances of zebrafish in the field of neurology show great promise for future utility in studying disease. Neurologic-disease-associated genes are conserved in the zebrafish, enabling identification of molecular drug targets.112114 The ex vivo development of the zebrafish embryos allows observation of these conserved proteins with fluorescent tags, and the ease of genetic manipulation in the zebrafish has led to the creation of several zebrafish neurologic transgenic models. These models permit studies interrogating neurodevelopmental disorders such as autism,115 neuropsychiatric disorders including depression and anxiety,116,117 and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease.118121 Similar to other disease pathways, promise of the application of the CRISPR-Cas9 system to generate zebrafish knock-in and knock-out mutations is now set to rapidly expand the availability of transgenic models for drug discovery for neurological diseases and disorders.122

OCULAR TOXICITY

The vertebrate eye is highly conserved;123 thus, zebrafish offer an excellent model for studying ocular toxicity. Besides obvious anatomical similarities, including the cornea, lens, choroid, and retina as well as vascularization and innervation, the zebrafish and human eye have conserved gene expression, cellular makeup, and tissue architecture.124,125

Vision develops rapidly in zebrafish embryos, demonstrated by the reliance of 4 dpf larvae on visual cues for predation and evasive behavior.126 Primary retinal cell types are organized into recognizable layers, and the optic tectum is innervated by axons from the ganglion layer by 3 dpf.127 Homologous retinal layers and cell types are found in zebrafish for all mammalian counterparts;127129 these include the nerve fiber, ganglion, inner and outer plexiform, inner and outer nuclear, photo-receptor, and the pigmented epithelium.127,129 Zebrafish are diurnal and have color vision owing to a cone-dense retina, like that of humans; comparatively, rats and mice have relatively fewer cones and weak color vision.128

Even though the zebrafish retina is cone-dense, they lack a fovea, or a cone-concentrated area. Some other differences in zebrafish compared to mammalian retinas are that there are more cones than rods in zebrafish retinas, and they have cones that are sensitive to ultraviolet radiation as well as double cones that consist of a red-sensitive (principle) compartment and a green-sensitive (accessory) compartment.129 A key difference pertaining to the use of zebrafish as a model of toxicity is that the zebrafish retina (larval and adult) can regenerate. Retinal progenitor cells with the capacity to differentiate into any of the primary retinal neurons are derived from dividing Müller glial cells in response to tissue injury.130

Overall, the zebrafish retina is vascularized similarly to that of mammals, but there are some differences. Like mammals, angiogenesis from a central retinal artery forms the vasculature. And, initially, hyaloid vasculature is found in the lens but diminishes with age. The ganglion cell layer is vascularized throughout adulthood.131 However, unlike mature mammalian retinal vascularization, the inner and outer plexiform layers of the zebrafish retina are not vascularized. It has been proposed that less vascularization is needed for the thinner zebrafish retina, because it can rely on diffusion from surface vessels (hyaloid, choroid).131

Given the above-mentioned similarities between zebrafish and mammalian eyes, it is no surprise that several investigators have evaluated zebrafish as a model to interrogate pharmaceutical ocular toxicities. These studies have demonstrated concordance between results from zebrafish vision assays and human ocular reactions to drugs,132134 including chlorpromazine, cisplatin, gentamicin, quinine, deferoxamine, minoxidil, thioridazine, and vardenafil, among others. By testing drugs with no established human ocular effects as well as the oculartoxic drugs, those authors reported that the zebrafish assays were sensitive 68–83% of the time and specific 75–100% of the time, demonstrating utility for detecting oculartoxic chemicals. The optokinetic135,136 and optomotor response137 assays are two common ways to evaluate vision in adult and larval zebrafish. In the optokinetic assay, dark and light alternating vertical stripes are passed around an immobilized fish, and eye saccades are counted as an indicator of a healthy eye response to moving stimuli; in the optomotor assay, the fish are free swimming and allowed to respond to temporal or spatial changes in light. This latter assay often uses a second, comparator, stimulus (sound or touch) to assess general mobility. Both assays typically rely on video recording to help assess response. Recently, zebrafish larvae were used for detecting retinal toxicity in pharmaceutical pipeline discovery compounds, replacing studies in mammalian models85 and demonstrating clear value from zebrafish for drug discovery toxicology (Figure 5).

Figure 5.

Figure 5.

Representative ocular histology (top, 40×) and transmission electron microscopy (2000×) from larval zebrafish treated with vehicle (A + C) or a chemotherapeutic discovery compound (B + D). The vehicle-treated eye (A) depicts normal retinal features with homogeneously spaced cells. The retina from the compound-treated larva has decreased cell numbers in both the ganglion (GC) and inner nuclear (IN) layers (red arrows in B), and the inner plexiform (IP) layer has a disorganized, vacuolated appearance. The electron microscopic images reveal evenly spaced nuclei in the ganglion and inner nuclear layers for the vehicle-treated larva (C) but apoptosis (asterisk and red arrows), nuclear vacuolation/fragmentation (red block arrows), and vacuolation, associated with cell loss, in the retinal layers of the compound-treated larva. Similar effects were found in retinas of rats and dogs treated with that compound. NF, nerve fiber layer; OP, outer plexiform layer; PR, photoreceptor layer. Copied with permission from ref 84 and Oxford University Press, copyright 2014.

INTESTINE, PANCREAS, AND HEPATOBILIARY TOXICITY

The postesophageal digestive system in zebrafish consists of the intestine, pancreas, liver, and gall bladder. Among these organs, there are similarities and differences between zebrafish and mammals that should be considered when planning studies with, and analyzing results from, zebrafish. Numerous reports have highlighted the utility of zebrafish for interrogating drug toxicities on the digestive system.

Intestine.

Zebafish are agastric throughout their lives, but the anterior portion of the intestine, referred to as the intestinal bulb, has an enlarged lumen compared to the rest of the intestine and can act as a food reservoir; however, the intestinal bulb lacks gastric glands and thus has a neutral pH.138 Other similarities and differences concerning intestinal anatomy and physiology are reviewed in Brugman.139 Important similarities for drug toxicology are that, like mammals, intestinal movement relies on smooth muscles controlled by the enteric nervous system140 and that the intestine consists of an epithelium containing villi, enterocytes, goblet cells, and enteroendocrine cells. The epithelial cell maturation, differentiation, and turnover are like those of mammals.141 The key differences between zebrafish and mammalian intestines lie in the absence of structures or cell types in zebrafish. Missing from zebrafish are intestinal crypts, Peyer’s patches, Paneth cells, and a submucosal layer; smooth muscles connect directly to the mucosal layer.139

Zebrafish have been used extensively to model intestinal disorders, including investigations into microbiome influences, congenital disorders, intestinal inflammation, enteric nervous system/motility disorders, and intestinal tumorigenesis.142 Methods for imaging gut movement and measuring gut transit time in zebrafish have been recently improved143,144 allowing for higher sensitivity and throughput for toxicology (Figure 6). However, reports demonstrating the use of zebrafish for detecting potential intestinal toxicities from compounds in drug development are uncommon. Two reports, focused on assessing the predictive value of zebrafish for intestinal/gut toxicities, have been published. Those reports132,143 tested marketed drugs with and without GI effects in humans for effects on zebrafish gut contractility or transit time, respectively. Gut contractility was reportedly highly variable among the larval fish tested. This variability may have been caused by the poor/inconsistent bioavailability of some of the drugs, according to the authors. The gut transit time was also highly variable; however, by testing more (up to 24) larvae per treatment group, the sensitivity of the assay was improved. Both reports indicate that false negative results (i.e., no toxicity detected, where it was expected) are more common than false positive results. This may be due to low absorption for some compounds but also may be driven by the lack of certain cell types/structures in the zebrafish intestine or the agastric nature of the model. However, as a first-tier screen for intestinal toxicities, based on these reports, the zebrafish would be expected to identify >50% of toxic discovery compounds, allowing for prioritization of safer compounds for mammalian testing.

Figure 6.

Figure 6.

Fluorescent food transit can be detected and measured as a loss of signal over time from microscopic imaging (left) or, in a high-throughput fashion, from a corresponding gain in signal by plate-based spectrophotometry (right). The latter method allows the measurement of intestinal transit from dozens of larvae simultaneously in a microwell plate. Modified with permission from ref 143 and Elsevier, copyright 2015.

Pancreas.

The basic structure and function of the pancreas is conserved between zebrafish and mammals. An exocrine compartment secretes enzymes into the digestive tract, and an endocrine compartment produces insulin, glucagon, somatostatin, and ghrelin. Polypeptide-producing cells (PP cells) have not been positively identified in zebrafish pancreas.145 Most studies on pancreas using zebrafish have focused on the endocrine compartment, examining islet damage/protection for the sake of diabetes research.146 As an indicator of potential to translate chemical toxicity in the zebrafish pancreas to mammalian models/humans, these studies use challenge chemicals to damage zebrafish islets that are commonly used in mammalian models to drive the same ends.147150 Reports on the chemical toxicity of zebrafish exocrine pancreas are focused on embryo developmental toxicity driven by environmental contaminants;151,152 these are being used to expose sensitivities of the developing pancreas to common environmental pollutants as well as to shed light on cellular mechanisms involved in those toxicities.

Liver.

Drug-induced liver injury (DILI) is a major concern in the drug development industry.153,154 The most common assays for hepatotoxicity testing in drug development use human hepatocytes or HepG2 cells in culture. Two-dimensional (2D) and 3D in vitro human tissue models, including multiple cell types, are becoming popular, as they may more closely reflect what happens in the whole organ.155 The zebrafish liver performs the same functions as those of the human liver156 and is fully functional 5 days post fertilization, so larval zebrafish can be used to interrogate xenobiotics for hepatotoxicity in an in vitro format, providing whole organ/whole animal data from multiwell plates.157 Gene expression analysis after exposure to established human hepatotoxins revealed similar gene changes among in vitro models (human hepatocytes, mouse hepatocytes, rat hepatocytes, and zebrafish embryos) and in vivo models (mouse liver, rat liver, and zebrafish embryos), highlighting that zebrafish embryos have in vitro- and in vivo-like properties.158 As reported to date, there is good overlap in the metabolic capacity of the zebrafish liver and that of humans.159,160 The common metabolic pathways indicate a potential to predict hepatotoxic metabolites using zebrafish. The tissue architecture of the zebrafish liver differs from mammalian livers in that it is less organized, but it contains all the same cell types.156,160,161 In zebrafish, hepatocytes are arranged in tubules, rather than bilayered plates, as in mammals. Small bile ducts are found within the hepatocyte tubules, and these function to transfer bile to the gall bladder.161,162

Based on the biological similarities given above, and the relative ease for whole liver toxicology, the zebrafish offers an attractive model.163 Fluorescent markers for various cell types make it possible to visualize and quantify cell-specific compound effects in the zebrafish liver in real-time.156,164,165 More simpler techniques employ lipid stains, like oil-red-O, to visualize gross liver changes in the larval zebrafish.166 A study conducted by two major pharmaceutical companies demonstrated added predictive value from zebrafish liver toxicity assays for predicting DILI when they were used in conjunction with high-content cellular (hepatocyte) toxicity assays.167 Other studies have also shown congruence between zebrafish and human DILI using marketed drugs with known hepatotoxicity, like tetracycline, aspirin, erythromycin, cyclosporin A, amiodarone, and acetaminophen.25,27,158,168,169

Zebrafish have delivered popular models for demonstrating, or investigating mechanisms of, the hepatotoxicity from traditional medicines170172 and environmental contaminants, including those of pharmaceutical173175 and of other industrial origin.176178 In addition to these applications, zebrafish hepatotoxicity assays are being directly employed in early drug discovery toxicology. Evidence for this can be found in recent reports on studies designed to discover liver-protective compounds against known hepatotoxins,179 improve formulations or therapeutic indices for promising chemotherapies,180,181 and demonstrate novel compound efficacy in a zebrafish autoimmune disease model, which was coupled with an assessment of hepatotoxicity.182

Gall Bladder.

Lipophilic dyes can be used to visualize both lipid metabolism and transport in vivo in larval zebrafish.183 Lipid uptake, enzymatic metabolism, and transport in zebrafish have high homology to the human processes.184 Recent efforts at AbbVie have focused on using the reporter dye, PED6 (N-([6-(2,4-dinitro-phenyl)amino]hexanoyl)-1-palmitoyl-2-BODIPY-FLpentanoyl-sn-glycerol-3-phosphoethanolamine)183 to monitor for biliary toxicities from internal discovery compounds in zebrafish. PED6, as well as fluorescent transgenic zebrafish expressing reporter tags in gall bladder cells, have been used to identify biliary toxins and their mechanisms.185188 Although there appear to be no reports yet on the use of zebrafish for early drug discovery biliary toxicity, the utility is evident, and this is likely an underutilized application of the model.

NEPHROTOXICITY

The kidney is especially vulnerable to xenobiotic toxicity, because its function to remove toxic molecules from circulation relies on higher blood exposure than other organs. Thus, monitoring biomarkers that track kidney failure and kidney histology are primary end points in preclinical toxicology.189 Based on tractability of zebrafish larvae for research into vertebrate biology, they have recently been used to model kidney disease or nephrotoxicity.190193

The higher complexity of the mammalian kidney compared to that of zebrafish renders comprehensive modeling of kidney toxicity in zebrafish unpractical. However, nephrotoxicity can be reasonably interrogated with this model. The zebrafish larval pronephros has a simpler anatomy than that of the mammalian metanephros; it consists of only two nephrons with fused glomeruli. Despite the simple anatomy, similarities between the cellular constitution and function of the zebrafish pronephros and those of the mammalian metanephros make the zebrafish attractive. The zebrafish glomerulus contains fenestrated capillary endothelial cells and podocytes. Also, polarized epithelial cells with primary cilia line the nephron tubules, which are segmented into specialized regions for differential secretion from, and reabsorption into, the blood.194

Several investigators have demonstrated proof-of-concept that larval zebrafish can be used to monitor for drug-induced nephrotoxicity. Treatment with puromycin or knockdown of either CD2-associated protein or podicin caused effacement of podocytes and proteinuria;195 these authors also introduced a fluorescent tracer (FITC-labeled 70 kDa dextran) for live quantification of podocyte failure. More recently, electron microscopy was used to visualize the glomerular injury induced by puromycin in larval zebrafish.196 Three drugs known to cause tubulopathy in humans (paracetamol, gentamicin, and tenofovir) were shown to induce functional, morphological, and histopathological kidney changes in zebrafish larvae.192 Metabolic changes in biomarkers (purines, glutathione, and amino acids) associated with acute kidney injury were detected in larval zebrafish exposed to those same three drugs.197 Beyond proof-of-concept for detecting nephrotoxicity, the larval zebrafish are being used in phenotypic screens to discover compounds with a potential to repair acute kidney injury.198

ENDOCRINE TOXICITY

The activity of the endocrine system in vertebrates is mostly controlled by the hypothalamic–pituitary axis. The hypothalamus controls the activity of the adenohypophyseal through the release of neuroendocrine peptides, to which adenohypophyseal cells respond by secretion of specific hormones to evoke peripheral organ responses.199 Although their regulation and effects are tightly interconnected, the hypothalamic–pituitary axis can be functionally subdivided into the hypothalamic–pituitary–adrenal (HPA) axis, related mainly with stress and immune responses; the hypothalamic–pituitary–thyroid (HPT) axis, correlated with metabolism; and the hypothalamic–pituitary–gonadal (HPG) axis, related to reproduction.200

The structure and function of the endocrine system are strongly conserved among vertebrates, even if some differences can be observed between humans and zebrafish.201 Terrestrial vertebrates present a hypothalamic–pituitary portal system. Consequently, neuroendocrine peptides produced in the hypothalamus are secreted to the blood vessels and reach the adenohypophysis through the bloodstream. In contrast, in zebrafish, no hypothalamic–pituitary portal system is present, and the neurosecretory fibers enter the pituitary and release their hormones directly onto the adenohypophyseal cells.202 Similarly, zebrafish lack a distinctive adrenal gland but present a functionally comparable interrenal gland. This organ has no separation between the adrenal cortex and the medulla, containing both steroidogenic and chromaffin cells.201 Never theless, development of the steroidogenic cell lineage is well-conserved. For instance, both mammalian NR5A1 and its zebrafish homologue nr5a1a are essential for adrenal and interrenal gland development, respectively,203,204 as well as for activation of the side-chain cleavage enzyme cyp11a1, which is the rate-limiting enzyme in steroid biosynthesis.205

Most of the endocrine system in zebrafish develops during the first 5 dpf.201 Pituitary hormone gene expression starts at 48 hpf.206 Estrogen receptors are already present at 24 hpf,207 and aromatase gene expression, coding for the enzyme that catalyzes the biosynthesis of estrogens from precursor androgens on the brain, can be detected from 24 hpf.208 Key steroidogenic gene expression of the interrenal organ originates at 2 dpf.209 Thyroxin production of the thyroid gland starts at 3 dpf.210

Endocrine-disrupting chemicals (EDCs) are of high relevance for human and wildlife health, since endocrine signaling controls many essential physiological processes that impact the individual’s health, such as growth and development, stress response, and ultimately reproduction and population development.201 Studies on endocrine disruption in fish have focused mainly on the estrogen, androgen, thyroid, and steroidogenesis (EATS) pathways, in the context of environmental risk assessment of new chemical substances.211

Specific chemical biomarkers can be measured in zebrafish to screen for the endocrine activity of xenobiotics. For instance, one of the most commonly assessed responses is the induction of vitellogenin (vtg), an egg-yolk protein precursor produced in the liver and induced by estrogenic exposure.211 VTG is strongly induced after exposure to 10 ng/L of ethinylestradiol.212 In the same way, other markers related to endocrine activity such as thyroidal213,214 or steroidal hormones215 may be of interest but not amenable to high-throughput assays.

Gene expression analysis has been a useful method to identify specific pathways involved in the biological effects of EDC on zebrafish embryos. For instance, exposure to fadrozole led to a downregulation of vitellogenin (vtg) and brain aromatase (cyp19a1b) transcript levels in 96 hpf embryos.216 This key event produced an apical adverse effect of the sex ratio shifting toward male during sexual differentiation, indicating that the existing adverse outcome pathway (AOP) for aromatase inhibition in fish can be translated to the life-stage of sexual differentiation. A microarray study demonstrated the compensatory induction of androgen-pathway-related genes, sult2st3 and cyp2k22, after exposure to steroidal hormones.217 In the same way, expression of HPT axis-related marker genes thyroperoxidase (tpo), transthyretin (ttr), thyroid receptor α (trα), and deiodinase 2 (dio2) were altered in zebrafish embryos after exposure to perfluorinated compounds,218 polybrominated diphenyl ethers (PBDEs),219 and triazole fungicides,220 all well-established human endocrine disruptors.221,222

Multiple fluorescent reporter lines have been developed for testing of xenobiotics for endocrine activity. The EASZY assay makes use of the tg(cyp19a1b-GFP) transgenic zebrafish line to screen for estrogen active substances. This line expresses GFP in a concentration-dependent manner under the control of the gene encoding for the cyp19a1b brain aromatase.223 The model is highly sensitive to natural estrogens such as E2 (17ß-estradiol) as well as synthetic estrogens such as 17aethinylestradiol or diethylstilbestrol at nanomolar concentrations. The EASZY assay has been recently used to monitor estrogenic activities of waste and surface waters sampled across Europe.224 Another estrogen-responsive model was developed in a pigment-free “Casper” phenotype to identify the specific target tissues and quantify the response in whole fish.225 In the same way, different reporter lines for thyroid226,227 and glucocorticoid228 pathways are available. Those reporter lines are fast and cost-effective methods for the detection of the endocrine activity of xenobiotics and hence have considerable potential for both a high-content and high-throughput screen of endocrine disruptors.27

HEMATOLOGIC TOXICITY

Zebrafish have many blood cell types that are consistent with human peripheral blood cells. This includes red blood cells, neutrophils, monocytes, macrophages, T and B cells, and dendritic cells. The equivalent of a platelet is the thrombocyte. Reporters for each of these lineages have been generated as tissue-specific promoters driving fluorescent proteins. It is therefore possible to visualize all the blood cell types in different colors and evaluate toxicity.

Zebrafish hematopoietic stem cells have been discovered based on GFP reporters.229 It is possible to transplant the cells into irradiated adult fish, and the graft shows long-term stability for more than a year. There are many studies evaluating the trafficking of blood stem cells and their developmental biology. Most of the processes are conserved with those of humans.

A number of zebrafish mutants exist that represent hematologic disorders. In the original screen, there were 26 mutant complementation groups, and five of those genes proved to be novel genes, which later became associated with human diseases.230 It is possible to use zebrafish models and undertake genetic or chemical suppressor screens. This would potentially lead to novel therapeutics.

A few studies on hematologic toxicology have been undertaken using zebrafish. The hematopoietic system can be assayed in many ways.231 Using a double transgenic fish that has a GATA-1 promoter driving dsRED and globin LCR driving GFP, the stages of erythroid development can be studied.232 Adding drugs of the phenyl hydrazine family leads to lysis specifically of the mature red blood cells. This establishes a facile system for the evaluation of toxicity of chemicals and drugs.

CARDIOVASCULAR TOXICITY

Cardiovascular physiology is conserved between humans and zebrafish at anatomical, cellular, and membrane-biology levels. Zebrafish have been shown to provide a good model for cardiotoxicity. Many human cardiovascular drugs have shown comparable effects on zebrafish physiology,233 and numerous human cardiovascular disorders have been recapitulated in zebrafish genetic models.234 Systematic studies of drugs that cause QT prolongation in humans show a >95% conservation of effect in zebrafish.45 QT prolongation is a common cardiotoxicity discovered during new drug development.

Milan et al.235 developed an automated, high-throughput assay for bradycardia in zebrafish embryos, which was shown to correlate with QT prolongation in humans. They tested 100 compounds in the assay and showed that 22 of 23 drugs known to cause QT prolongation in humans cause bradycardia in zebrafish. In addition, the assay was able to detect drug– drug interactions that lead to QT prolongation, such as the well-known synergistic interactions between erythromycin and cisapride and between cimetidine and terfenadine. These interactions result from the physiological effects of one compound influencing the metabolism of the second compound and can only be detected in a whole organism. These results highlight the value of performing toxicity studies in zebrafish—zebrafish assays can achieve the scale and throughput of in vitro assays, but they occur in a relevant physiological setting, in which complex pharmacokinetic and pharmacodynamic processes remain intact.60 In early work, it was possible to show that more than 90% of drugs that cause repolarization toxicity in humans result in cognate electro-physiological effects in the zebrafish even as early as 48 hpf. Based on those data from 100 drugs, the specificity for reporting on repolarization toxicity in humans was 76%; the sensitivity was 80%, which increased to 96% when poorly absorbed drugs were injected.235

Letamendia et al.236 also showed how zebrafish is a model amenable for the automation of cardiotoxicity screenings. They developed an automated high-throughput platform for in vivo chemical screening on zebrafish embryos that includes automated methods for embryo dispensation, compound delivery, incubation, imaging, and analysis of the results. A validation of this platform with known positive and negative compounds was successfully carried out based on the rationale of compounds inhibiting the zERG channel, similar to the hERG channel in humans. This produces a 2:1 atrio:ventricular arrhythmia that resembles the QT prolongation in humans.

In vitro hERG binding and patchclamp assays are accepted by both the pharmaceutical industry and regulatory bodies as suitable methods for investigating the potential of compounds to cause QT prolongation. However, these assays only measure the affinity of compounds for the hERG channel. By comparison, the distinctive 2:1 atrial:ventricular arrhythmia observed in larval zebrafish can be used to identify compounds that block not only the ERG channel but also other cardiac ion channels. While this could be argued as advantageous, because the zebrafish assay can identify cardiotoxicity arising from the blockage of myriad ion channels, it raises problems when investigators attempt to compare their results with those from single target in vitro assays. Data indicating cardiotoxic effects in zebrafish from compounds that are apparently safe according to in vitro data sets might be interpreted as false positive results from the zebrafish; however, they may be caused by the compound acting on a target not represented in in vitro models.237

Moreover, zebrafish can regenerate heart muscle. Several research groups are working to discover factors involved in this process to help develop methods of repairing heart tissue in humans. One group revealed the role of TGFβ signaling in the regenerative capacity of the zebrafish heart after myocardial infarction using a cryoinjury procedure.238 Understanding the key healing processes after myocardial infarction in zebrafish may result in identification of the barriers to efficient cardiac regeneration in mammals and enable the design of novel therapeutic strategies for improved regeneration of the infarcted mammalian heart.

OTOTOXICITY

Drug-induced ototoxicity is often reversible but sometimes is not, leading to a severe impact on the quality of life for patients faced with no alternative treatment, as is with aminoglycosides for life-threatening infection and cisplatin for cancer.239 Screening for ototoxicity in mammalian preclinical models is very difficult, and given few regulatory requirements, this is rarely done.240 A zebrafish model for ototoxicity may fill this gap in preclinical safety testing. Zebrafish and human auditory systems most likely share an evolutionary origin with those of all vertebrates. This is indicated by homologous hair cell physiology,241,242 similar auditory anatomy, and neuronal signaling243 and is further supported by apparently conserved genetic regulation.244246 A large part of our understanding of the genetics of human hearing and balance has come from studies with zebrafish.247249

The zebrafish ear is composed of three semicircular canals arranged orthogonally to one another for sensing the direction and the speed of movement. It also contains two otoliths that help transmit vibrations to associated maculae (the utricular macula for sensing balance and the saccular macula for hearing) and a third macula (lagenar) that has both balance and hearing functions; these work together to stimulate anterior, posterior, and lateral cristae containing hair cells, which transduce signals to the brain.250252 This parallels the basic anatomy and physiology of the mammalian inner ear245,250,253 except that there is no cochlea in zebrafish. Zebrafish lack homologous structures for the mammalian outer ear (visible part), middle ear (ear drum and tympanic bones), and cochlea. The cochlea, in mammals, helps to amplify sounds.254 Sound amplification in zebrafish is accomplished via the Weberian apparatus, a series of bones which convey vibrations from the resonant and relatively large swim bladder to the lagenar macula in the ear.252

Sound is transmitted to the brain via mechanosensory hair cells.241 In zebrafish, these cells are either bathed in the endolymph of the ear or positioned along the lateral line system, which is a series of neuromasts (bundles of hair cells), located on, or near, the body surface in canals, for sensing water flow changes.255,256 Hair cells transduce sound (or water) waves into electrical nerve signals and transmit them to the brain through associated afferent nerve endings.242,257,258 The synapses between vertebrate hair cells and neurons are highly specialized to allow fast transduction of mechanical stimulation.259 Most hearing loss in humans (90%) stems from a loss of function during this transition and is referred to as sensorineural hearing loss.260 Zebrafish and human hair cells are homologous at the cellular and molecular levels and synapse in the same way with afferent neurons.257,258,261 Recent research has exposed a plethora of shared genetic involvement in hearing, comparing zebrafish to humans,244,246,248,262267 including proof-of-concept that gene knock-out studies in zebrafish can identify genes that cause nonsyndromic deafness in humans.249

Hair cell functionality is well-developed in zebrafish in advance of 5 dpf, as demonstrated by tests for auditory and somatosensory responses.252,268,269 The superficial location of hair cells in zebrafish allows for the ability to monitor hair cell changes in vivo in a high-throughput manner. This has made zebrafish a very popular model for ototoxicity interrogation and protection.247 Many drugs that are ototoxic to humans are ototoxic to developing, larval, and adult zebrafish; these include aminoglycoside antibiotics, carbonic anhydrase inhibitors, platinum-based chemotherapies, and environmental metal contaminants.247,270272 These data indicate that this model is a promising tool to screen novel chemical entities for ototoxicity.

Several screening platforms were recently tested and proven useful for detecting hair cell damage.273279 Using platforms such as these, many potentially protective cotreatments have been discovered in zebrafish that may mitigate human ototoxicity of useful, and yet unimproved, therapeutics. Some methods, through live imaging and forward genetic screens, can provide insight into the chemical and genetic mechanisms of ototoxicity as well as information about whether a protective cotreatment interferes with the therapy.280 The following are some examples of protective cotreatments discovered using this model: for radiotherapy for head and neck cancer, p38 inhibition;281 to mitigate cisplatin-induced ototoxiticy, quercetin,282 curcuminoids,283 CDK2 inhibition,284 or sirtuin 1 activation;285 for neomycin ototoxicity, melatonin,286 astaxanthin nanoemulsion;287 and recently discovered potential aminoglycoside-protective cotreatments.55,288 The latter work resulted in repurposing a preapproved drug into clinical trials for aminoglycoside protection, demonstrating the utility for zebrafish to help advance discoveries that circumvent human ototoxicity.

PINEAL/CIRCADIAN RHYTHM

Sleep disturbances are induced by many classes of pharmaceuticals, including beta-blockers, benzodiazepines, opioids, and amphetamines. Understanding the nature and severity of the sleep disturbance is necessary to assess the drug safety; for instance, reduced sleep is detrimental to human health and can contribute to diabetes, hormonal deficiencies, and neurological disorders. Impacts on sleep may be desirable for sleep aids and acceptable for severe indications such as cancer but unacceptable for drugs to treat less severe conditions; in any case, understanding effects on sleep early in the development process is important.

In zebrafish, sleep/wake behavior based on locomotor activity can be easily determined in a high-throughput manner.289 An advantage of the diurnal zebrafish over nocturnal rodent models is the greater similarity of sleep regulation to other diurnal vertebrates including humans. In both zebrafish and humans, melatonin production by the pineal organ (Figure 7) regulates the circadian regulation of sleep, downstream of the light-entrained circadian clock.290 The hypothalamic hypocretinergic system is also conserved between zebrafish and mammals.291 Clinically used hypnotics and stimulants have similar effects on the sleep/wake cycle in zebrafish as they do in humans, further demonstrating the translatability of effects on sleep/wake behavior,292 with occasional exceptions, e.g., dopamine D1 agonists, which increase rest in zebrafish but wakefulness in mammals.289

Figure 7.

Figure 7.

Photoreceptive pineal organ of a 4 dpf larval zebrafish, medially located in the forebrain (top of image = rostral). Nuclei are blue (DAPI), cone photoreceptors are red (antiarrestin 3a antibody), and rod photoreceptors are green (antirhodopsin antibody). Previously unpublished, provided by J. Gamse. Photo credit: Joshua Clanton.

Zebrafish larvae have been successfully used for identifying previously unappreciated effects of pharmaceuticals with known targets on sleep/wake behavior, indicating the utility of this system for identifying potential sleep-disturbing effects of new pharmaceuticals in preclinical screening.289 Moreover, using fine-grained analysis of locomotor behavior in combination with the statistical power available with zebrafish, relatively subtle effects on sleep/wake behavior can be teased out292 and then confirmed in other preclinical models such as cynomolgus monkeys with telemetered polysomnography.293

LATEST ADVANCES AND FUTURE DIRECTION

In general, zebrafish are a useful tool for pharmaceutical toxicity testing based on the evidence for translatable toxicology from zebrafish to mammals and augmented by the tractable nature of zebrafish for testing a wide spectrum of toxicities. New and innovative applications of zebrafish in relation to pharmaceutical toxicology appear regularly in the literature. These include studies interrogating toxic mechanisms, adverse outcome pathways (AOPs), drug abuse liability, endocrine disruption, as well as pertinent studies around metabolism, bioavailability, transcriptomics, and proteomics.

Mechanistic evidence can be used as a key to help escape toxicities that are encountered during new drug development. The mechanism through which toxicity is enacted can be driven through the therapeutic target (on-target toxicity) or another protein/molecule (off-target). Also, the precise molecular interaction between a xenobiotic and the therapeutic target may drive the desired therapeutic effect and a toxic effect based on the site and nature of the interaction. Knowledge about the toxic mechanism can help guide structure–activity relationship (SAR) studies and allow for faster discovery of less toxic drug candidates. The concept of using zebrafish for interrogating toxic mechanisms of environmental pollutants is not new,294,295 and in that arena, publications have steadily grown over the past 10 years to about 300 per year. Although the application of zebrafish in pharmaceutical toxicity mechanism studies296,297 has lagged, the numbers have recently begun to grow (Figure 8) to nearly 100 per year.

Figure 8.

Figure 8.

Number of publications reported from PubMed using search phrases “zebrafish (environmental or pharmaceutical) toxicity mechanism of action”.

The pharmaceutical mechanism studies have revealed cell types, pathways, and genes involved in cardiovascular,298,299 neuromuscular,300 neuronal,301303 ocular,85 auditory,55,281288 and embryo-development304307 toxicities. Those same studies introduce models by which safer compounds can be discovered by conducting SAR within or around a chemical series as has been lately shown for several chemotherapies.308311 Equally important, those models can be used to interrogate species-specific mechanisms of toxicities, which is of great value for predicting clinical toxicity. For example, promising immunomodulatory (IMiDS) chemotherapies, thalidomide, lenalidomide, and pomalidomide, all have teratogenic properties but with different potencies, depending on the species. Recent studies including knockdown and transgene expression in zebrafish, mice, and human stem cells312314 have shed light on the mechanisms and molecular players that make a developing embryo sensitive or insensitive to those drugs.

Recently, mechanistic studies using zebrafish have discovered personalized therapies for rare genetic diseases. Full exome next generation sequencing and refined gene editing tools combined with a rapidly developing tiny vertebrate with strong genetic homology to humans creates a proving ground for repurposing drugs to cure, or suppress symptoms of, life-threatening diseases.315 After patient-specific rare alleles are discovered, they are engineered into the genome of zebrafish, and if disease symptoms are recapitulated, a high-throughput platform is created for discovering personal therapies. Moreover, these models can deliver novel insight about mechanisms through which rare diseases are manifested and may lend themselves to our understanding of more common diseases.316 Rare diseases for which therapies have been discovered using zebrafish include pediatric diseases involving the following systems: musculoskeletal, blood, endocrine, neuronal, genitourinary, lymphatic, congenital malformations,52,317 and cancer.318 In this new application of zebrafish lies great potential for a positive impact on human health and medicine.

The impact of industrial waste on the environment is a growing concern keeping pace with the growing human population, and pharmaceutical waste is a major component of that pollution. Ecopharmacology is a new field that is concerned with the presence, persistence, and effects of parent drugs and metabolites in/on the environment.319 Pharmaceutical waste enters the environment through three ways; patient excretion, improper disposal, and manufacturing discharge. Pharmaceutical pollution is especially dangerous to life, because drugs are designed to be biologically active; therefore, all life forms are potentially at risk of being harmed or changed by this type of waste. Some ecopharmacological findings are microbial drug resistance, gene expression changes, epigenetic effects, genotoxicity, carcinogenicity, endocrine and immune disruption, and biofilm formation.

Environmental risk assessment is requested of all new drug products marketed for human consumption by the U.S. Food and Drug Administration and the European Medicines Agency. Those assessments compare the predicted environmental concentration to the acute toxic potency of the new drug on various species, which can include an algal species, an aquatic invertebrate, and fish. Zebrafish are being used regularly in those studies due to the ease of raising them in a laboratory environment.320 Increasing awareness of compounded chemical mixture effects on biological systems321323 and recent discoveries of effects of endocrine-disrupting chemicals in the environment324327 are heralding times for different and more detailed interrogation into potential environmental impact of new drugs.328 Recent zebrafish work that is pioneering the way toward such ends includes studies on transcriptomics,321,329331 chemical mixture effects,322,332 lipid metabolism,333,334 reproduction,335 and development.336

When evaluating drug safety, attention is frequently directed toward identifying cellular and organ toxicities, but there are important safety considerations that go well beyond toxicity. Prominent among these is the potential for drug abuse liability. As the current opioid abuse epidemic highlights, highly efficacious and relatively nontoxic medications can be unsafe if they exhibit potential for abuse. Testing drug candidates for abuse liability remains expensive and time-consuming because of the complexity of the rat and primate models used. Published reports offer some hope that zebrafish could be developed as a model for testing drug abuse liability, thereby enabling screening for abuse liability earlier, more efficiently, and on a larger scale. The earliest demonstration of zebrafish responsiveness to a drug of abuse was by Darland et al., who demonstrated that zebrafish conditioned place preference for cocaine almost 2 decades ago.337 Since then, several noncontingent (passive) behavioral assays have demonstrated zebrafish conditioning to opioids, amphetamines, alcohol, nicotine, and other drugs with abuse liability.338344 More recently, contingent models such as an opioid self-administration model have been developed for zebrafish.345 In this model, zebrafish can be conditioned in as few as 5 days to self-administer opioids, and their motivation to seek additional doses can be quantified. Because zebrafish perform so well in both noncontingent and contingent models of drug seeking, it may be possible in the future to use zebrafish to test new drugs for their abuse liability.

CONCLUSION

Zebrafish can be used to assess the toxicity of drug candidates in early screening assays, sometimes in a high-throughput manner. Due to their small size and transparency, such testing requires a small mass of test article, very little lab space, and data can be collected noninvasively over time in vivo. These data can help prioritize safer compounds for mammalian testing, disclose mechanisms of toxicity, and identify cotherapies that may mitigate toxicity of promising therapeutics. Employed to interrogate xenobiotics around which evidence points to conserved vertebrate biology, zebrafish toxicity assays can quickly and easily provide translatable data on a spectrum of tissues, organs, and systems.

Biographies

Steven Cassar is an Investigative Toxicologist for AbbVie Pharmaceutical Research and Development. For several years, studies with zebrafish at AbbVie have helped prioritize the development of less toxic compounds and have provided insights into mechanisms of toxicity. Steven received his BS in Biology from Alma College in Michigan and his MS in Mycology from Louisiana State University. Steven has over 20 years of experience in drug discovery and development, specifically in translational medicine, pharmacology, and genetics.

Isaac Adatto is an active participant of the zebrafish community. He joined the Zon lab at Boston Children’s Hospital in 2007. He has published his work on zebrafish husbandry in several journals, presented his research findings nationally and internationally, and has served as president of the Zebrafish Husbandry Association. Adatto currently works at Harvard’s Department of Stem Cell and Regenerative Biology as the Senior Zebrafish Research Manager supporting and assisting in research for multiple investigators. He holds an M.S. from the University of Massachusetts Intercampus Marine Science Program.

Dr. Jennifer L. Freeman is an Associate Professor of Toxicology in the School of Health Sciences, a faculty affiliate in Environmental and Ecological Engineering, and member of the Public Health Graduate Program at Purdue University. She received her doctorate in Environmental Toxicology and Molecular Cytogenetics from the University of Illinois at Urbana-Champaign and was a postdoctoral fellow at Harvard Medical School and Brigham and Women’s Hospital, Boston. The interests of the Freeman laboratory are to define the underlying genetic and epigenetic mechanisms of toxicity. All projects are currently utilizing the zebrafish vertebrate model system as a tool to investigate toxicity.

Joshua Gamse is a Senior Research Investigator in the Developmental and Reproductive Toxicology group within Drug Safety Evaluation at Bristol-Myers Squibb, where he has worked since 2014. Prior to Bristol-Myers Squibb, he was an Associate Professor in the Department of Biology at Vanderbilt University, where his laboratory’s research focused on development and function of the epithalamus in zebrafish.

Iñaki Iturria has a Bachelor’s degree in Pharmacy by the University of the Basque Country and a Master’s Degree in Biotechnology by the Autonomous University of Barcelona. He obtained his Ph.D. in 2016 by developing a model to evaluate the probiotic activity of lactic acid bacteria in zebrafish. He actually works as a Study Director at Biobide, a CRO specialized in toxicity and efficacy assays in zebrafish.

Christian Lawrence is currently the Manager of the Aquatic Resources Program (ARP) at Boston Children’s Hospital (BCH). ARP administers the zebrafish program at BCH, which is one of the largest and most active of its kind in the world. Mr. Lawrence also serves as a faculty member for the Health and Colony Management of Laboratory Fish course at the Mount Desert Island Biological Laboratory and was a Fulbright Specialist in Israel in 2014. He is coauthor of The Laboratory Zebrafish and has written a number of scientific publications on zebrafish biology and culture.

Arantza Muriana achieved her MPharm from Navarra University, with an MBA from San Pablo CEU University (Madrid, Spain) added to her scientist background, improving her project management and business skills. She began working in zebrafish after training in zebrafish biology at the Salk Institute (California, US). She has worked in clinical and preclinical CROs for 16 years, with more than 13 years of experience working in zebrafish with several papers, especially in the toxicology and neurodevelopmental areas. She has been part of several consortiums with regulatory agencies (EPA, FDA, NIH-NIEH, or OECD) for the validation and standardization of zebrafish as an alternative model.

Randall T. Peterson, Ph.D., is a chemical biologist seeking to discover new drug candidates for cardiovascular and nervous system disorders. The Peterson lab uses chemical screens in living zebrafish to identify novel compounds that modify organismal processes in vivo. Several of the compounds discovered by the Peterson laboratory have become widely used research tools or are in clinical development. After two decades as a student and faculty member at Harvard University, Randy has recently moved to the University of Utah, where he serves as dean of the College of Pharmacy and L. S. Skaggs Presidential Endowed Professor.

Steven Van Cruchten graduated as DVM at Ghent University in 1999 and obtained his Ph.D. at Ghent University in 2004. He then joined Johnson & Johnson in Belgium as a reproductive toxicologist and he also became involved into the field of juvenile toxicology. In 2008, he joined AstraZeneca in Sweden as a Toxicology Project Leader. He also kept his expert role in reproductive and juvenile toxicology. S. Van Cruchten returned to Belgium in 2011 to take up the position of associate professor at the University of Antwerp. His research focuses on alternative and animal models for juvenile and reproductive toxicology.

Dr. Zon is the Grousbeck Professor of Pediatric Medicine at Harvard Medical School, an Investigator at Howard Hughes Medical Institute, and the Director of the Stem Cell Program at Boston Children’s Hospital. He is internationally recognized for his pioneering work in stem cell biology and cancer genetics and has been the pre-eminent figure in establishing zebrafish as an invaluable genetic model for the study of the blood and hematopoietic development.

Footnotes

The authors declare the following competing financial interest(s): L.I.Z. is a founder and stockholder of Fate Therapeutics, CAMP4 Therapeutics, and Scholar Rock. He is a consultant for Celularity.

REFERENCES

  • (1).Blomme EA, and Will Y (2016) Toxicology Strategies for Drug Discovery: Present and Future. Chem. Res. Toxicol 29 (4), 473–504. [DOI] [PubMed] [Google Scholar]
  • (2).Hay M, Thomas DW, Craighead JL, Economides C, and Rosenthal J (2014) Clinical development success rates for investigational drugs. Nat. Biotechnol 32 (1), 40–51. [DOI] [PubMed] [Google Scholar]
  • (3).Munos B (2009) Lessons from 60 years of pharmaceutical innovation. Nat. Rev. Drug Discovery 8 (12), 959–68. [DOI] [PubMed] [Google Scholar]
  • (4).Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, and Pangalos MN (2014) Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework. Nat. Rev. Drug Discovery 13 (6), 419–31. [DOI] [PubMed] [Google Scholar]
  • (5).Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, Pairaudeau G, Pennie WD, Pickett SD, Wang J, Wallace O, and Weir A (2015) An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat. Rev. Drug Discovery 14 (7), 475–86. [DOI] [PubMed] [Google Scholar]
  • (6).Mangipudy R, Burkhardt J, and Kadambi VJ (2014) Use of animals for toxicology testing is necessary to ensure patient safety in pharmaceutical development. Regul. Toxicol. Pharmacol 70 (2), 439–41. [DOI] [PubMed] [Google Scholar]
  • (7).Monticello TM, Jones TW, Dambach DM, Potter DM, Bolt MW, Liu M, Keller DA, Hart TK, and Kadambi VJ (2017) Current nonclinical testing paradigm enables safe entry to First-In-Human clinical trials: The IQ consortium nonclinical to clinical translational database. Toxicol. Appl. Pharmacol 334, 100–109. [DOI] [PubMed] [Google Scholar]
  • (8).Beilmann M, Boonen H, Czich A, Dear G, Hewitt P, Mow T, Newham P, Oinonen T, Pognan F, Roth A, Valentin JP, Van Goethem F, Weaver RJ, Birk B, Boyer S, Caloni F, Chen AE, Corvi R, Cronin MTD, Daneshian M, Ewart LC, Fitzgerald RE, Hamilton GA, Hartung T, Kangas JD, Kramer NI, Leist M, Marx U, Polak S, Rovida C, Testai E, Van der Water B, Vulto P, and Steger-Hartmann T (2018) Optimizing drug discovery by Investigative Toxicology: Current and future trends. ALTEX 36 (2), 289–313. [DOI] [PubMed] [Google Scholar]
  • (9).Hornberg JJ, Laursen M, Brenden N, Persson M, Thougaard AV, Toft DB, and Mow T (2014) Exploratory toxicology as an integrated part of drug discovery. Part I: Why and how. Drug Discovery Today 19 (8), 1131–6. [DOI] [PubMed] [Google Scholar]
  • (10).Russell WM, and Burch RL (2009) Prefatory Note. ATLA, Altern. Lab. Anim 37 (3), 267–8. [DOI] [PubMed] [Google Scholar]
  • (11).Tannenbaum J, and Bennett BT (2015) Russell and Burch’s 3Rs then and now: the need for clarity in definition and purpose. J. Am. Assoc Lab Anim Sci 54 (2), 120–132. [PMC free article] [PubMed] [Google Scholar]
  • (12).Kim TW, Che JH, and Yun JW (2019) Use of stem cells as alternative methods to animal experimentation in predictive toxicology. Regul. Toxicol. Pharmacol 105, 15–29. [DOI] [PubMed] [Google Scholar]
  • (13).Stummann TC, and Bremer S (2012) Embryonic stem cells in safety pharmacology and toxicology. Adv. Exp. Med. Biol 745, 14–25. [DOI] [PubMed] [Google Scholar]
  • (14).Kuna L, Bozic I, Kizivat T, Bojanic K, Mrso M, Kralj E, Smolic R, Wu GY, and Smolic M (2018) Models of Drug Induced Liver Injury (DILI) - Current Issues and Future Perspectives. Curr. Drug Metab 19 (10), 830–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (15).Ma X, Liu J, Zhu W, Tang M, Lawrence N, Yu C, Gou M, and Chen S (2018) 3D bioprinting of functional tissue models for personalized drug screening and in vitro disease modeling. Adv. Drug Delivery Rev 132, 235–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (16).Zscheppang K, Berg J, Hedtrich S, Verheyen L, Wagner DE, Suttorp N, Hippenstiel S, and Hocke AC (2018) Human Pulmonary 3D Models For Translational Research. Biotechnol. J 13 (1), 1700341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (17).Weinhart M, Hocke A, Hippenstiel S, Kurreck J, and Hedtrich S (2019) 3D organ models-Revolution in pharmacological research? Pharmacol. Res 139, 446–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (18).Kar S, and Leszczynski J (2019) Exploration of Computational Approaches to Predict the Toxicity of Chemical Mixtures. Toxics 7 (1), 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (19).Wu Y, and Wang G (2018) Machine Learning Based Toxicity Prediction: From Chemical Structural Description to Transcriptome Analysis. Int. J. Mol. Sci 19 (8), 2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (20).Bissig KD, Han W, Barzi M, Kovalchuk N, Ding L, Fan X, Pankowicz FP, Zhang QY, and Ding X (2018) P450-Humanized and Human Liver Chimeric Mouse Models for Studying Xenobiotic Metabolism and Toxicity. Drug Metab. Dispos 46 (11), 1734–1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (21).Hunt PR (2017) The C. elegans model in toxicity testing. J. Appl. Toxicol 37 (1), 50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (22).Barros TP, Alderton WK, Reynolds HM, Roach AG, and Berghmans S (2008) Zebrafish: an emerging technology for in vivo pharmacological assessment to identify potential safety liabilities in early drug discovery. Br. J. Pharmacol 154 (7), 1400–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (23).Horzmann KA, and Freeman JL (2018) Making Waves: New Developments in Toxicology With the Zebrafish. Toxicol. Sci 163 (1), 5–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (24).Kanungo J, Cuevas E, Ali SF, and Paule MG (2014) Zebrafish model in drug safety assessment. Curr. Pharm. Des 20 (34), 5416–29. [DOI] [PubMed] [Google Scholar]
  • (25).McGrath P, and Li CQ (2008) Zebrafish: a predictive model for assessing drug-induced toxicity. Drug Discovery Today 13 (9–10), 394–401. [DOI] [PubMed] [Google Scholar]
  • (26).Peterson RT, and Macrae CA (2012) Systematic approaches to toxicology in the zebrafish. Annu. Rev. Pharmacol. Toxicol 52, 433–53. [DOI] [PubMed] [Google Scholar]
  • (27).Raldua D, and Pina B (2014) In vivo zebrafish assays for analyzing drug toxicity. Expert Opin. Drug Metab. Toxicol 10 (5), 685–97. [DOI] [PubMed] [Google Scholar]
  • (28).Rovida C, and Hartung T (2009) Re-evaluation of animal numbers and costs for in vivo tests to accomplish REACH legislation requirements for chemicals - a report by the transatlantic think tank for toxicology (t(4)). ALTEX 26 (3), 187–208. [PubMed] [Google Scholar]
  • (29).Walker ES, and Roberts RA (2018) Collaboration and competition: ethics in toxicology. Toxicol. Res. (Cambridge, U. K.) 7 (4), 576–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (30).Freires IA, Sardi JC, de Castro RD, and Rosalen PL (2017) Alternative Animal and Non-Animal Models for Drug Discovery and Development: Bonus or Burden? Pharm. Res 34 (4), 681–686. [DOI] [PubMed] [Google Scholar]
  • (31).MacArthur Clark J (2018) The 3Rs in research: a contemporary approach to replacement, reduction and refinement. Br. J. Nutr 120 (s1), S1–S7. [DOI] [PubMed] [Google Scholar]
  • (32).Russell WM (1995) The development of the three Rs concept. Altern Lab Anim 23 (3), 298–304. [PubMed] [Google Scholar]
  • (33).Strahle U, Scholz S, Geisler R, Greiner P, Hollert H, Rastegar S, Schumacher A, Selderslaghs I, Weiss C, Witters H, and Braunbeck T (2012) Zebrafish embryos as an alternative to animal experiments–a commentary on the definition of the onset of protected life stages in animal welfare regulations. Reprod. Toxicol 33 (2), 128–32. [DOI] [PubMed] [Google Scholar]
  • (34).Grech A, Tebby C, Brochot C, Bois FY, Bado-Nilles A, Dorne JL, Quignot N, and Beaudouin R (2019) Generic physiologically-based toxicokinetic modelling for fish: Integration of environmental factors and species variability. Sci. Total Environ 651, 516–531. [DOI] [PubMed] [Google Scholar]
  • (35).Villacrez M, Hellman K, Ono T, Sugihara Y, Rezeli M, Ek F, Marko-Varga G, and Olsson R (2018) Evaluation of Drug Exposure and Metabolism in Locust and Zebrafish Brains Using Mass Spectrometry Imaging. ACS Chem. Neurosci 9 (8), 1994–2000. [DOI] [PubMed] [Google Scholar]
  • (36).Gemberling M, Bailey TJ, Hyde DR, and Poss KD (2013) The zebrafish as a model for complex tissue regeneration. Trends Genet 29 (11), 611–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (37).Mokalled MH, and Poss KD (2018) A Regeneration Toolkit. Dev. Cell 47 (3), 267–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (38).Mehta AS, and Singh A (2019) Insights into regeneration tool box: An animal model approach. Dev. Biol 453, 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (39).Lawrence C (2007) The husbandry of zebrafish (Danio rerio): A review. Aquaculture 269 (1), 1–20. [Google Scholar]
  • (40).Lawrence C (2016) New frontiers for zebrafish management. Methods Cell Biol 135, 483–508. [DOI] [PubMed] [Google Scholar]
  • (41).Kilkenny C, Browne WJ, Cuthill IC, Emerson M, and Altman DG (2010) Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol 8 (6), No. e1000412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (42).Teytelman L, Stoliartchouk A, Kindler L, and Hurwitz BL (2016) Protocols.io: Virtual Communities for Protocol Development and Discussion. PLoS Biol 14 (8), No. e1002538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (43).Hsieh JH, Ryan K, Sedykh A, Lin JA, Shapiro AJ, Parham F, and Behl M (2019) Application of Benchmark Concentration (BMC) Analysis on Zebrafish Data: A New Perspective for Quantifying Toxicity in Alternative Animal Models. Toxicol. Sci 167 (1), 92–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (44).Varga ZM, Ekker SC, and Lawrence C (2018) Workshop Report: Zebrafish and Other Fish Models-Description of Extrinsic Environmental Factors for Rigorous Experiments and Reproducible Results. Zebrafish 15 (6), 533–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (45).MacRae CA, and Peterson RT (2015) Zebrafish as tools for drug discovery. Nat. Rev. Drug Discovery 14 (10), 721–31. [DOI] [PubMed] [Google Scholar]
  • (46).Santoriello C, and Zon LI (2012) Hooked! Modeling human disease in zebrafish. J. Clin. Invest 122 (7), 2337–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (47).van Wijk RC, Krekels EHJ, Hankemeier T, Spaink HP, and van der Graaf PH (2016) Systems pharmacology of hepatic metabolism in zebrafish larvae. Drug Discovery Today: Dis. Models 22, 27–34. [Google Scholar]
  • (48).Garcia GR, Noyes PD, and Tanguay RL (2016) Advancements in zebrafish applications for 21st century toxicology. Pharmacol. Ther 161, 11–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (49).Tamplin OJ, White RM, Jing L, Kaufman CK, Lacadie SA, Li P, Taylor AM, and Zon LI (2012) Small molecule screening in zebrafish: swimming in potential drug therapies. Wiley Interdiscip Rev. Dev Biol 1 (3), 459–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (50).Wiley DS, Redfield SE, and Zon LI (2017) Chemical screening in zebrafish for novel biological and therapeutic discovery. Methods Cell Biol 138, 651–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (51).Cutler C, Multani P, Robbins D, Kim HT, Le T, Hoggatt J, Pelus LM, Desponts C, Chen YB, Rezner B, Armand P, Koreth J, Glotzbecker B, Ho VT, Alyea E, Isom M, Kao G, Armant M, Silberstein L, Hu P, Soiffer RJ, Scadden DT, Ritz J, Goessling W, North TE, Mendlein J, Ballen K, Zon LI, Antin JH, and Shoemaker DD (2013) Prostaglandin-modulated umbilical cord blood hematopoietic stem cell transplantation. Blood 122 (17), 3074–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (52).Li D, March ME, Gutierrez-Uzquiza A, Kao C, Seiler C, Pinto E, Matsuoka LS, Battig MR, Bhoj EJ, Wenger TL, Tian L, Robinson N, Wang T, Liu Y, Weinstein BM, Swift M, Jung HM, Kaminski CN, Chiavacci R, Perkins JA, Levine MA, Sleiman PMA, Hicks PJ, Strausbaugh JT, Belasco JB, Dori Y, and Hakonarson H (2019) ARAF recurrent mutation causes central conducting lymphatic anomaly treatable with a MEK inhibitor. Nat. Med 25 (7), 1116–1122. [DOI] [PubMed] [Google Scholar]
  • (53).Mandelbaum J, Shestopalov IA, Henderson RE, Chau NG, Knoechel B, Wick MJ, and Zon LI (2018) Zebrafish blastomere screen identifies retinoic acid suppression of MYB in adenoid cystic carcinoma. J. Exp. Med 215 (10), 2673–2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (54).North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, Weber GJ, Bowman TV, Jang IH, Grosser T, Fitzgerald GA, Daley GQ, Orkin SH, and Zon LI (2007) Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 447 (7147), 1007–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (55).Ryals M, Morell RJ, Martin D, Boger ET, Wu P, Raible DW, and Cunningham LL (2018) The Inner Ear Heat Shock Transcriptional Signature Identifies Compounds That Protect Against Aminoglycoside Ototoxicity. Front. Cell. Neurosci 12, 445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (56).White RM, Cech J, Ratanasirintrawoot S, Lin CY, Rahl PB, Burke CJ, Langdon E, Tomlinson ML, Mosher J, Kaufman C, Chen F, Long HK, Kramer M, Datta S, Neuberg D, Granter S, Young RA, Morrison S, Wheeler GN, and Zon LI (2011) DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature 471 (7339), 518–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (57).Yu PB, Deng DY, Lai CS, Hong CC, Cuny GD, Bouxsein ML, Hong DW, McManus PM, Katagiri T, Sachidanandan C, Kamiya N, Fukuda T, Mishina Y, Peterson RT, and Bloch KD (2008) BMP type I receptor inhibition reduces heterotopic [corrected] ossification. Nat. Med 14 (12), 1363–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (58).Yu PB, Hong CC, Sachidanandan C, Babitt JL, Deng DY, Hoyng SA, Lin HY, Bloch KD, and Peterson RT (2008) Dorsomorphin inhibits BMP signals required for embryo-genesis and iron metabolism. Nat. Chem. Biol 4 (1), 33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (59).Howe K, Clark MD, Torroja CF, Torrance J, Berthelot C, Muffato M, Collins JE, Humphray S, McLaren K, Matthews L, McLaren S, Sealy I, Caccamo M, Churcher C, Scott C, Barrett JC, Koch R, Rauch GJ, White S, Chow W, Kilian B, Quintais LT, Guerra-Assuncao JA, Zhou Y, Gu Y, Yen J, Vogel JH, Eyre T, Redmond S, Banerjee R, Chi J, Fu B, Langley E, Maguire SF, Laird GK, Lloyd D, Kenyon E, Donaldson S, Sehra H, Almeida-King J, Loveland J, Trevanion S, Jones M, Quail M, Willey D, Hunt A, Burton J, Sims S, McLay K, Plumb B, Davis J, Clee C, Oliver K, Clark R, Riddle C, Elliot D, Threadgold G, Harden G, Ware D, Begum S, Mortimore B, Kerry G, Heath P, Phillimore B, Tracey A, Corby N, Dunn M, Johnson C, Wood J, Clark S, Pelan S, Griffiths G, Smith M, Glithero R, Howden P, Barker N, Lloyd C, Stevens C, Harley J, Holt K, Panagiotidis G, Lovell J, Beasley H, Henderson C, Gordon D, Auger K, Wright D, Collins J, Raisen C, Dyer L, Leung K, Robertson L, Ambridge K, Leongamornlert D, McGuire S, Gilderthorp R, Griffiths C, Manthravadi D, Nichol S, Barker G, Whitehead S, Kay M, Brown J, Murnane C, Gray E, Humphries M, Sycamore N, Barker D, Saunders D, Wallis J, Babbage A, Hammond S, Mashreghi-Mohammadi M, Barr L, Martin S, Wray P, Ellington A, Matthews N, Ellwood M, Woodmansey R, Clark G, Cooper J, Tromans A, Grafham D, Skuce C, Pandian R, Andrews R, Harrison E, Kimberley A, Garnett J, Fosker N, Hall R, Garner P, Kelly D, Bird C, Palmer S, Gehring I, Berger A, Dooley CM, Ersan-Urun Z, Eser C, Geiger H, Geisler M, Karotki L, Kirn A, Konantz J, Konantz M, Oberlander M, Rudolph-Geiger S, Teucke M, Lanz C, Raddatz G, Osoegawa K, Zhu B, Rapp A, Widaa S, Langford C, Yang F, Schuster SC, Carter NP, Harrow J, Ning Z, Herrero J, Searle SM, Enright A, Geisler R, Plasterk RH, Lee C, Westerfield M, de Jong PJ, Zon LI, Postlethwait JH, Nusslein-Volhard C, Hubbard TJ, Roest Crollius H, Rogers J, and Stemple DL (2013) The zebrafish reference genome sequence and its relationship to the human genome. Nature 496 (7446), 498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (60).Zon LI, and Peterson RT (2005) In vivo drug discovery in the zebrafish. Nat. Rev. Drug Discovery 4 (1), 35–44. [DOI] [PubMed] [Google Scholar]
  • (61).Kitano H (2002) Systems biology: a brief overview. Science 295 (5560), 1662–4. [DOI] [PubMed] [Google Scholar]
  • (62).van der Graaf PH, and Benson N (2011) Systems pharmacology: bridging systems biology and pharmacokineticspharmacodynamics (PKPD) in drug discovery and development. Pharm. Res 28 (7), 1460–4. [DOI] [PubMed] [Google Scholar]
  • (63).OECD. Test No. 236: Fish Embryo Acute Toxicity (FET) Test OECD Guidelines for the Testing of Chemicals, Section 2 OECD Publishing: Paris, France, 2013. [Google Scholar]
  • (64).International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use Detection of Toxicity to Reproduction for Human Pharmaceuticals S5(R3) https://www.fda.gov/media/108894/download (accessed July 31, 2019).
  • (65).International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use Final Concept Paper S5(R3): Detection of Toxicity to Reproduction for Human Pharmaceuticals https://admin.ich.org/sites/default/files/inline-files/S5-R3EWG_Step2_Guideline_2017_0705.pdf (accessed July 31, 2019).
  • (66).Gustafson AL, Stedman DB, Ball J, Hillegass JM, Flood A, Zhang CX, Panzica-Kelly J, Cao J, Coburn A, Enright BP, Tornesi MB, Hetheridge M, and Augustine-Rauch KA (2012) Inter-laboratory assessment of a harmonized zebrafish developmental toxicology assay - progress report on phase I. Reprod. Toxicol 33 (2), 155–64. [DOI] [PubMed] [Google Scholar]
  • (67).Brannen KC, Panzica-Kelly JM, Danberry TL, and Augustine-Rauch KA (2010) Development of a zebrafish embryo teratogenicity assay and quantitative prediction model. Birth Defects Res., Part B 89 (1), 66–77. [DOI] [PubMed] [Google Scholar]
  • (68).Ball JS, Stedman DB, Hillegass JM, Zhang CX, Panzica-Kelly J, Coburn A, Enright BP, Tornesi B, Amouzadeh HR, Hetheridge M, Gustafson AL, and Augustine-Rauch KA (2014) Fishing for teratogens: a consortium effort for a harmonized zebrafish developmental toxicology assay. Toxicol. Sci 139 (1), 210–9. [DOI] [PubMed] [Google Scholar]
  • (69).Lee SH, Kang JW, Lin T, Lee JE, and Jin DI (2013) Teratogenic potential of antiepileptic drugs in the zebrafish model. BioMed Res. Int 2013, 726478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (70).Pruvot B, Quiroz Y, Voncken A, Jeanray N, Piot A, Martial JA, and Muller M (2012) A panel of biological tests reveals developmental effects of pharmaceutical pollutants on late stage zebrafish embryos. Reprod. Toxicol 34 (4), 568–83. [DOI] [PubMed] [Google Scholar]
  • (71).Selderslaghs IW, Blust R, and Witters HE (2012) Feasibility study of the zebrafish assay as an alternative method to screen for developmental toxicity and embryotoxicity using a training set of 27 compounds. Reprod. Toxicol 33 (2), 142–54. [DOI] [PubMed] [Google Scholar]
  • (72).Teixido E, Pique E, Gomez-Catalan J, and Llobet JM (2013) Assessment of developmental delay in the zebrafish embryo teratogenicity assay. Toxicol. In Vitro 27 (1), 469–78. [DOI] [PubMed] [Google Scholar]
  • (73).Van den Bulck K, Hill A, Mesens N, Diekman H, De Schaepdrijver L, and Lammens L (2011) Zebrafish developmental toxicity assay: A fishy solution to reproductive toxicity screening, or just a red herring? Reprod. Toxicol 32 (2), 213–9. [DOI] [PubMed] [Google Scholar]
  • (74).Yamashita A, Inada H, Chihara K, Yamada T, Deguchi J, and Funabashi H (2014) Improvement of the evaluation method for teratogenicity using zebrafish embryos. J. Toxicol. Sci 39 (3), 453–64. [DOI] [PubMed] [Google Scholar]
  • (75).Cassar S, Beekhuijzen M, Beyer B, Chapin R, Dorau M, Hoberman A, Krupp E, Leconte I, Stedman D, Stethem C, van den Oetelaar D, and Tornesi B (2019) A multi-institutional study benchmarking the zebrafish developmental assay for prediction of embryotoxic plasma concentrations from rat embryo-fetal development studies. Reprod. Toxicol 86, 33–44. [DOI] [PubMed] [Google Scholar]
  • (76).Daston GP, Beyer BK, Carney EW, Chapin RE, Friedman JM, Piersma AH, Rogers JM, and Scialli AR (2014) Exposure-based validation list for developmental toxicity screening assays. Birth Defects Res., Part B 101 (6), 423–8. [DOI] [PubMed] [Google Scholar]
  • (77).Saad M, Matheeussen A, Bijttebier S, Verbueken E, Pype C, Casteleyn C, Van Ginneken C, Apers S, Maes L, Cos P, and Van Cruchten S (2017) In vitro CYP-mediated drug metabolism in the zebrafish (embryo) using human reference compounds. Toxicol. In Vitro 42, 329–336. [DOI] [PubMed] [Google Scholar]
  • (78).Verbueken E, Bars C, Ball JS, Periz-Stanacev J, Marei WFA, Tochwin A, Gabriels IJ, Michiels EDG, Stinckens E, Vergauwen L, Knapen D, Van Ginneken CJ, and Van Cruchten SJ (2018) From mRNA Expression of Drug Disposition Genes to In Vivo Assessment of CYP-Mediated Biotransformation during Zebrafish Embryonic and Larval Development. Int. J. Mol. Sci 19 (12), 3976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (79).Giusti A, Nguyen XB, Kislyuk S, Mignot M, Ranieri C, Nicolai J, Oorts M, Wu X, Annaert P, De Croze N, Leonard M, Ny A, Cabooter D, and de Witte P (2019) Safety Assessment of Compounds after In Vitro Metabolic Conversion Using Zebrafish Eleuthero Embryos. Int. J. Mol. Sci 20 (7), 1712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (80).Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, and Schilling TF (1995) Stages of embryonic development of the zebrafish. Dev. Dyn 203 (3), 253–310. [DOI] [PubMed] [Google Scholar]
  • (81).Makris SL, Solomon HM, Clark R, Shiota K, Barbellion S, Buschmann J, Ema M, Fujiwara M, Grote K, Hazelden KP, Hew KW, Horimoto M, Ooshima Y, Parkinson M, and Wise LD (2009) Terminology of developmental abnormalities in common laboratory mammals (Version 2). Birth Defects Res., Part B 86 (4), 227–327. [DOI] [PubMed] [Google Scholar]
  • (82).Babin PJ, Goizet C, and Raldua D (2014) Zebrafish models of human motor neuron diseases: advantages and limitations. Prog. Neurobiol 118, 36–58. [DOI] [PubMed] [Google Scholar]
  • (83).Bandmann O, and Burton EA (2010) Genetic zebrafish models of neurodegenerative diseases. Neurobiol. Dis 40 (1), 58–65. [DOI] [PubMed] [Google Scholar]
  • (84).Lee J, and Freeman JL (2014) Zebrafish as a model for investigating developmental lead (Pb) neurotoxicity as a risk factor in adult neurodegenerative disease: a mini-review. NeuroToxicology 43, 57–64. [DOI] [PubMed] [Google Scholar]
  • (85).Cassar S, Dunn C, Olson A, Buck W, Fossey S, Ramos MF, Sancheti P, Stolarik D, Britton H, Cole T, Bratcher N, Huang X, Peterson R, Longenecker K, and LeRoy B (2018) From the Cover: Inhibitors of Nicotinamide Phosphoribosyltransferase Cause Retinal Damage in Larval Zebrafish. Toxicol. Sci 161 (2), 300–309. [DOI] [PubMed] [Google Scholar]
  • (86).de Esch C, Slieker R, Wolterbeek A, Woutersen R, and de Groot D (2012) Zebrafish as potential model for developmental neurotoxicity testing: a mini review. Neurotoxicol. Teratol 34 (6), 545–53. [DOI] [PubMed] [Google Scholar]
  • (87).Quevedo C, Behl M, Ryan K, Paules RS, Alday A, Muriana A, and Alzualde A (2019) Detection and Prioritization of Developmentally Neurotoxic and/or Neurotoxic Compounds Using Zebrafish. Toxicol. Sci 168 (1), 225–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (88).Mueller T, and Wullimann MF (2016) Chapter 2 - Atlas of Cellular Markers in Zebrafish Neurogenesis: Atlas. Atlas of Early Zebrafish Brain Development, 27–157. [Google Scholar]
  • (89).Kalueff AV, Echevarria DJ, and Stewart AM (2014) Gaining translational momentum: more zebrafish models for neuroscience research. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 55, 1–6. [DOI] [PubMed] [Google Scholar]
  • (90).Kalueff AV, Stewart AM, and Gerlai R (2014) Zebrafish as an emerging model for studying complex brain disorders. Trends Pharmacol. Sci 35 (2), 63–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (91).Ijaz S, and Hoffman EJ (2016) Zebrafish: A Translational Model System for Studying Neuropsychiatric Disorders. J. Am. Acad. Child Adolesc Psychiatry 55 (9), 746–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (92).Basnet RM, Zizioli D, Taweedet S, Finazzi D, and Memo M (2019) Zebrafish Larvae as a Behavioral Model in Neuropharmacology. Biomedicines 7 (1), 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (93).Cassar S, Breidenbach L, Olson A, Huang X, Britton H, Woody C, Sancheti P, Stolarik D, Wicke K, Hempel K, and LeRoy B (2017) Measuring drug absorption improves interpretation of behavioral responses in a larval zebrafish locomotor assay for predicting seizure liability. J. Pharmacol. Toxicol. Methods 88, 56–63. [DOI] [PubMed] [Google Scholar]
  • (94).Horzmann KA, and Freeman JL (2016) Zebrafish Get Connected: Investigating Neurotransmission Targets and Alterations in Chemical Toxicity. Toxics 4 (3), 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (95).Panula P, Chen YC, Priyadarshini M, Kudo H, Semenova S, Sundvik M, and Sallinen V (2010) The comparative neuro-anatomy and neurochemistry of zebrafish CNS systems of relevance to human neuropsychiatric diseases. Neurobiol. Dis 40 (1), 46–57. [DOI] [PubMed] [Google Scholar]
  • (96).Rico EP, Rosemberg DB, Seibt KJ, Capiotti KM, Da Silva RS, and Bonan CD (2011) Zebrafish neurotransmitter systems as potential pharmacological and toxicological targets. Neurotoxicol. Teratol 33 (6), 608–17. [DOI] [PubMed] [Google Scholar]
  • (97).d’Amora M, and Giordani S (2018) The Utility of Zebrafish as a Model for Screening Developmental Neurotoxicity. Front. Neurosci 12, 976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (98).Brustein E, Saint-Amant L, Buss RR, Chong M, McDearmid JR, and Drapeau P (2003) Steps during the development of the zebrafish locomotor network. J. Physiol 97 (1), 77–86. [DOI] [PubMed] [Google Scholar]
  • (99).Colwill RM, and Creton R (2011) Imaging escape and avoidance behavior in zebrafish larvae. Rev. Neurosci 22 (1), 63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (100).Legradi J, el Abdellaoui N, van Pomeren M, and Legler J (2015) Comparability of behavioural assays using zebrafish larvae to assess neurotoxicity. Environ. Sci. Pollut. Res 22 (21), 16277–89. [DOI] [PubMed] [Google Scholar]
  • (101).Li F, Lin J, Liu X, Li W, Ding Y, Zhang Y, Zhou S, Guo N, and Li Q (2018) Characterization of the locomotor activities of zebrafish larvae under the influence of various neuroactive drugs. Ann. Transl Med 6 (10), 173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (102).Yang X, Lin J, Peng X, Zhang Q, Zhang Y, Guo N, Zhou S, and Li Q (2017) Effects of picrotoxin on zebrafish larvae behaviors: A comparison study with PTZ. Epilepsy Behav 70, 224–231. [DOI] [PubMed] [Google Scholar]
  • (103).Liu X, Lin J, Zhang Y, Peng X, Guo N, and Li Q (2016) Effects of diphenylhydantoin on locomotion and thigmotaxis of larval zebrafish. Neurotoxicol. Teratol 53, 41–7. [DOI] [PubMed] [Google Scholar]
  • (104).Alzualde A, Behl M, Sipes NS, Hsieh JH, Alday A, Tice RR, Paules RS, Muriana A, and Quevedo C (2018) Toxicity profiling of flame retardants in zebrafish embryos using a battery of assays for developmental toxicity, neurotoxicity, cardiotoxicity and hepatotoxicity toward human relevance. Neurotoxicol. Teratol 70, 40–50. [DOI] [PubMed] [Google Scholar]
  • (105).Bogi E, Belovicova K, Ujhazy E, Mach M, Koprdova R, Zilava L, Garafova A, Jezova D, and Dubovicky M (2018) Perinatal exposure to venlafaxine leads to lower anxiety and depression-like behavior in the adult rat offspring. Behav. Pharmacol 29 (5), 445–452. [DOI] [PubMed] [Google Scholar]
  • (106).Stanley KA, Curtis LR, Massey Simonich SL, and Tanguay RL (2009) Endosulfan I and endosulfan sulfate disrupts zebrafish embryonic development. Aquat. Toxicol 95 (4), 355–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (107).Stehr CM, Linbo TL, Incardona JP, and Scholz NL (2006) The developmental neurotoxicity of fipronil: notochord degeneration and locomotor defects in zebrafish embryos and larvae. Toxicol. Sci 92 (1), 270–8. [DOI] [PubMed] [Google Scholar]
  • (108).Ganzen L, Venkatraman P, Pang CP, Leung YF, and Zhang M (2017) Utilizing Zebrafish Visual Behaviors in Drug Screening for Retinal Degeneration. Int. J. Mol. Sci 18 (6), 1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (109).Zeddies DG, and Fay RR (2005) Development of the acoustically evoked behavioral response in zebrafish to pure tones. J. Exp. Biol 208 (7), 1363–1372. [DOI] [PubMed] [Google Scholar]
  • (110).Stewart AM, Braubach O, Spitsbergen J, Gerlai R, and Kalueff AV (2014) Zebrafish models for translational neuroscience research: from tank to bedside. Trends Neurosci 37 (5), 264–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (111).Maximino C, Marques de Brito T, Dias CA, Gouveia A Jr., and Morato S (2010) Scototaxis as anxiety-like behavior in fish. Nat. Protoc 5 (2), 209–16. [DOI] [PubMed] [Google Scholar]
  • (112).Fan CY, Cowden J, Simmons SO, Padilla S, and Ramabhadran R (2010) Gene expression changes in developing zebrafish as potential markers for rapid developmental neurotoxicity screening. Neurotoxicol. Teratol 32 (1), 91–8. [DOI] [PubMed] [Google Scholar]
  • (113).Lee J, Peterson SM, and Freeman JL (2016) Alzheimer’s disease risk genes in wild-type adult zebrafish exhibit gender-specific expression changes during aging. Neurogenetics 17 (3), 197–9. [DOI] [PubMed] [Google Scholar]
  • (114).Xi Y, Noble S, and Ekker M (2011) Modeling neurodegeneration in zebrafish. Curr. Neurol. Neurosci. Rep 11 (3), 274–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (115).Meshalkina DA, Kizlyk MN, Kysil EV, Collier AD, Echevarria DJ, Abreu MS, Barcellos LJG, Song C, Warnick JE, Kyzar EJ, and Kalueff AV (2018) Zebrafish models of autism spectrum disorder. Exp. Neurol 299, 207–216. [DOI] [PubMed] [Google Scholar]
  • (116).Marcon M, Mocelin R, de Oliveira DL, da Rosa Araujo AS, Herrmann AP, and Piato A (2019) Acetyl-L-carnitine as a putative candidate for the treatment of stress-related psychiatric disorders: Novel evidence from a zebrafish model. Neuropharmacology 150, 145–152. [DOI] [PubMed] [Google Scholar]
  • (117).Peng X, Lin J, Zhu Y, Liu X, Zhang Y, Ji Y, Yang X, Zhang Y, Guo N, and Li Q (2016) Anxiety-related behavioral responses of pentylenetetrazole-treated zebrafish larvae to light-dark transitions. Pharmacol., Biochem. Behav 145, 55–65. [DOI] [PubMed] [Google Scholar]
  • (118).Hu ZY, Chen B, Zhang JP, and Ma YY (2017) Up-regulation of autophagy-related gene 5 (ATG5) protects dopaminergic neurons in a zebrafish model of Parkinson’s disease. J. Biol. Chem 292 (44), 18062–18074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (119).Lee J, Peterson SM, and Freeman JL (2017) Sex-specific characterization and evaluation of the Alzheimer’s disease genetic risk factor sorl1 in zebrafish during aging and in the adult brain following a 100 ppb embryonic lead exposure. J. Appl. Toxicol 37 (4), 400–407. [DOI] [PubMed] [Google Scholar]
  • (120).Prabhudesai S, Bensabeur FZ, Abdullah R, Basak I, Baez S, Alves G, Holtzman NG, Larsen JP, and Møller SG (2016) LRRK2 knockdown in zebrafish causes developmental defects, neuronal loss, and synuclein aggregation. J. Neurosci. Res 94 (8), 717–735. [DOI] [PubMed] [Google Scholar]
  • (121).Pu YZ, Liang L, Fu AL, Liu Y, Sun L, Li Q, Wu D, Sun MJ, Zhang YG, and Zhao BQ (2017) Generation of Alzheimer’s Disease Transgenic Zebrafish Expressing Human APP Mutation Under Control of Zebrafish appb Promotor. Curr. Alzheimer Res 14 (6), 668–679. [DOI] [PubMed] [Google Scholar]
  • (122).Baxendale S, van Eeden F, and Wilkinson R (2017) The Power of Zebrafish in Personalised Medicine. Adv. Exp. Med. Biol 1007, 179. [DOI] [PubMed] [Google Scholar]
  • (123).Sanes JR, and Zipursky SL (2010) Design principles of insect and vertebrate visual systems. Neuron 66 (1), 15–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (124).Chhetri J, Jacobson G, and Gueven N (2014) Zebrafish– on the move towards ophthalmological research. Eye (London, U. K.) 28 (4), 367–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (125).Greiling TM, and Clark JI (2008) The transparent lens and cornea in the mouse and zebra fish eye. Semin. Cell Dev. Biol 19 (2), 94–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (126).Easter SS Jr., and Nicola GN (1996) The development of vision in the zebrafish (Danio rerio). Dev. Biol 180 (2), 646–663. [DOI] [PubMed] [Google Scholar]
  • (127).Avanesov A, and Malicki J (2010) Analysis of the retina in the zebrafish model. Methods Cell Biol 100, 153–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (128).Goldsmith P, and Harris WA (2003) The zebrafish as a tool for understanding the biology of visual disorders. Semin. Cell Dev. Biol 14 (1), 11–8. [DOI] [PubMed] [Google Scholar]
  • (129).Slijkerman RW, Song F, Astuti GD, Huynen MA, van Wijk E, Stieger K, and Collin RW (2015) The pros and cons of vertebrate animal models for functional and therapeutic research on inherited retinal dystrophies. Prog. Retinal Eye Res 48, 137–59. [DOI] [PubMed] [Google Scholar]
  • (130).Wan J, and Goldman D (2016) Retina regeneration in zebrafish. Curr. Opin. Genet. Dev 40, 41–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (131).Alvarez Y, Cederlund ML, Cottell DC, Bill BR, Ekker SC, Torres-Vazquez J, Weinstein BM, Hyde DR, Vihtelic TS, and Kennedy BN (2007) Genetic determinants of hyaloid and retinal vasculature in zebrafish. BMC Dev. Biol 7, 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (132).Berghmans S, Butler P, Goldsmith P, Waldron G, Gardner I, Golder Z, Richards FM, Kimber G, Roach A, Alderton W, and Fleming A (2008) Zebrafish based assays for the assessment of cardiac, visual and gut function–potential safety screens for early drug discovery. J. Pharmacol. Toxicol. Methods 58 (1), 59–68. [DOI] [PubMed] [Google Scholar]
  • (133).Deeti S, O’Farrell S, and Kennedy BN (2014) Early safety assessment of human oculotoxic drugs using the zebrafish visualmotor response. J. Pharmacol. Toxicol. Methods 69 (1), 1–8. [DOI] [PubMed] [Google Scholar]
  • (134).Richards FM, Alderton WK, Kimber GM, Liu Z, Strang I, Redfern WS, Valentin JP, Winter MJ, and Hutchinson TH (2008) Validation of the use of zebrafish larvae in visual safety assessment. J. Pharmacol. Toxicol. Methods 58 (1), 50–8. [DOI] [PubMed] [Google Scholar]
  • (135).Brockerhoff SE (2006) Measuring the optokinetic response of zebrafish larvae. Nat. Protoc 1 (5), 2448–51. [DOI] [PubMed] [Google Scholar]
  • (136).Fleisch VC, and Neuhauss SC (2006) Visual behavior in zebrafish. Zebrafish 3 (2), 191–201. [DOI] [PubMed] [Google Scholar]
  • (137).Emran F, Rihel J, and Dowling JE (2008) A behavioral assay to measure responsiveness of zebrafish to changes in light intensities. J. Visualized Exp. No 20, e923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (138).Nalbant P, Boehmer C, Dehmelt L, Wehner F, and Werner A (1999) Functional characterization of a Na+-phosphate cotransporter (NaPi-II) from zebrafish and identification of related transcripts. J. Physiol 520 (1), 79–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (139).Brugman S (2016) The zebrafish as a model to study intestinal inflammation. Dev. Comp. Immunol 64, 82–92. [DOI] [PubMed] [Google Scholar]
  • (140).Ganz J (2018) Gut feelings: Studying enteric nervous system development, function, and disease in the zebrafish model system. Dev. Dyn 247 (2), 268–278. [DOI] [PubMed] [Google Scholar]
  • (141).Ng ANY, de Jong-Curtain TA, Mawdsley DJ, White SJ, Shin J, Appel B, Dong PDS, Stainier DYR, and Heath JK (2005) Formation of the digestive system in zebrafish: III. Intestinal epithelium morphogenesis. Dev. Biol 286 (1), 114–135. [DOI] [PubMed] [Google Scholar]
  • (142).Zhao X, and Pack M (2017) Modeling intestinal disorders using zebrafish. Methods Cell Biol 138, 241–270. [DOI] [PubMed] [Google Scholar]
  • (143).Cassar S, Huang X, and Cole T (2015) A high-throughput method for predicting drug effects on gut transit time using larval zebrafish. J. Pharmacol. Toxicol. Methods 76, 72–5. [DOI] [PubMed] [Google Scholar]
  • (144).Rich A (2018) Improved Imaging of Zebrafish Motility. Neurogastroenterol. Motil 30 (9), No. e13435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (145).Kinkel MD, and Prince VE (2009) On the diabetic menu: zebrafish as a model for pancreas development and function. BioEssays 31 (2), 139–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (146).Zang L, Maddison LA, and Chen W (2018) Zebrafish as a Model for Obesity and Diabetes. Front. Cell Dev. Biol 6, 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (147).Ko JH, Nam YH, Joo SW, Kim HG, Lee YG, Kang TH, and Baek NI (2018) Flavonoid 8-O-Glucuronides from the Aerial Parts of Malva verticillata and Their Recovery Effects on Alloxan-Induced Pancreatic Islets in Zebrafish. Molecules 23 (4), 833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (148).Krishnan J, and Rohner N (2019) Sweet fish: Fish models for the study of hyperglycemia and diabetes. J. Diabetes 11 (3), 193–203. [DOI] [PubMed] [Google Scholar]
  • (149).Matsuda H (2018) Zebrafish as a model for studying functional pancreatic beta cells development and regeneration. Dev Growth Differ 60 (6), 393–399. [DOI] [PubMed] [Google Scholar]
  • (150).Prince VE, Anderson RM, and Dalgin G (2017) Zebrafish Pancreas Development and Regeneration: Fishing for Diabetes Therapies. Curr. Top. Dev. Biol 124, 235–276. [DOI] [PubMed] [Google Scholar]
  • (151).Jacobs HM, Sant KE, Basnet A, Williams LM, Moss JB, and Timme-Laragy AR (2018) Embryonic exposure to Mono(2-ethylhexyl) phthalate (MEHP) disrupts pancreatic organogenesis in zebrafish (Danio rerio). Chemosphere 195, 498–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (152).Sant KE, Jacobs HM, Borofski KA, Moss JB, and Timme-Laragy AR (2017) Embryonic exposures to perfluorooctanesulfonic acid (PFOS) disrupt pancreatic organogenesis in the zebrafish, Danio rerio. Environ. Pollut 220, 807–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (153).Pavlik L, Regev A, Ardayfio PA, and Chalasani NP (2019) Drug-Induced Steatosis and Steatohepatitis: The Search for Novel Serum Biomarkers Among Potential Biomarkers for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Drug Saf 42 (6), 701–711. [DOI] [PubMed] [Google Scholar]
  • (154).Real M, Barnhill MS, Higley C, Rosenberg J, and Lewis JH (2019) Drug-Induced Liver Injury: Highlights of the Recent Literature. Drug Saf 42 (3), 365–387. [DOI] [PubMed] [Google Scholar]
  • (155).Weaver RJ, and Valentin JP (2019) Today’s Challenges to De-Risk and Predict Drug Safety in Human ″Mind-the-Gap. Toxicol. Sci 167 (2), 307–321. [DOI] [PubMed] [Google Scholar]
  • (156).Goessling W, and Sadler KC (2015) Zebrafish: an important tool for liver disease research. Gastroenterology 149 (6), 1361–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (157).Wang S, Miller SR, Ober EA, and Sadler KC (2017) Making It New Again: Insight Into Liver Development, Regeneration, and Disease From Zebrafish Research. Curr. Top. Dev. Biol 124, 161–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (158).Driessen M, Vitins AP, Pennings JL, Kienhuis AS, Water B, and van der Ven LT (2015) A transcriptomics-based hepatotoxicity comparison between the zebrafish embryo and established human and rodent in vitro and in vivo models using cyclosporine A, amiodarone and acetaminophen. Toxicol. Lett 232 (2), 403–12. [DOI] [PubMed] [Google Scholar]
  • (159).de Souza Anselmo C, Sardela VF, de Sousa VP, and Pereira HMG (2018) Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of xenobiotics in humans? Comp. Biochem. Physiol., Part C: Toxicol. Pharmacol 212, 34–46. [DOI] [PubMed] [Google Scholar]
  • (160).Vliegenthart ADB, Tucker CS, Del Pozo J, and Dear JW (2014) Zebrafish as model organisms for studying drug-induced liver injury. Br. J. Clin. Pharmacol 78 (6), 1217–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (161).Tao T, and Peng J (2009) Liver development in zebrafish (Danio rerio). J. Genet. Genomics 36 (6), 325–334. [DOI] [PubMed] [Google Scholar]
  • (162).Lorent K, Yeo SY, Oda T, Chandrasekharappa S, Chitnis A, Matthews RP, and Pack M (2004) Inhibition of Jagged-mediated Notch signaling disrupts zebrafish biliary development and generates multi-organ defects compatible with an Alagille syndrome phenocopy. Development 131 (22), 5753–66. [DOI] [PubMed] [Google Scholar]
  • (163).Vernetti LA, Vogt A, Gough A, and Taylor DL (2017) Evolution of Experimental Models of the Liver to Predict Human Drug Hepatotoxicity and Efficacy. Clin Liver Dis 21 (1), 197–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (164).Poon KL, Wang X, Lee SG, Ng AS, Goh WH, Zhao Z, Al-Haddawi M, Wang H, Mathavan S, Ingham PW, McGinnis C, and Carney TJ (2017) Transgenic Zebrafish Reporter Lines as Alternative In Vivo Organ Toxicity Models. Toxicol. Sci 156 (1), 133–148. [DOI] [PubMed] [Google Scholar]
  • (165).Zhang Y, Han L, He Q, Chen W, Sun C, Wang X, Chen X, Wang R, Hsiao CD, and Liu K (2017) A rapid assessment for predicting drug-induced hepatotoxicity using zebrafish. J. Pharmacol. Toxicol. Methods 84, 102–110. [DOI] [PubMed] [Google Scholar]
  • (166).Cornet C, Calzolari S, Minana-Prieto R, Dyballa S, van Doornmalen E, Rutjes H, Savy T, D’Amico D, and Terriente J (2017) ZeGlobalTox: An Innovative Approach to Address Organ Drug Toxicity Using Zebrafish. Int. J. Mol. Sci 18 (4), 864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (167).Hill A, Mesens N, Steemans M, Xu JJ, and Aleo MD (2012) Comparisons between in vitro whole cell imaging and in vivo zebrafish-based approaches for identifying potential human hepatotoxicants earlier in pharmaceutical development. Drug Metab. Rev 44 (1), 127–40. [DOI] [PubMed] [Google Scholar]
  • (168).He JH, Guo SY, Zhu F, Zhu JJ, Chen YX, Huang CJ, Gao JM, Dong QX, Xuan YX, and Li CQ (2013) A zebrafish phenotypic assay for assessing drug-induced hepatotoxicity. J. Pharmacol. Toxicol. Methods 67 (1), 25–32. [DOI] [PubMed] [Google Scholar]
  • (169).Mesens N, Crawford AD, Menke A, Hung PD, Van Goethem F, Nuyts R, Hansen E, Wolterbeek A, Van Gompel J, De Witte P, and Esguerra CV (2015) Are zebrafish larvae suitable for assessing the hepatotoxicity potential of drug candidates? J. Appl. Toxicol 35 (9), 1017–29. [DOI] [PubMed] [Google Scholar]
  • (170).Quan Y, Gong L, He J, Zhou Y, Liu M, Cao Z, Li Y, and Peng C (2019) Aloe emodin induces hepatotoxicity by activating NF-kappaB inflammatory pathway and P53 apoptosis pathway in zebrafish. Toxicol. Lett 306, 66–79. [DOI] [PubMed] [Google Scholar]
  • (171).Zhong R, Chen Y, Ling J, Xia Z, Zhan Y, Sun E, Shi Z, Feng L, Jia X, Song J, and Wei Y (2019) The Toxicity and Metabolism Properties of Herba Epimedii Flavonoids on Laval and Adult Zebrafish. Evid Based Complement Alternat Med 2019, 3745051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (172).Zhou LL, Chen HJ, He QQ, Li C, Wei LX, and Shang J (2019) Evaluation of hepatotoxicity potential of a potent traditional Tibetan medicine Zuotai. J. Ethnopharmacol 234, 112–118. [DOI] [PubMed] [Google Scholar]
  • (173).Chen Q, Jia A, Snyder SA, Gong Z, and Lam SH (2016) Glucocorticoid activity detected by in vivo zebrafish assay and in vitro glucocorticoid receptor bioassay at environmental relevant concentrations. Chemosphere 144, 1162–9. [DOI] [PubMed] [Google Scholar]
  • (174).Gomes TB, Fernandes Sales Junior S, Saint’Pierre TD, Correia FV, Hauser-Davis RA, and Saggioro EM (2019) Sublethal psychotropic pharmaceutical effects on the model organism Danio rerio: Oxidative stress and metal dishomeostasis. Ecotoxicol. Environ. Saf 171, 781–789. [DOI] [PubMed] [Google Scholar]
  • (175).Pohl J, Bjorlenius B, Brodin T, Carlsson G, Fick J, Larsson DGJ, Norrgren L, and Orn S (2018) Effects of ozonated sewage effluent on reproduction and behavioral endpoints in zebrafish (Danio rerio). Aquat. Toxicol 200, 93–101. [DOI] [PubMed] [Google Scholar]
  • (176).Li C, Li P, Tan YM, Lam SH, Chan EC, and Gong Z (2016) Metabolomic Characterizations of Liver Injury Caused by Acute Arsenic Toxicity in Zebrafish. PLoS One 11 (3), No. e0151225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (177).Plhalova L, Stepanova S, Blahova J, Praskova E, Hostovsky M, Skoric M, Zelnickova L, Svobodova Z, and Bedanova I (2012) The effects of subchronic exposure to terbuthylazine on zebrafish. Neuro Endocrinol Lett 33 (Suppl 3), 113–119. [PubMed] [Google Scholar]
  • (178).Simmons DBD, Cowie AM, Koh J, Sherry JP, and Martyniuk CJ (2019) Label-free and iTRAQ proteomics analysis in the liver of zebrafish (Danio rerio) following dietary exposure to the organochlorine pesticide dieldrin. J. Proteomics 202, 103362. [DOI] [PubMed] [Google Scholar]
  • (179).Huang Q, Li S, Yin H, Wang C, Lee SMY, and Wang R (2018) Alleviating the hepatotoxicity of trazodone via supra-molecular encapsulation. Food Chem. Toxicol 112, 421–426. [DOI] [PubMed] [Google Scholar]
  • (180).Calienni MN, Cagel M, Montanari J, Moretton MA, Prieto MJ, Chiappetta DA, and Alonso SDV (2018) Zebrafish (Danio rerio) model as an early stage screening tool to study the biodistribution and toxicity profile of doxorubicin-loaded mixed micelles. Toxicol. Appl. Pharmacol 357, 106–114. [DOI] [PubMed] [Google Scholar]
  • (181).Lin HS, Huang YL, Wang YS, Hsiao E, Hsu TA, Shiao HY, Jiaang WT, Sampurna BP, Lin KH, Wu MS, Lai GM, and Yuh CH (2019) Identification of Novel Anti-Liver Cancer Small Molecules with Better Therapeutic Index than Sorafenib via Zebrafish Drug Screening Platform. Cancers 11 (6), 739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (182).Sunke R, Bankala R, Thirupataiah B, Ramarao E, Kumar JS, Doss HM, Medishetti R, Kulkarni P, Kapavarapu RK, Rasool M, Mudgal J, Mathew JE, Shenoy GG, Parsa KVL, and Pal M (2019) InCl3 mediated heteroarylation of indoles and their derivatization via CH activation strategy: Discovery of 2-(1H-indol-3-yl)-quinoxaline derivatives as a new class of PDE4B selective inhibitors for arthritis and/or multiple sclerosis. Eur. J. Med. Chem 174, 198–215. [DOI] [PubMed] [Google Scholar]
  • (183).Carten JD, and Farber SA (2009) A new model system swims into focus: using the zebrafish to visualize intestinal metabolism in vivo. Clin Lipidol 4 (4), 501–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (184).Quinlivan VH, and Farber SA (2017) Lipid Uptake, Metabolism, and Transport in the Larval Zebrafish. Front. Endocrinol 8, 319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (185).Davenport M (2016) Biliary atresia: From Australia to the zebrafish. J. Pediatr Surg 51 (2), 200–5. [DOI] [PubMed] [Google Scholar]
  • (186).Koo KA, Lorent K, Gong W, Windsor P, Whittaker SJ, Pack M, Wells RG, and Porter JR (2015) Biliatresone, a Reactive Natural Toxin from Dysphania glomulifera and D. littoralis: Discovery of the Toxic Moiety 1,2-Diaryl-2-Propenone. Chem. Res. Toxicol 28 (8), 1519–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (187).Ningappa M, So J, Glessner J, Ashokkumar C, Ranganathan S, Min J, Higgs BW, Sun Q, Haberman K, Schmitt L, Vilarinho S, Mistry PK, Vockley G, Dhawan A, Gittes GK, Hakonarson H, Jaffe R, Subramaniam S, Shin D, and Sindhi R (2015) The Role of ARF6 in Biliary Atresia. PLoS One 10 (9), No. e0138381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (188).Zhao X, Lorent K, Wilkins BJ, Marchione DM, Gillespie K, Waisbourd-Zinman O, So J, Koo KA, Shin D, Porter JR, Wells RG, Blair I, and Pack M (2016) Glutathione antioxidant pathway activity and reserve determine toxicity and specificity of the biliary toxin biliatresone in zebrafish. Hepatology 64 (3), 894–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (189).Fuchs TC, and Hewitt P (2011) Biomarkers for drug-induced renal damage and nephrotoxicity-an overview for applied toxicology. AAPS J 13 (4), 615–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (190).Elmonem MA, Berlingerio SP, van den Heuvel LP, de Witte PA, Lowe M, and Levtchenko EN (2018) Genetic Renal Diseases: The Emerging Role of Zebrafish Models. Cells 7 (9), 130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (191).Gehrig J, Pandey G, and Westhoff JH (2018) Zebrafish as a Model for Drug Screening in Genetic Kidney Diseases. Front Pediatr 6, 183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (192).Gorgulho R, Jacinto R, Lopes SS, Pereira SA, Tranfield EM, Martins GG, Gualda EJ, Derks RJE, Correia AC, Steenvoorden E, Pintado P, Mayboroda OA, Monteiro EC, and Morello J (2018) Usefulness of zebrafish larvae to evaluate drug-induced functional and morphological renal tubular alterations. Arch. Toxicol 92 (1), 411–423. [DOI] [PubMed] [Google Scholar]
  • (193).Schenk H, Muller-Deile J, Kinast M, and Schiffer M (2017) Disease modeling in genetic kidney diseases: zebrafish. Cell Tissue Res 369 (1), 127–141. [DOI] [PubMed] [Google Scholar]
  • (194).Kramer-Zucker AG, Wiessner S, Jensen AM, and Drummond IA (2005) Organization of the pronephric filtration apparatus in zebrafish requires Nephrin, Podocin and the FERM domain protein Mosaic eyes. Dev. Biol 285 (2), 316–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (195).Hentschel DM, Mengel M, Boehme L, Liebsch F, Albertin C, Bonventre JV, Haller H, and Schiffer M (2007) Rapid screening of glomerular slit diaphragm integrity in larval zebrafish. Am. J. Physiol Renal Physiol 293 (5), F1746–50. [DOI] [PubMed] [Google Scholar]
  • (196).Rider SA, Bruton FA, Collins RG, Conway BR, and Mullins JJ (2018) The Efficacy of Puromycin and Adriamycin for Induction of Glomerular Failure in Larval Zebrafish Validated by an Assay of Glomerular Permeability Dynamics. Zebrafish 15 (3), 234–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (197).Morello J, Derks RJE, Lopes SS, Steenvoorden E, Monteiro EC, Mayboroda OA, and Pereira SA (2018) Zebrafish Larvae Are a Suitable Model to Investigate the Metabolic Phenotype of Drug-Induced Renal Tubular Injury. Front. Pharmacol 9, 1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (198).Hukriede N, Vogt A, and de Caestecker M (2017) Drug Discovery to Halt the Progression of Acute Kidney Injury to Chronic Kidney Disease: A Case for Phenotypic Drug Discovery in Acute Kidney Injury. Nephron 137 (4), 268–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (199).Biran J, Tahor M, Wircer E, and Levkowitz G (2015) Role of developmental factors in hypothalamic function. Front. Neuroanat 9, 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (200).Bruggemann M, Licht O, Fetter E, Teigeler M, Schafers C, and Eilebrecht E (2018) Knotting nets: Molecular junctions of interconnecting endocrine axes identified by application of the adverse outcome pathway concept. Environ. Toxicol. Chem 37 (2), 318–328. [DOI] [PubMed] [Google Scholar]
  • (201).Lohr H, and Hammerschmidt M (2011) Zebrafish in endocrine systems: recent advances and implications for human disease. Annu. Rev. Physiol 73, 183–211. [DOI] [PubMed] [Google Scholar]
  • (202).Zohar Y, Munoz-Cueto JA, Elizur A, and Kah O (2010) Neuroendocrinology of reproduction in teleost fish. Gen. Comp. Endocrinol 165 (3), 438–55. [DOI] [PubMed] [Google Scholar]
  • (203).Chai C, and Chan WK (2000) Developmental expression of a novel Ftz-F1 homologue, ff1b (NR5A4), in the zebrafish Danio rerio. Mech. Dev 91 (1–2), 421–6. [DOI] [PubMed] [Google Scholar]
  • (204).von Hofsten J, Jones I, Karlsson J, and Olsson PE (2001) Developmental expression patterns of FTZ-F1 homologues in zebrafish (Danio rerio). Gen. Comp. Endocrinol 121 (2), 146–55. [DOI] [PubMed] [Google Scholar]
  • (205).Hsu HJ, Lin G, and Chung BC (2004) Parallel early development of zebrafish interrenal glands and pronephros: differential control by wt1 and ff1b. Endocr. Res 30 (4), 803. [DOI] [PubMed] [Google Scholar]
  • (206).Herzog W, Zeng X, Lele Z, Sonntag C, Ting JW, Chang CY, and Hammerschmidt M (2003) Adenohypophysis formation in the zebrafish and its dependence on sonic hedgehog. Dev. Biol 254 (1), 36–49. [DOI] [PubMed] [Google Scholar]
  • (207).Mouriec K, Lareyre JJ, Tong SK, Le Page Y, Vaillant C, Pellegrini E, Pakdel F, Chung BC, Kah O, and Anglade I (2009) Early regulation of brain aromatase (cyp19a1b) by estrogen receptors during zebrafish development. Dev. Dyn 238 (10), 2641–51. [DOI] [PubMed] [Google Scholar]
  • (208).Lassiter CS, and Linney E (2007) Embryonic expression and steroid regulation of brain aromatase cyp19a1b in zebrafish (Danio rerio). Zebrafish 4 (1), 49–57. [DOI] [PubMed] [Google Scholar]
  • (209).To TT, Hahner S, Nica G, Rohr KB, Hammerschmidt M, Winkler C, and Allolio B (2007) Pituitary-interrenal interaction in zebrafish interrenal organ development. Mol. Endocrinol 21 (2), 472–85. [DOI] [PubMed] [Google Scholar]
  • (210).Porazzi P, Calebiro D, Benato F, Tiso N, and Persani L (2009) Thyroid gland development and function in the zebrafish model. Mol. Cell. Endocrinol 312 (1–2), 14–23. [DOI] [PubMed] [Google Scholar]
  • (211).Scholz S, Renner P, Belanger SE, Busquet F, Davi R, Demeneix BA, Denny JS, Leonard M, McMaster ME, Villeneuve DL, and Embry MR (2013) Alternatives to in vivo tests to detect endocrine disrupting chemicals (EDCs) in fish and amphibians–screening for estrogen, androgen and thyroid hormone disruption. Crit. Rev. Toxicol 43 (1), 45–72. [DOI] [PubMed] [Google Scholar]
  • (212).Orn S, Yamani S, and Norrgren L (2006) Comparison of vitellogenin induction, sex ratio, and gonad morphology between zebrafish and Japanese medaka after exposure to 17alpha- ethinylestradiol and 17beta-trenbolone. Arch. Environ. Contam. Toxicol 51 (2), 237–43. [DOI] [PubMed] [Google Scholar]
  • (213).Chen X, Walter KM, Miller GW, Lein PJ, and Puschner B (2018) Simultaneous quantification of T4, T3, rT3, 3,5-T2 and 3,3′-T2 in larval zebrafish (Danio rerio) as a model to study exposure to polychlorinated biphenyls. Biomed. Chromatogr 32 (6), No. e4185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (214).Yu LQ, Zhao GF, Feng M, Wen W, Li K, Zhang PW, Peng X, Huo WJ, and Zhou HD (2014) Chronic exposure to pentachlorophenol alters thyroid hormones and thyroid hormone pathway mRNAs in zebrafish. Environ. Toxicol. Chem 33 (1), 170–6. [DOI] [PubMed] [Google Scholar]
  • (215).Nyakubaya VT, Durney BC, Ellington MC, Kantes AD, Reed PA, Walter SE, Stueckle JR, and Holland LA (2015) Quantification of circulating steroids in individual zebrafish using stacking to achieve nanomolar detection limits with capillary electrophoresis and UV-visible absorbance detection. Anal. Bioanal. Chem 407 (23), 6985–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (216).Muth-Kohne E, Westphal-Settele K, Bruckner J, Konradi S, Schiller V, Schafers C, Teigeler M, and Fenske M (2016) Linking the response of endocrine regulated genes to adverse effects on sex differentiation improves comprehension of aromatase inhibition in a Fish Sexual Development Test. Aquat. Toxicol 176, 116–27. [DOI] [PubMed] [Google Scholar]
  • (217).Fetter E, Smetanova S, Baldauf L, Lidzba A, Altenburger R, Schuttler A, and Scholz S (2015) Identification and Characterization of Androgen-Responsive Genes in Zebrafish Embryos. Environ. Sci. Technol 49 (19), 11789–98. [DOI] [PubMed] [Google Scholar]
  • (218).Shi X, Liu C, Wu G, and Zhou B (2009) Waterborne exposure to PFOS causes disruption of the hypothalamus-pituitary-thyroid axis in zebrafish larvae. Chemosphere 77 (7), 1010–8. [DOI] [PubMed] [Google Scholar]
  • (219).Yu L, Deng J, Shi X, Liu C, Yu K, and Zhou B (2010) Exposure to DE-71 alters thyroid hormone levels and gene transcription in the hypothalamic-pituitary-thyroid axis of zebrafish larvae. Aquat. Toxicol 97 (3), 226–33. [DOI] [PubMed] [Google Scholar]
  • (220).Yu L, Chen M, Liu Y, Gui W, and Zhu G (2013) Thyroid endocrine disruption in zebrafish larvae following exposure to hexaconazole and tebuconazole. Aquat. Toxicol 138–139, 35–42. [DOI] [PubMed] [Google Scholar]
  • (221).Crofton KM (2008) Thyroid disrupting chemicals: mechanisms and mixtures. Int. J. Androl 31 (2), 209–23. [DOI] [PubMed] [Google Scholar]
  • (222).Boas M, Feldt-Rasmussen U, and Main KM (2012) Thyroid effects of endocrine disrupting chemicals. Mol. Cell. Endocrinol 355 (2), 240–8. [DOI] [PubMed] [Google Scholar]
  • (223).Brion F, Le Page Y, Piccini B, Cardoso O, Tong SK, Chung BC, and Kah O (2012) Screening estrogenic activities of chemicals or mixtures in vivo using transgenic (cyp19a1b-GFP) zebrafish embryos. PLoS One 7 (5), No. e36069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (224).Brion F, De Gussem V, Buchinger S, Hollert H, Carere M, Porcher JM, Piccini B, Feray C, Dulio V, Konemann S, Simon E, Werner I, Kase R, and Ait-Aissa S (2019) Monitoring estrogenic activities of waste and surface waters using a novel in vivo zebrafish embryonic (EASZY) assay: Comparison with in vitro cell-based assays and determination of effect-based trigger values. Environ. Int 130, 104896. [DOI] [PubMed] [Google Scholar]
  • (225).Green JM, Metz J, Lee O, Trznadel M, Takesono A, Brown AR, Owen SF, Kudoh T, and Tyler CR (2016) High-Content and Semi-Automated Quantification of Responses to Estrogenic Chemicals Using a Novel Translucent Transgenic Zebrafish. Environ. Sci. Technol 50 (12), 6536–45. [DOI] [PubMed] [Google Scholar]
  • (226).Ji C, Jin X, He J, and Yin Z (2012) Use of TSHbeta:EGFP transgenic zebrafish as a rapid in vivo model for assessing thyroid-disrupting chemicals. Toxicol. Appl. Pharmacol 262 (2), 149–55. [DOI] [PubMed] [Google Scholar]
  • (227).Opitz R, Maquet E, Huisken J, Antonica F, Trubiroha A, Pottier G, Janssens V, and Costagliola S (2012) Transgenic zebrafish illuminate the dynamics of thyroid morphogenesis and its relationship to cardiovascular development. Dev. Biol 372 (2), 203–16. [DOI] [PubMed] [Google Scholar]
  • (228).Benato F, Colletti E, Skobo T, Moro E, Colombo L, Argenton F, and Dalla Valle L (2014) A living biosensor model to dynamically trace glucocorticoid transcriptional activity during development and adult life in zebrafish. Mol. Cell. Endocrinol 392 (1–2), 60–72. [DOI] [PubMed] [Google Scholar]
  • (229).Choudhuri A, Fast EM, and Zon LI (2017) Using Zebrafish to Study Pathways that Regulate Hematopoietic Stem Cell Self-Renewal and Migration. Stem Cell Rep 8 (6), 1465–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (230).Ransom DG, Haffter P, Odenthal J, Brownlie A, Vogelsang E, Kelsh RN, Brand M, van Eeden FJ, Furutani-Seiki M, Granato M, Hammerschmidt M, Heisenberg CP, Jiang YJ, Kane DA, Mullins MC, and Nusslein-Volhard C (1996) Characterization of zebrafish mutants with defects in embryonic hematopoiesis. Development 123, 311–319. [DOI] [PubMed] [Google Scholar]
  • (231).Boatman S, Barrett F, Satishchandran S, Jing L, Shestopalov I, and Zon LI (2013) Assaying hematopoiesis using zebrafish. Blood Cells, Mol., Dis 51 (4), 271–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (232).Lenard A, Alghisi E, Daff H, Donzelli M, McGinnis C, and Lengerke C (2016) Using zebrafish to model erythroid lineage toxicity and regeneration. Haematologica 101 (5), e164–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (233).MacRae CA, and Fishman MC (2002) Zebrafish: the complete cardiovascular compendium. Cold Spring Harbor Symp. Quant. Biol 67, 301–7. [DOI] [PubMed] [Google Scholar]
  • (234).Santoro MM, Beltrame M, Panakova D, Siekmann AF, Tiso N, Venero Galanternik M, Jung HM, and Weinstein BM (2019) Advantages and Challenges of Cardiovascular and Lymphatic Studies in Zebrafish Research. Front. Cell Dev. Biol 7, 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (235).Milan DJ, Peterson TA, Ruskin JN, Peterson RT, and MacRae CA (2003) Drugs that induce repolarization abnormalities cause bradycardia in zebrafish. Circulation 107 (10), 1355–8. [DOI] [PubMed] [Google Scholar]
  • (236).Letamendia A, Quevedo C, Ibarbia I, Virto JM, Holgado O, Diez M, Izpisua Belmonte JC, and Callol-Massot C (2012) Development and validation of an automated high-throughput system for zebrafish in vivo screenings. PLoS One 7 (5), No. e36690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (237).Fleming A, Diekmann H, and Goldsmith P (2013) Functional characterisation of the maturation of the blood-brain barrier in larval zebrafish. PLoS One 8 (10), No. e77548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (238).Chablais F, and Jazwinska A (2012) The regenerative capacity of the zebrafish heart is dependent on TGFbeta signaling. Development 139 (11), 1921–30. [DOI] [PubMed] [Google Scholar]
  • (239).Lanvers-Kaminsky C, Zehnhoff-Dinnesen AA, Parfitt R, and Ciarimboli G (2017) Drug-induced ototoxicity: Mechanisms, Pharmacogenetics, and protective strategies. Clin. Pharmacol. Ther 101 (4), 491–500. [DOI] [PubMed] [Google Scholar]
  • (240).Gauvin DV, Yoder J, Tapp RL, and Cole PI (2016) Defining “Best Practices” For Critical Endpoints In Preclinical Screening of New Chemical Entities For Ototoxicity Liability. Otolaryngology - Open Journal 2 (2), 58–69. [Google Scholar]
  • (241).McPherson DR (2018) Sensory Hair Cells: An Introduction to Structure and Physiology. Integr. Comp. Biol 58 (2), 282–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (242).Pickett SB, and Raible DW (2019) Water Waves to Sound Waves: Using Zebrafish to Explore Hair Cell Biology. J. Assoc Res. Otolaryngol 20 (1), 1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (243).Manley GA (2017) Comparative Auditory Neuroscience: Understanding the Evolution and Function of Ears. J. Assoc Res. Otolaryngol 18 (1), 1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (244).Barta CL, Liu H, Chen L, Giffen KP, Li Y, Kramer KL, Beisel KW, and He DZ (2018) RNA-seq transcriptomic analysis of adult zebrafish inner ear hair cells. Sci. Data 5, 180005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (245).Fritzsch B, Beisel KW, and Bermingham NA (2000) Developmental evolutionary biology of the vertebrate ear: conserving mechanoelectric transduction and developmental pathways in diverging morphologies. NeuroReport 11 (17), R35–44. [DOI] [PubMed] [Google Scholar]
  • (246).Nicolson T (2017) The genetics of hair-cell function in zebrafish. J. Neurogenet 31 (3), 102–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (247).Coffin AB, and Ramcharitar J (2016) Chemical Ototoxicity of the Fish Inner Ear and Lateral Line. Adv. Exp. Med. Biol 877, 419–37. [DOI] [PubMed] [Google Scholar]
  • (248).Nicolson T (2005) The genetics of hearing and balance in zebrafish. Annu. Rev. Genet 39, 9–22. [DOI] [PubMed] [Google Scholar]
  • (249).Varshney GK, Pei W, LaFave MC, Idol J, Xu L, Gallardo V, Carrington B, Bishop K, Jones M, Li M, Harper U, Huang SC, Prakash A, Chen W, Sood R, Ledin J, and Burgess SM (2015) High-throughput gene targeting and phenotyping in zebrafish using CRISPR/Cas9. Genome Res 25 (7), 1030–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (250).Alsina B, and Whitfield TT (2017) Sculpting the labyrinth: Morphogenesis of the developing inner ear. Semin. Cell Dev. Biol 65, 47–59. [DOI] [PubMed] [Google Scholar]
  • (251).Baxendale S, and Whitfield TT (2016) Methods to study the development, anatomy, and function of the zebrafish inner ear across the life course. Methods Cell Biol 134, 165–209. [DOI] [PubMed] [Google Scholar]
  • (252).Lu Z, and DeSmidt AA (2013) Early development of hearing in zebrafish. J. Assoc Res. Otolaryngol 14 (4), 509–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (253).Anthwal N, and Thompson H (2016) The development of the mammalian outer and middle ear. J. Anat 228 (2), 217–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (254).Basch ML, Brown RM, Jen HI, and Groves AK (2016) Where hearing starts: the development of the mammalian cochlea. J. Anat 228 (2), 233–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (255).Ghysen A, and Dambly-Chaudiere C (2004) Development of the zebrafish lateral line. Curr. Opin. Neurobiol 14 (1), 67–73. [DOI] [PubMed] [Google Scholar]
  • (256).Suli A, Watson GM, Rubel EW, and Raible DW (2012) Rheotaxis in larval zebrafish is mediated by lateral line mechanosensory hair cells. PLoS One 7 (2), No. e29727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (257).Nicolson T (2015) Ribbon synapses in zebrafish hair cells. Hear. Res 330 (Pt B), 170–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (258).Olt J, Johnson SL, and Marcotti W (2014) In vivo and in vitro biophysical properties of hair cells from the lateral line and inner ear of developing and adult zebrafish. J. Physiol 592 (10), 2041–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (259).Fettiplace R, and Kim KX (2014) The physiology of mechanoelectrical transduction channels in hearing. Physiol. Rev 94 (3), 951–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (260).Eggermont JJ (2017) Acquired hearing loss and brain plasticity. Hear. Res 343, 176–190. [DOI] [PubMed] [Google Scholar]
  • (261).Kindt KS, and Sheets L (2018) Transmission Disrupted: Modeling Auditory Synaptopathy in Zebrafish. Front. Cell Dev. Biol 6, 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (262).Chang-Chien J, Yen YC, Chien KH, Li SY, Hsu TC, and Yang JJ (2014) The connexin 30.3 of zebrafish homologue of human connexin 26 may play similar role in the inner ear. Hear. Res 313, 55–66. [DOI] [PubMed] [Google Scholar]
  • (263).Gou Y, Vemaraju S, Sweet EM, Kwon HJ, and Riley BB (2018) sox2 and sox3 Play unique roles in development of hair cells and neurons in the zebrafish inner ear. Dev. Biol 435 (1), 73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (264).Han S, Liu X, Xie S, Gao M, Liu F, Yu S, Sun P, Wang C, Archacki S, Lu Z, Hu X, Qin Y, Qu Z, Huang Y, Lv Y, Tu J, Li J, Yimer TA, Jiang T, Tang Z, Luo D, Chen F, and Liu M (2018) Knockout of ush2a gene in zebrafish causes hearing impairment and late onset rod-cone dystrophy. Hum. Genet 137 (10), 779–794. [DOI] [PubMed] [Google Scholar]
  • (265).Hartwell RD, England SJ, Monk NAM, van Hateren NJ, Baxendale S, Marzo M, Lewis KE, and Whitfield TT (2019) Anteroposterior patterning of the zebrafish ear through Fgfand Hh-dependent regulation of hmx3a expression. PLoS Genet 15 (4), No. e1008051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (266).Mulvaney JF, Thompkins C, Noda T, Nishimura K, Sun WW, Lin SY, Coffin A, and Dabdoub A (2016) Kremen1 regulates mechanosensory hair cell development in the mammalian cochlea and the zebrafish lateral line. Sci. Rep 6, 31668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (267).Schrauwen I, Kari E, Mattox J, Llaci L, Smeeton J, Naymik M, Raible DW, Knowles JA, Crump JG, Huentelman MJ, and Friedman RA (2018) De novo variants in GREB1L are associated with non-syndromic inner ear malformations and deafness. Hum. Genet 137 (6–7), 459–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (268).Bagnall MW, and Schoppik D (2018) Development of vestibular behaviors in zebrafish. Curr. Opin. Neurobiol 53, 83–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (269).Bhandiwad AA, Zeddies DG, Raible DW, Rubel EW, and Sisneros JA (2013) Auditory sensitivity of larval zebrafish (Danio rerio) measured using a behavioral prepulse inhibition assay. J. Exp. Biol 216, 3504–3513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (270).Matsumoto H, Fujiwara S, Miyagi H, Nakamura N, Shiga Y, Ohta T, and Tsuzuki M (2017) Carbonic Anhydrase Inhibitors Induce Developmental Toxicity During Zebrafish Embryogenesis, Especially in the Inner Ear. Mar. Biotechnol 19 (5), 430–440. [DOI] [PubMed] [Google Scholar]
  • (271).Rah YC, Yoo MH, Choi J, Park S, Park HC, Oh KH, Lee SH, and Kwon SY (2017) In vivo assessment of hair cell damage and developmental toxicity caused by gestational caffeine exposure using zebrafish (Danio rerio) models. Neurotoxicol. Teratol 64, 1–7. [DOI] [PubMed] [Google Scholar]
  • (272).Uribe PM, Sun H, Wang K, Asuncion JD, Wang Q, Chen CW, Steyger PS, Smith ME, and Matsui JI (2013) Aminoglycoside-induced hair cell death of inner ear organs causes functional deficits in adult zebrafish (Danio rerio). PLoS One 8 (3), No. e58755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (273).Niihori M, Platto T, Igarashi S, Hurbon A, Dunn AM, Tran P, Tran H, Mudery JA, Slepian MJ, and Jacob A (2015) Zebrafish swimming behavior as a biomarker for ototoxicity-induced hair cell damage: a high-throughput drug development platform targeting hearing loss. Transl Res 166 (5), 440–50. [DOI] [PubMed] [Google Scholar]
  • (274).Ou H, Simon JA, Rubel EW, and Raible DW (2012) Screening for chemicals that affect hair cell death and survival in the zebrafish lateral line. Hear. Res 288 (1–2), 58–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (275).Philip RC, Rodriguez JJ, Niihori M, Francis RH, Mudery JA, Caskey JS, Krupinski E, and Jacob A (2018) Automated High-Throughput Damage Scoring of Zebrafish Lateral Line Hair Cells After Ototoxin Exposure. Zebrafish 15 (2), 145–155. [DOI] [PubMed] [Google Scholar]
  • (276).Stengel D, Zindler F, and Braunbeck T (2017) An optimized method to assess ototoxic effects in the lateral line of zebrafish (Danio rerio) embryos. Comp. Biochem. Physiol., Part C: Toxicol. Pharmacol 193, 18–29. [DOI] [PubMed] [Google Scholar]
  • (277).Todd DW, Philip RC, Niihori M, Ringle RA, Coyle KR, Zehri SF, Zabala L, Mudery JA, Francis RH, Rodriguez JJ, and Jacob A (2017) A Fully Automated High-Throughput Zebrafish Behavioral Ototoxicity Assay. Zebrafish 14 (4), 331–342. [DOI] [PubMed] [Google Scholar]
  • (278).Yang Q, Sun P, Chen S, Li H, and Chen F (2017) Behavioral methods for the functional assessment of hair cells in zebrafish. Front Med 11 (2), 178–190. [DOI] [PubMed] [Google Scholar]
  • (279).Yao Q, DeSmidt AA, Tekin M, Liu X, and Lu Z (2016) Hearing Assessment in Zebrafish During the First Week Postfertilization. Zebrafish 13 (2), 79–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (280).Stawicki TM, Esterberg R, Hailey DW, Raible DW, and Rubel EW (2015) Using the zebrafish lateral line to uncover novel mechanisms of action and prevention in drug-induced hair cell death. Front. Cell. Neurosci 9, 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (281).Shin YS, Hwang HS, Kang SU, Chang JW, Oh YT, and Kim CH (2014) Inhibition of p38 mitogen-activated protein kinase ameliorates radiation-induced ototoxicity in zebrafish and cochlea-derived cell lines. NeuroToxicology 40, 111–22. [DOI] [PubMed] [Google Scholar]
  • (282).Lee SK, Oh KH, Chung AY, Park HC, Lee SH, Kwon SY, and Choi J (2015) Protective role of quercetin against cisplatin-induced hair cell damage in zebrafish embryos. Hum. Exp. Toxicol 34 (11), 1043–52. [DOI] [PubMed] [Google Scholar]
  • (283).Monroe JD, Millay MH, Patty BG, and Smith ME (2018) The curcuminoid, EF-24, reduces cisplatin-mediated reactive oxygen species in zebrafish inner ear auditory and vestibular tissues. J. Clin. Neurosci 57, 152–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (284).Teitz T, Fang J, Goktug AN, Bonga JD, Diao S, Hazlitt RA, Iconaru L, Morfouace M, Currier D, Zhou Y, Umans RA, Taylor MR, Cheng C, Min J, Freeman B, Peng J, Roussel MF, Kriwacki R, Guy RK, Chen T, and Zuo J (2018) CDK2 inhibitors as candidate therapeutics for cisplatin- and noise-induced hearing loss. J. Exp. Med 215 (4), 1187–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (285).Pang J, Xiong H, Zhan T, Cheng G, Jia H, Ye Y, Su Z, Chen H, Lin H, Lai L, Ou Y, Xu Y, Chen S, Huang Q, Liang M, Cai Y, Zhang X, Xu X, Zheng Y, and Yang H (2019) Sirtuin 1 and Autophagy Attenuate Cisplatin-Induced Hair Cell Death in the Mouse Cochlea and Zebrafish Lateral Line. Front. Cell. Neurosci 12, 515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (286).Oh KH, Rah YC, Hwang KH, Lee SH, Kwon SY, Cha JH, and Choi J (2017) Melatonin mitigates neomycin-induced hair cell injury in zebrafish. Drug Chem. Toxicol 40 (4), 390–396. [DOI] [PubMed] [Google Scholar]
  • (287).Takemoto Y, Hirose Y, Sugahara K, Hashimoto M, Hara H, and Yamashita H (2018) Protective effect of an astaxanthin nanoemulsion against neomycin-induced hair-cell damage in zebrafish. Auris, Nasus, Larynx 45 (1), 20–25. [DOI] [PubMed] [Google Scholar]
  • (288).Kenyon EJ, Kirkwood NK, Kitcher SR, O’Reilly M, Derudas M, Cantillon DM, Goodyear RJ, Secker A, Baxendale S, Bull JC, Waddell SJ, Whitfield TT, Ward SE, Kros CJ, and Richardson GP (2017) Identification of ion-channel modulators that protect against aminoglycoside-induced hair cell death. JCI Insight 2 (24), e96773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (289).Rihel J, Prober DA, Arvanites A, Lam K, Zimmerman S, Jang S, Haggarty SJ, Kokel D, Rubin LL, Peterson RT, and Schier AF (2010) Zebrafish behavioral profiling links drugs to biological targets and rest/wake regulation. Science 327 (5963), 348–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (290).Gandhi AV, Mosser EA, Oikonomou G, and Prober DA (2015) Melatonin is required for the circadian regulation of sleep. Neuron 85 (6), 1193–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (291).Yokogawa T, Marin W, Faraco J, Pezeron G, Appelbaum L, Zhang J, Rosa F, Mourrain P, and Mignot E (2007) Characterization of sleep in zebrafish and insomnia in hypocretin receptor mutants. PLoS Biol 5 (10), No. e277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (292).Nishimura Y, Okabe S, Sasagawa S, Murakami S, Ashikawa Y, Yuge M, Kawaguchi K, Kawase R, and Tanaka T (2015) Pharmacological profiling of zebrafish behavior using chemical and genetic classification of sleep-wake modifiers. Front. Pharmacol 6, 257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (293).Authier S, Bassett L, Pouliot M, Rachalski A, Troncy E, Paquette D, and Mongrain V (2014) Effects of amphetamine, diazepam and caffeine on polysomnography (EEG, EMG, EOG)-derived variables measured using telemetry in Cynomolgus monkeys. J. Pharmacol. Toxicol. Methods 70 (1), 86–93. [DOI] [PubMed] [Google Scholar]
  • (294).Ankley GT, and Johnson RD (2004) Small fish models for identifying and assessing the effects of endocrine-disrupting chemicals. ILAR J 45 (4), 469–83. [DOI] [PubMed] [Google Scholar]
  • (295).Law JM (2001) Mechanistic considerations in small fish carcinogenicity testing. ILAR J 42 (4), 274–84. [DOI] [PubMed] [Google Scholar]
  • (296).Gibert Y, Trengove MC, and Ward AC (2013) Zebrafish as a genetic model in pre-clinical drug testing and screening. Curr. Med. Chem 20 (19), 2458–66. [DOI] [PubMed] [Google Scholar]
  • (297).Strahle U, and Grabher C (2010) The zebrafish embryo as a model for assessing off-target drug effects. Dis. Models & Mech 3 (11–12), 689–92. [DOI] [PubMed] [Google Scholar]
  • (298).Han Y, Zhang J, and Hu C (2018) A systematic toxicity evaluation of cephalosporins via transcriptomics in zebrafish and in silico ADMET studies. Food Chem. Toxicol 116, 264–271. [DOI] [PubMed] [Google Scholar]
  • (299).Ye Q, Liu H, Fang C, Liu Y, Liu X, Liu J, Zhang C, Zhang T, Peng C, and Guo L (2019) Cardiotoxicity evaluation and comparison of diterpene alkaloids on zebrafish. Drug Chem. Toxicol, 1–8. [DOI] [PubMed] [Google Scholar]
  • (300).Paatero I, Veikkolainen V, Maenpaa M, Schmelzer E, Belting HG, Pelliniemi LJ, and Elenius K (2019) ErbB4 tyrosine kinase inhibition impairs neuromuscular development in zebrafish embryos. Mol. Biol. Cell 30 (2), 209–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (301).Mairesse J, Zinni M, Pansiot J, Hassan-Abdi R, Demene C, Colella M, Charriaut-Marlangue C, Rideau Batista Novais A, Tanter M, Maccari S, Gressens P, Vaiman D, Soussi-Yanicostas N, and Baud O (2019) Oxytocin receptor agonist reduces perinatal brain damage by targeting microglia. Glia 67 (2), 345–359. [DOI] [PubMed] [Google Scholar]
  • (302).Miller GW, Chandrasekaran V, Yaghoobi B, and Lein PJ (2018) Opportunities and challenges for using the zebrafish to study neuronal connectivity as an endpoint of developmental neurotoxicity. NeuroToxicology 67, 102–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (303).Xiao C, Han Y, Liu Y, Zhang J, and Hu C (2018) Relationship Between Fluoroquinolone Structure and Neurotoxicity Revealed by Zebrafish Neurobehavior. Chem. Res. Toxicol 31 (4), 238–250. [DOI] [PubMed] [Google Scholar]
  • (304).Garcia GR, Shankar P, Dunham CL, Garcia A, La Du JK, Truong L, Tilton SC, and Tanguay RL (2018) Signaling Events Downstream of AHR Activation That Contribute to Toxic Responses: The Functional Role of an AHR-Dependent Long Noncoding RNA (slincR) Using the Zebrafish Model. Environ. Health Perspect 126 (11), 117002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (305).Hahn ME, Timme-Laragy AR, Karchner SI, and Stegeman JJ (2015) Nrf2 and Nrf2-related proteins in development and developmental toxicity: Insights from studies in zebrafish (Danio rerio). Free Radical Biol. Med 88, 275–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (306).Qian J, Han Y, Li J, Zhang J, and Hu C (2018) Toxic effect prediction of cefatirizine amidine sodium and its impurities by structure-toxicity relationship of cephalosporins. Toxicol. In Vitro 46, 137–147. [DOI] [PubMed] [Google Scholar]
  • (307).Yoshioka W, and Tohyama C (2019) Mechanisms of Developmental Toxicity of Dioxins and Related Compounds. Int. J. Mol. Sci 20 (3), 617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (308).Chimote G, Sreenivasan J, Pawar N, Subramanian J, Sivaramakrishnan H, and Sharma S (2014) Comparison of effects of anti-angiogenic agents in the zebrafish efficacy-toxicity model for translational anti-angiogenic drug discovery. Drug Des., Dev. Ther 8, 1107–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (309).Farooq M, Sharma A, Almarhoon Z, Al-Dhfyan A, El-Faham A, Taha NA, Wadaan MAM, Torre BG, and Albericio F (2019) Design and synthesis of mono-and di-pyrazolyl-striazine derivatives, their anticancer profile in human cancer cell lines, and in vivo toxicity in zebrafish embryos. Bioorg. Chem 87, 457–464. [DOI] [PubMed] [Google Scholar]
  • (310).Veselinovic JB, Veselinovic AM, Ilic-Tomic T, Davis R, O’Connor K, Pavic A, and Nikodinovic-Runic J (2017) Potent anti-melanogenic activity and favorable toxicity profile of selected 4-phenyl hydroxycoumarins in the zebrafish model and the computational molecular modeling studies. Bioorg. Med. Chem 25 (24), 6286–6296. [DOI] [PubMed] [Google Scholar]
  • (311).Zhang CH, Chen K, Jiao Y, Li LL, Li YP, Zhang RJ, Zheng MW, Zhong L, Huang SZ, Song CL, Lin WT, Yang J, Xiang R, Peng B, Han JH, Lu GW, Wei YQ, and Yang SY (2016) From Lead to Drug Candidate: Optimization of 3-(Phenylethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine Derivatives as Agents for the Treatment of Triple Negative Breast Cancer. J. Med. Chem 59 (21), 9788–9805. [DOI] [PubMed] [Google Scholar]
  • (312).Donovan KA, An J, Nowak RP, Yuan JC, Fink EC, Berry BC, Ebert BL, and Fischer ES (2018) Thalidomide promotes degradation of SALL4, a transcription factor implicated in Duane Radial Ray syndrome. eLife 7, 38430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (313).Harvey SA (2006) sall4 acts downstream of tbx5 and is required for pectoral fin outgrowth. Development 133 (6), 1165–1173. [DOI] [PubMed] [Google Scholar]
  • (314).Matyskiela ME, Couto S, Zheng X, Lu G, Hui J, Stamp K, Drew C, Ren Y, Wang M, Carpenter A, Lee CW, Clayton T, Fang W, Lu CC, Riley M, Abdubek P, Blease K, Hartke J, Kumar G, Vessey R, Rolfe M, Hamann LG, and Chamberlain PP (2018) SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol 14 (10), 981–987. [DOI] [PubMed] [Google Scholar]
  • (315).Adamson KI, Sheridan E, and Grierson AJ (2018) Use of zebrafish models to investigate rare human disease. J. Med. Genet 55 (10), 641–649. [DOI] [PubMed] [Google Scholar]
  • (316).Hayes MN, and Langenau DM (2017) Discovering novel oncogenic pathways and new therapies using zebrafish models of sarcoma. Methods Cell Biol 138, 525–561. [DOI] [PubMed] [Google Scholar]
  • (317).Varga M, Ralbovszki D, Balogh E, Hamar R, Keszthelyi M, and Tory K (2018) Zebrafish Models of Rare Hereditary Pediatric Diseases. Diseases 6 (2), 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (318).He S, Jing CB, and Look AT (2017) Zebrafish models of leukemia. Methods Cell Biol 138, 563–592. [DOI] [PubMed] [Google Scholar]
  • (319).Jena M, Mishra A, and Maiti R (2019) Environmental pharmacology: source, impact and solution. Rev. Environ. Health 34 (1), 69–79. [DOI] [PubMed] [Google Scholar]
  • (320).Perkins EJ, Ankley GT, Crofton KM, Garcia-Reyero N, LaLone CA, Johnson MS, Tietge JE, and Villeneuve DL (2013) Current perspectives on the use of alternative species in human health and ecological hazard assessments. Environ. Health Perspect 121 (9), 1002–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (321).Aguirre-Martinez GV, Reinardy HC, Martin-Diaz ML, and Henry TB (2017) Response of gene expression in zebrafish exposed to pharmaceutical mixtures: Implications for environmental risk. Ecotoxicol. Environ. Saf 142, 471–479. [DOI] [PubMed] [Google Scholar]
  • (322).Horie Y, Yamagishi T, Takahashi H, Shintaku Y, Iguchi T, and Tatarazako N (2017) Assessment of the lethal and sublethal effects of 20 environmental chemicals in zebrafish embryos and larvae by using OECD TG 212. J. Appl. Toxicol 37 (10), 1245–1253. [DOI] [PubMed] [Google Scholar]
  • (323).Lazarevic N, Barnett AG, Sly PD, and Knibbs LD (2019) Statistical Methodology in Studies of Prenatal Exposure to Mixtures of Endocrine-Disrupting Chemicals: A Review of Existing Approaches and New Alternatives. Environ. Health Perspect 127 (2), 026001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (324).Genuis SJ, and Kyrillos E (2017) The chemical disruption of human metabolism. Toxicol. Mech. Methods 27 (7), 477–500. [DOI] [PubMed] [Google Scholar]
  • (325).Jarque S, and Pina B (2014) Deiodinases and thyroid metabolism disruption in teleost fish. Environ. Res 135, 361–75. [DOI] [PubMed] [Google Scholar]
  • (326).Petrie B, Barden R, and Kasprzyk-Hordern B (2015) A review on emerging contaminants in wastewaters and the environment: current knowledge, understudied areas and recommendations for future monitoring. Water Res 72, 3–27. [DOI] [PubMed] [Google Scholar]
  • (327).Safholm M, Ribbenstedt A, Fick J, and Berg C (2014) Risks of hormonally active pharmaceuticals to amphibians: a growing concern regarding progestagens. Philos. Trans. R. Soc., B 369 (1656), 20130577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (328).Scholz S, Fischer S, Gundel U, Kuster E, Luckenbach T, and Voelker D (2008) The zebrafish embryo model in environmental risk assessment–applications beyond acute toxicity testing. Environ. Sci. Pollut. Res 15 (5), 394–404. [DOI] [PubMed] [Google Scholar]
  • (329).Baker ME, and Hardiman G (2014) Transcriptional analysis of endocrine disruption using zebrafish and massively parallel sequencing. J. Mol. Endocrinol 52 (3), R241–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (330).Jarque S, Ibarra J, Rubio-Brotons M, Garcia-Fernandez J, and Terriente J (2019) Multiplex Analysis Platform for Endocrine Disruption Prediction Using Zebrafish. Int. J. Mol. Sci 20 (7), 1739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (331).Schiller V, Wichmann A, Kriehuber R, Schafers C, Fischer R, and Fenske M (2013) Transcriptome alterations in zebrafish embryos after exposure to environmental estrogens and anti-androgens can reveal endocrine disruption. Reprod. Toxicol 42, 210–23. [DOI] [PubMed] [Google Scholar]
  • (332).Spaan K, Haigis AC, Weiss J, and Legradi J (2019) Effects of 25 thyroid hormone disruptors on zebrafish embryos: A literature review of potential biomarkers. Sci. Total Environ 656, 1238–1249. [DOI] [PubMed] [Google Scholar]
  • (333).Li DL, Huang YJ, Gao S, Chen LQ, Zhang ML, and Du ZY (2019) Sex-specific alterations of lipid metabolism in zebrafish exposed to polychlorinated biphenyls. Chemosphere 221, 768–777. [DOI] [PubMed] [Google Scholar]
  • (334).Maradonna F, and Carnevali O (2018) Lipid Metabolism Alteration by Endocrine Disruptors in Animal Models: An Overview. Front. Endocrinol. (Lausanne, Switz.) 9, 654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (335).Huang Y, Wang XL, Zhang JW, and Wu KS (2015) Impact of endocrine-disrupting chemicals on reproductive function in zebrafish (Danio rerio). Reprod Domest Anim 50 (1), 1–6. [DOI] [PubMed] [Google Scholar]
  • (336).Vancamp P, Houbrechts AM, and Darras VM (2019) Insights from zebrafish deficiency models to understand the impact of local thyroid hormone regulator action on early development. Gen. Comp. Endocrinol 279, 45–52. [DOI] [PubMed] [Google Scholar]
  • (337).Darland T, and Dowling JE (2001) Behavioral screening for cocaine sensitivity in mutagenized zebrafish. Proc. Natl. Acad. Sci. U. S. A 98 (20), 11691–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (338).Kily LJ, Cowe YC, Hussain O, Patel S, McElwaine S, Cotter FE, and Brennan CH (2008) Gene expression changes in a zebrafish model of drug dependency suggest conservation of neuro-adaptation pathways. J. Exp. Biol 211 (Pt 10), 1623–34. [DOI] [PubMed] [Google Scholar]
  • (339).Klee EW, Schneider H, Clark KJ, Cousin MA, Ebbert JO, Hooten WM, Karpyak VM, Warner DO, and Ekker SC (2012) Zebrafish: a model for the study of addiction genetics. Hum. Genet 131 (6), 977–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (340).Ninkovic J, Folchert A, Makhankov YV, Neuhauss SC, Sillaber I, Straehle U, and Bally-Cuif L (2006) Genetic identification of AChE as a positive modulator of addiction to the psychostimulant D-amphetamine in zebrafish. J. Neurobiol 66 (5), 463–75. [DOI] [PubMed] [Google Scholar]
  • (341).Ponzoni L, Braida D, and Sala M (2016) Abuse potential of methylenedioxymethamphetamine (MDMA) and its derivatives in zebrafish: role of serotonin 5HT2-type receptors. Psychopharmacology (Berl) 233 (15–16), 3031–3039. [DOI] [PubMed] [Google Scholar]
  • (342).Mathur P, Berberoglu MA, and Guo S (2011) Preference for ethanol in zebrafish following a single exposure. Behav. Brain Res 217 (1), 128–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (343).Bretaud S, Li Q, Lockwood BL, Kobayashi K, Lin E, and Guo S (2007) A choice behavior for morphine reveals experience-dependent drug preference and underlying neural substrates in developing larval zebrafish. Neuroscience 146 (3), 1109–16. [DOI] [PubMed] [Google Scholar]
  • (344).Cachat J, Canavello P, Elegante M, Bartels B, Hart P, Bergner C, Egan R, Duncan A, Tien D, Chung A, Wong K, Goodspeed J, Tan J, Grimes C, Elkhayat S, Suciu C, Rosenberg M, Chung KM, Kadri F, Roy S, Gaikwad S, Stewart A, Zapolsky I, Gilder T, Mohnot S, Beeson E, Amri H, Zukowska Z, Soignier RD, and Kalueff AV (2010) Modeling withdrawal syndrome in zebrafish. Behav. Brain Res 208 (2), 371–6. [DOI] [PubMed] [Google Scholar]
  • (345).Bosse GD, and Peterson RT (2017) Development of an opioid self-administration assay to study drug seeking in zebrafish. Behav. Brain Res 335, 158–166. [DOI] [PubMed] [Google Scholar]

RESOURCES