Next Article in Journal
The Hematological and Biochemical Effects from Pesticide Exposure on Thai Vegetable Farmers
Previous Article in Journal
Gourmet Table Salts: The Mineral Composition Showdown
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rapid Assessment of Ocular Toxicity from Environmental Contaminants Based on Visually Mediated Zebrafish Behavior Studies

by
Jia Yi
1,†,
Yilei Ma
1,†,
Jiahui Ma
1,†,
Haiyang Yu
1,
Kun Zhang
2,
Libo Jin
3,
Qinsi Yang
4,
Da Sun
1,3,* and
Dejun Wu
5,*
1
Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
2
Bioengineering College of Chongqing University, Chongqing 400030, China
3
National and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
4
Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
5
Emergency Department, Quzhou People’s Hospital, Quzhou 324000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Toxics 2023, 11(8), 706; https://doi.org/10.3390/toxics11080706
Submission received: 29 June 2023 / Revised: 4 August 2023 / Accepted: 14 August 2023 / Published: 16 August 2023
(This article belongs to the Special Issue Environmental Pollutants Exposure and Human Eye Health)

Abstract

:
The presence of contaminants in the environment has increased in recent years, and studies have demonstrated that these contaminants have the ability to penetrate the blood–retinal barrier and directly affect the visual systems of organisms. Zebrafish are recognized as an ideal model for human eye diseases due to their anatomical and functional similarities to the human eye, making them an efficient and versatile organism for studying ocular toxicity caused by environmental contaminants in the field of environmental toxicology. Meanwhile, zebrafish exhibit a diverse repertoire of visually mediated behaviors, and their visual system undergoes complex changes in behavioral responses when exposed to environmental contaminants, enabling rapid assessment of the ocular toxicity induced by such pollutants. Therefore, this review aimed to highlight the effectiveness of zebrafish as a model for examining the effects of environmental contaminants on ocular development. Special attention is given to the visually mediated behavior of zebrafish, which allows for a rapid assessment of ocular toxicity resulting from exposure to environmental contaminants. Additionally, the potential mechanisms by which environmental contaminants may induce ocular toxicity are briefly outlined.

1. Introduction

As a superficial organ, the eye is frequently directly exposed to the surrounding environment, leaving it susceptible to direct or indirect harm from hazardous elements in the environment that can cause eye injury [1,2]. With an estimated 295 million people worldwide suffering from moderate and severe visual impairment as of 2020, including approximately 43.3 million who are even blind [3], environmental contaminants are gaining attention as an important direct or indirect causative factor [4,5]. Meanwhile, several epidemiological studies suggest that environmental pollutants may be associated with congenital eye malformations [6]. These include exposure to environmental chemical pollutants during pregnancy or during the early stages of newborn life, which may cause vision problems in newborns through the placenta or breast milk [7,8,9,10]. As shown in Figure 1, environmental contaminants that have been shown to act on the visual system, which include endocrine disrupting chemicals (EDCs), brominated flame retardants (BFRs), pharmaceutical and personal care products (PPCPs), microplastics (MPs), agrochemicals, and heavy metals, which adversely impact embryonic development and the visual performance of the neural retina of organism [11], causing multiple visual system defects at different levels of biological tissues (Figure 1) (e.g., genes, proteins, organology, and physiology) [12,13,14]. Moreover, water-insoluble contaminants are commonly investigated through the utilization of organic solvents, among which are dimethyl sulfoxide, methanol, and acetone, serving as co-solvents [15,16,17]. In the face of such a severe threat from environmental contaminants, there is an urgent need for a scientifically rational animal model to be developed for a rapid and accurate evaluation of the ocular toxicity of environmental contaminants on organisms and to study their mechanisms.
The zebrafish (Danio rerio), a typical biological model in ecotoxicology research, has a fully sequenced genome, with the majority of zebrafish genes shared with humans [18]. Furthermore, 84% of known human disease-related genes have zebrafish counterparts, and zebrafish exhibit a wide range of morphological and biochemical similarities with humans [19,20]. Since visual development and functional databases for the zebrafish are now accessible, they have become as an essential model in hard-boned fish research for toxicological investigations [21].
Similar to mammals, zebrafish also possess a variety of social behaviors, including social interaction, anxiety, learning, memory, and defense mechanisms [22,23,24]. Foraging and predator avoidance are both closely related to vision, which is the most fundamental sensory system in zebrafish. In addition, environmental contaminants can accumulate in the eyes of an organism, altering the expression of essential visual proteins, disrupting retinal structure, and impairing the function of the visual system [25,26,27], so behavioral endpoints can be utilized to rapidly investigate the effects of pollutants on the visual system. Such behavioral studies in living organisms can provide a more comprehensive understanding of the effects of contaminants [28].
Furthermore, the advantage of zebrafish models in ophthalmic research is derived from the similarities between human and zebrafish embryo eye development architecture, as well as the remarkable adaptability of zebrafish to experimental and genetic modification [29]. In comparison to mammals, which have long cycles, large economic costs, and complex operations, zebrafish exhibit high fecundity fertilization capacity in vitro, a quick embryonic development process, a small size, and plentiful embryonic resources, making them excellent for high-throughput screening [30].
This review comprehensively introduces the ocular anatomy and function of zebrafish, summarizes its methods for rapidly evaluating the ocular toxicity of environmental contaminants, and describes the underlying mechanisms responsible for such toxicity. Finally, the challenges and prospective development trends in utilizing the zebrafish model to assess the ocular toxicity of environmental contaminants are further discussed.

2. The Eye Structure of Zebrafish

The zebrafish has emerged as a promising model for studying human eye disorders due to its remarkable similarities in anatomy, function, tissue composition, circuitry, and biochemistry with other vertebrates [31]. The highly conserved nature of zebrafish eyes among vertebrates has facilitated the successful modeling of numerous eye disorders [32], including cataracts, glaucoma, diabetic retinopathy, and age-related macular degeneration [16,33]. Zebrafish retinas exhibit the typical morphology found in vertebrate retinas, resembling adult retinas, and share both anatomical and functional similarities with human retinas [29,34]. While the macula is a cone-shaped, dense region responsible for central high-resolution color vision in the human retina [35], recent discoveries have identified a comparable macular area in the retinas of zebrafish larvae, which plays a crucial role in extremely sensitive vision, akin to the human macula [36,37]. Furthermore, the arrangement of neural networks and gene expression patterns in zebrafish photoreceptors closely resemble those observed in humans [38]. Consequently, the zebrafish has emerged as the preferred model for investigating human retinal diseases (Figure 2).
The retina of the zebrafish consists of three layers: the outer nuclear layer (ONL), the inner nuclear layer (INL), and the ganglion cell layer (GCL), which are separated by synapse-rich plexiform layers (outer plexiform layer, OPL; inner plexiform layer, IPL) [35]. There are the same retinal neurons in zebrafish as in humans, including retinal ganglion cells (RGC), bipolar cells, horizontal cells, and amacrine cells, as well as the same glial cells, including Müller cells, astrocytes, and microglia [39]. The retina is covered by Müller glial cells, which serve as scaffolds for neurons, interact with all retinal neuronal bodies and processes, and contribute to the regulation of neurotransmitters as well as gas and ion homeostasis [40,41]. The ONL consists of one rod type and four morphologically and spectrally distinct cone subtypes, namely short single cones (ultraviolet (UV)-sensitive), long single cones (blue-sensitive), double cone accessory members (green-sensitive), and double cone principle members (red-sensitive) [42,43]. The INL is composed of interneurons (bipolar cells, horizontal cells, and amacrine cells) and Müller glia [36]. The plexus layer lies between the nuclear layers and serves as the primary site of synaptic connections between retinal neurons. The OPL contains photoreceptors, bipolar cells, and horizontal cells, while the IPL consists of bipolar cells, amacrine cells, and ganglion cells [36].
ONL is responsible for the initial step in light signal reception and conversion into electrical signals [44]. When ambient light enters the eye, it reaches the retina through the refractive medium of the bipolar cells and photoreceptors, with the latter playing a crucial role in this process. Visual signals from photoreceptors, including cones and rods, are transferred from the retina to ganglion cells in the GCL and subsequently relayed to the brain [45]. Additionally, the retinal pigment epithelium (RPE), which is a layer of pigment cells located outside the neurosensory retina, supports the retinal vision cells and serves as a critical interface between photoreceptors and the choroid in the vertebrate eye [11,46]. Extensive molecular interactions occur between the photoreceptors in the neural retina and the RPE [40].
Zebrafish provide reliable endpoints for the study of retinal injury and the investigation of developmental toxicity in the ocular system as an excellent vertebrate model. One prominent method utilized in both rodent and zebrafish retina research is optical coherence tomography retinal imaging [47]. This non-invasive interference technique provides cross-sectional and facial images of the rodent and fish retina, enabling precise identification of minimal lesions and significant changes in retinal structure, thus achieving high-resolution retinal imaging [35,48]. Yet, it is worthwhile to emphasize that a variety of behavioral paradigms can be utilized to quickly, intuitively, and directly identify impairments in zebrafish visual function. Visually mediated behavioral changes have been widely utilized to evaluate the ocular toxicity of pollutants in the early stages of zebrafish life [49]. In visual research, mice and rats are commonly used as primary animal models, whose retinas are mainly composed of rod cells, resulting in color vision defects or loss, relatively low visual acuity, and more reliance on olfactory, tactile, and auditory cues in bright environments [35]. In contrast, the zebrafish serves as a valuable animal model for studying visually mediated behaviors, offering notable advantages. Firstly, zebrafish have a cone-dominated vision similar to humans, unlike mice, which have rod-dominated vision. Zebrafish possess cone photoreceptors that are sensitive to UV, blue, green, and red light, resulting in excellent color vision and visual acuity [35,36]. Secondly, zebrafish exhibit strong reproductive capabilities, with externally developing embryos that are optically transparent, facilitating easy observation. Additionally, zebrafish have relatively large eyes in proportion to their body size, allowing for manipulation of the eye bud during early embryonic development [39,50]. Thirdly, the visual system of zebrafish develops rapidly, with eye development completed by 72 h post-fertilization (hpf), enabling the direct observation of morphological defects within a short timeframe. Functional assessments can be conducted after 120 hpf, enabling faster identification of issues related to mature visual acuity compared to mice, which typically take around 15–20 days [51]. Fourthly, the small size of zebrafish makes them well-suited for behavioral tests in multiwell plates, and their behavior can be effortlessly monitored using automated video tracking systems [26]. Lastly, both adult zebrafish and larvae exhibit easily analyzable visual behaviors without the need for training, and there exist various robust behavioral paradigms for rapidly assessing the visual function of zebrafish and detecting visual impairments [35,40].

3. Quick Approaches for Assessing Ocular Toxicity

Behavior integrates the responses of animals to internal and external stimuli and can be used as an indicator of the effects of environmental pollutants on animals at molecular, biochemical, and physiological levels [52]. Such behavioral disorders arise from changes in neuronal structure and/or physiology. Particularly, zebrafish behavior exhibits high sensitivity to environmental pollutants, making it a particularly effective model for assessing the toxic effects of these pollutants [52]. In recent times, Zebrafish behavior has been quantitatively assessed in a growing number of studies to evaluate the toxicity endpoints of pollutants in zebrafish [53,54].
Zebrafish possess a visual processing circuitry that comprises the neural retina, optic nerve, optic tectum, and extraocular muscles. Their behavior heavily relies on a mature retina, which enables stable high-resolution imaging and eye movement reflexes that respond to fast-moving visual stimuli [51]. The activity of innate visual reflexes supports a variety of visually mediated behaviors and effectively assesses the visual function of zebrafish [11]. Several commonly used methods for studying zebrafish behavior include the optokinetic reflex (OKR), optomotor response (OMR), phototaxis, light–dark preference test, free-swimming activity, and visual avoidance behavior analysis (Figure 3) [11,35,55]. These behavioral responses have been extensively utilized in toxicology studies to identify different types of visual cues driving behavior, genes involved in vision and visual impairments, and the specific neurons and circuits responsible for perceiving visual stimuli and eliciting behavioral responses, often utilizing in vivo functional imaging techniques [32].

3.1. OKR

OKR is a convenient method for evaluating visual function in awake animals, as it provides visual stimulation and relies on the functional retina without the need for training [56]. The test typically involves immobilizing zebrafish in a transparent tank and stimulating them with gradually increasing or decreasing circular or vertical stripes, allowing only their eyes to move as they track the rotating stripes, followed by observing their eye movements [51,57]. This behavior matures between 73 and 80 hpf and persists throughout adulthood [58]. During OKR testing, fish with expected visual function deficiencies are anticipated to exhibit a reduced frequency of eye scanning or abnormal eye movements [59,60]. However, there are variations in the experimental protocol for conducting the OKR test in practice. These differences include the growth stage of the zebrafish, fixation methods, rotation speed of the stripe pattern, and the parameters used for behavioral evaluation of the experimental zebrafish, as indicated in Table 1.
OKR is also frequently used in assessing visual function in larvae, and it has been employed in studies investigating the effects of environmental contaminants on zebrafish [16]. These investigations have revealed that zebrafish larvae exposed to certain polybrominated diphenyl ether (PBDE) congeners, such as 2,2′,4,4′,5-pentabromodiphenyl ether (BDE99), exhibited decreased saccadic responses and responses to blue and green light stimuli in OKR tests assessing visual behavior [16]. Similarly, exposure to polycyclic aromatic hydrocarbons (PAHs) present in crude oil led to diminished eye movements [61], while exposure to the plasticizer bisphenol S (BPS) significantly impaired eye-tracking ability during visual behavior OKR testing [56]. Furthermore, exposure to triphenyl phosphate (TPhP) and triclocarban (TCC) was found to reduce the OKR response, with TPhP demonstrating a dose-dependent decrease [44,62]. In general, exposure to these environmental contaminants may have detrimental effects on the visual function of zebrafish. However, it is important to note that exposure to certain environmental contaminants can also cause zebrafish to exhibit a positive OKR. For instance, exposure to the commercial PBDE mixture, DE-71, significantly increased visual–motor response [63].
Table 1. Comparative analysis of variations in OKR implementation process.
Table 1. Comparative analysis of variations in OKR implementation process.
CompoundsStageApparatusStripe PatternBehavior Evaluation ParametersRef.
BDE-99120 hpf35 mm Petri dish
containing 3% methylcellulose
Rotational black and white stripes (10 rpm)Saccadic movements per minute[16]
Crude Oil96 hpf35 mm Petri dish
Containing 3% methylcellulose
Rotating black and white vertical stripes (6 rpm)[61]
DE-7115 dpf35 mm Petri dish
containing 6% methylcellulose
A drum lined with 18° vertical black and white stripes. (8.1 rpm)[63]
BPS120 days post-fertilization(dpf)A specially constructed cylindrical water-filled glass chamberRotating drum with black and white stripes (Unstated speed)Slow phase gain[56]
TPhP144 hpf35 mm Petri dish
containing 6% methylcellulose
Sine-wave grating (with a constant angular velocity of 7.5 per second and a spatial frequency of 0.6 cycles per degree)Ratio of eye velocity to stimulus velocity[44]
TCC144 hpfFixed in a glass slide with 1.6% agarose solutionMoving vertical black-white, blue-white, green-white, and red-white stripes (12 cycles/360°, 30°/s) [62]

3.2. OMR

The OMR is a rapid, straightforward, and efficient method for assessing visual function in zebrafish [32], which involves visual stimulation of the fish with moving stripe patterns presented alternately from below or beside the fish, consisting of vertical stripes of different colors, such as black and white, gray with different contrasts, or other different colors [32,64]. The OMR is an innate visuomotor reflex behavior in which fish swim in the same direction as high-contrast visual stimuli [65]. These behaviors may be displayed both while swimming freely or while immobilized with the head of zebrafish larvae embedded in agarose while allowing the trunk and tail to move [32].
Scoring the response of larvae is possible through video recordings of their swimming behavior, typically with fixed larvae, or by recording the distance moved relative to the direction of the stimulus [32]. The latter approach can be performed in a high-throughput manner using 384-well plates [32]. While OMR is similar to OKR, the stimulus in OMR drives the movement of the head and body rather than the eyes. These tests have different visual functional features, while OMR and OKR test results overlap. OMR emphasizes the visual motor ability of adult zebrafish, which depends on functional retinal projection [56]. Additionally, the design of OMR experiments may vary depending on the developmental stage of the zebrafish, the experimental equipment used, and the parameters for behavioral evaluation, as presented in Table 2.
Zebrafish larvae exposed to polychlorinated biphenyls, specifically PCB1254, exhibited abnormal behavior in OMR tests, with a significant reduction in the proportion of actively swimming fish compared with control larvae [15]. Similarly, male zebrafish exposed to BPS exhibited a significant reduction in their tracking ability during OMR testing [56]. However, no significant difference was observed in the OMR tests of zebrafish following acute exposure to the insecticides atrazine and diazauron, as all groups exhibited positive visuomotor responses [66].
Table 2. Comparative analysis of variations in OMR implementation process.
Table 2. Comparative analysis of variations in OMR implementation process.
CompoundsStageApparatusBehavior Evaluation ParametersRef.
PCB12547 dpfTransparent test tanks with black and white stripes are generated using custom-designed raster software.The proportion of fish that positive swimming, opposite swimming, hesitant swimming, and immobility[15]
BPS120 dpf A round transparent tank with an opaque column in the middle and a rotating drum with black and white stripes outside the tank. (Turn the drum clockwise for 1 min, then counterclockwise)Concordance ratio (the quotient of male zebrafish stripe tracking time divided by the whole recording time)[56]
Atrazine, diazauron7 dpfPetri dishes over an LCD monitor with moving animations in black and white stripesProportion of zebrafish that moved to the end of the dish following the stripes[66]

3.3. Phototaxis Behavior Response

Phototaxis is an innate behavior in which zebrafish larvae exhibit a response to light, whereby changes in the sensitivity of their eyes to light stimuli decrease the perceived difference between light and dark areas [16,49]. The phototaxis behavioral response test serves as a rapid and straightforward screening tool used to assess eye defects induced by environmental contaminants [44,67,68]. This test capitalizes on the natural inclination of zebrafish to move toward the lighted chamber, enabling the assessment of the coordination between motor movements and sensory perception in their visual system. It is widely employed to identify recessive visual defects in model biological systems [16,35]. To prevent any discrepancies in individual larva movements, zebrafish larvae with normal swimming function are typically selected for investigations.
The phototaxis behavioral response test typically involves the utilization of a rectangular acrylic box with a sliding partition that separates compartments A and B. A specific number of zebrafish larvae are placed in darkened chamber B for a predetermined period. Once the larvae have acclimated to the dark environment, the partition is opened to expose chamber A to ambient light, while chamber B remains dark. The larvae in each chamber are counted, and the percentage of larvae that swim from the dark chamber to the light chamber after a designated time is used to assess their responsiveness to light. The test can be conducted using blue, green, and red light in the lighting room to evaluate the impact of color perception. To assess whether the larvae accidentally entered chamber A, a controlled experiment was performed in which the chamber remains darkened after the partition is raised [62]. The detailed experimental protocol is shown in Table 3.
Environmental contaminants have the potential to induce functional impairments in the eye or visual processing centers, resulting in damage to photoreceptors, retinal morphological changes, and cell apoptosis [46]. Photoreceptors and their accompanying opsins are particularly sensitive to exogenous substances among the numerous components of the visual system [16]. Several studies have demonstrated that zebrafish larvae are less responsive to light after being exposed to environmental contaminants [69].
The phototaxis behavior of zebrafish larvae was observed to be inhibited following a single exposure to TCC [62], BDE99 [16], phenanthrene polycyclic aromatic hydrocarbon (Phe) [46], TPhP [44], and the agricultural chemical boscalid [17]. Notably, zebrafish larvae exposed to TPhP exhibited a dose-dependent reduction in the phototaxis response [44]. Various wavelengths of light signals (short, medium, and long) were employed to assess the ability of zebrafish larval cone cells to perceive different wavelength spectra. It was found that exposure to BPS inhibited the phototaxis of zebrafish larvae across all wavelength bands (short, medium, and long) in order to test the ability of larval cone cells to perceive varying wavelength spectra [70]. In contrast, exposure to DE-71 resulted in an increase in light-seeking behavior, as indicated by the phototaxis test results of zebrafish larvae [63].
Table 3. Comparative analysis of variations in the practical application of phototaxis behavior response.
Table 3. Comparative analysis of variations in the practical application of phototaxis behavior response.
CompoundsStageExperiment DurationBehavior Evaluation ParametersRef.
Duration in the Dark ChamberDuration after Removing the Partition
TCC144 hpf10 min5 minNumber of larvae swimming from the dark chamber into the illuminated chamber.[62]
BDE-99120 hpf5 min5 min, 10 min[16]
Phe7 dpf 2 min30 min[46]
Boscalid8 dpf10 min1 min[17]
TPhP144 hpf5 min1 min[44]
DE-7115 dpf2 min1 min[63]
BPS5 dpf15 min15 minNumbers of larvae travelled to the light chamber within 1 min.[70]

3.4. Light-Dark Preference Test

The light–dark preference test is a well-established approach to assessing anxiety-like behavior in mice and investigating the mechanisms underlying drug-induced neurobehavioral changes [71]. Recently, this test has also been recognized as an extremely valuable tool for evaluating the vision of zebrafish as well as behavioral alterations due to retinal damage and regeneration [38]. The light–dark preference test is particularly advantageous for studying the behavioral neuroscience of zebrafish, as opposed to rodents, who typically prefer darkness [72]. Research involving zebrafish commonly utilizes this test, including high-throughput analysis of neural phenotypes and drug screening [22].
Typically, the light–dark preference test involves exposing zebrafish to alternate light and dark cycles [73]. The experimental setup can vary in terms of the developmental stage of zebrafish larvae, the number of holes in the porous plate, and the duration of the experiment, including the duration of light and dark conditions, as summarized in Table 4. Several key behavioral parameters are measured during the light–dark preference test, including movement times, average speed, maximum speed, chemotaxis, and the total distance traveled [74].
Exposure of zebrafish to agrochemicals, specifically the phenylurea herbicides linuron and pyrethroid esfenvalerate, has been demonstrated to exert inhibitory effects on locomotor activity [75,76]. Notably, locomotion under dark conditions was also found to be reduced following exposure to 2,2′,4,4′-tetrabromodiphenyl ether (PBDE-47) [77]. In contrast, exposure to BPS, retinoic acid, and the substitute for perfluorooctane sulfonic acid, F-53B, results in hyperactive swimming behavior, as indicated by the results of light-dark preference tests [69,78,79]. Furthermore, the presence of pharmaceutical compounds in aquatic environments can exert an influence on the development of zebrafish eyes. For instance, exposure of zebrafish to amitriptyline (AMI), venlafaxine (VEN), and sertraline (SER) leads to significant alterations in their swimming distance under both dark and light conditions [80]. Additionally, exposure to cyclophosphamide (CP) markedly inhibits the swimming speed of zebrafish larvae [81]. Conversely, the locomotor activity of zebrafish larvae remains unaffected by exposure to the beta-blocker atenolol [82]. However, under prednisolone exposure, the light–dark preference test results reveal that prednisolone alters the embryonic response to darkness, although these changes are not directly mediated by visual alterations [49].
Table 4. Comparative analysis of variations in the practical application of light-dark preference test.
Table 4. Comparative analysis of variations in the practical application of light-dark preference test.
CompoundsStage and Well PlateProtocolsBehavior Evaluation ParametersRef.
Retinoic acid120 hpf, 24 well plate (1 fish/well)Adaptation to 10 min in the dark was followed by a 10 min light (visible light) interval followed by a 10 min dark (infrared light) interval and repeated for 50 min. Net speed change (or difference) in average swimming speeds between the last minute of 1 light state and the first minute of another light state and the average swimming speed of 10 min intervals for each state (light or dark)[78]
AMI, VEN, SER144 hpf, 96-microwell platesAdaptation to light for 10 min, 16 alternating dark and light periods, each 5 min long, 90 min.Swimming distance[80]
Prednisolone120 hpf, the apparatus consisted of a 6 cm diameter petri dish marked with a grid floor (4.5 mm2; approx. one body length), equally divided into a white and black area. The floor of the dish was further divided into three circular areas (outer, middle and central).Individual embryos were initially acclimated within a small transparent chamber positioned within the central area for 1 min to explore the tank for 5 min.The percentage of time spent in each compartment (light vs. dark), the proportion of time within each of the three circular areas within the light compartment, and the total level of activity (total number of lines crossed) while in the white compartment.[49]
PBDE-47 4, 5, and 6 dpf, 96-well microplatesAdaptation to the dark took 20 min, and then recording started with a dark period of 10 min, followed by three cycles of alternating 10 min light and dark periods. The total testing time was 70 min.Distance moved[77]
Atenololexposed to atenolol for 7 days, 96-well platesAlternating 10 min periods of light and dark, beginning with a dark period (i.e., dark photokinesis), for a total of 50 min.[82]
Linuron7 dpf larvae, 96-well plate[76]
Esfenvalerate5 dpf, 6 dpf, or 7 dpf, 96-well platesAlternating light-dark periods (10 min light–10 min dark–10 min light–1 min dark) for 50 min.[75]
F-53B120 hpf, 24-well plates (one larva for each well)Adaptation was performed 30 min before the start of the test, followed by a light (10 min) –dark (10 min) –light (10 min) stimulus.Swimming speed (mm/s)[69]
CPlarvae, 48-well platesAlternating light-dark periods (10 min light–10 min dark–10 min light–1 min dark) for 50 min.Swimming speed (mm/min)[81]
BPSAfter 120 h of BPS exposure, 24-well plates (1 larvae/well, 2 mL solution/well)Alternating light-dark periods (1 min light–1 min dark–1 min light–1 min dark) for 4 min.The traveled distance per minute, 4 min total distances, and average speed.[79]

3.5. Free Swimming

Swimming ability is a critical behavioral parameter for fish as it plays a pivotal role in their survival, reproduction, and defense [83]. Impairments in visual and sensory processes can lead to lower swimming capability in zebrafish, which can have a negative impact on feeding, growth, and predator avoidance, eventually jeopardizing their ability to flourish in natural settings [68]. The free-swimming activity test assesses the visual function of zebrafish by monitoring their movements in an unrestricted state. This test involves placing zebrafish in a transparent tank, observing their swimming behavior without any interference from shock or noise, and recording their swimming trajectory under continuous visible light using a high-resolution camera. One of the primary parameters assessed in this test is swimming speed [68]. The specific protocols for conducting free swimming tests can vary, including the developmental stage of zebrafish larvae and the experimental design. More detailed information can be found in Table 5.
Nanoplastics (NPs) are emerging contaminants that have adverse effects on the environment and can serve as carriers for other coexisting pollutants [84,85]. For instance, the sunscreen butyl methoxydibenzoylmethane (Avobenzone, AVO) can adsorb onto the surface of nanoplastic particles, hindering their degradation and preventing their entry into the aquatic environment via conventional wastewater treatment methods 52. Exposure to NPs has been found to significantly decrease the swimming activity of zebrafish larvae. Further, zebrafish larvae showed a significant reduction in swimming activity when exposed to the combined exposure of NPs and AVO [86,87]. Nevertheless, research has demonstrated that retinoic acid at a dose of 2 nM improves the free swimming speed of zebrafish under both light and dark conditions [78].
Table 5. Comparative analysis of variations in the practical application of free swimming.
Table 5. Comparative analysis of variations in the practical application of free swimming.
CompoundsStage and Well PlateExperimental PlanBehavior Evaluation ParametersRef.
Retinoic acid120 hpf, 24-well plates (1 fish/well)Two 20 min swim tests in the light and darkSwimming speed (mm/s)[78]
The combined exposure of NPs and AVO Exposure lasted for 144 h of morphologically normal zebrafish larvae, 24-well
plates (1 larva/well)
After 10 min of daptation, a 15 min swim test was performed under continuous visible light.[87]
120 hpf, 24-well plate (1 larva/well)[86]

3.6. Visual Avoidance Behavior

Zebrafish embryos exhibit unique avoidance behavior, which makes them a valuable model for assessing their motion detection ability. Simple animations composed of basic shapes are employed to elicit avoidance responses in zebrafish during the early developmental stages. The perception of motion necessitates image formation and tracking of positional changes over time, encompassing intricate visual processes that interact with the nervous system [49]. The specific implementation of this test varies depending on the growth stage of the zebrafish, the patterns employed for visual stimulation, and the individual experiment plan, as outlined in Table 6.
Previous research has demonstrated that zebrafish larvae exposed to MPs, Cu, prednisolone, PCB-95, and combined exposures of Cu and MPs exhibit abnormal behavior during early developmental stages. These abnormalities manifest as either a lack of significant avoidance response towards visual stimuli or reduced avoidance behavior [49,88,89].
Table 6. Comparative analysis of variations in practical applications of visual avoidance behavior.
Table 6. Comparative analysis of variations in practical applications of visual avoidance behavior.
CompoundsStage and Well PlateVisual Stimuli PatternsProtocolsBehavior Evaluation ParametersRef.
Prednisolone144 hpf, six-well platesProduced by Microsoft PowerPoint, it consists of two black rectangles (0.4–14.6 cm) placed parallel to each other.Acclimatization was performed on a white screen for 5 min, followed by 5 min of recording before stimulus onset, after which the animation was played for 5 min.(Pre-animation, during, and post-animation) The amount of time embryos spent in the outer area of the well[49]
PCB-955 dpf, 6-well platesProduced by Microsoft PowerPoint, red lines (RGB values were 255, 0, 0) were spaced 1 mm apart and moved from the top to the bottom relative to the dish at a speed of 7 mm every 8 s.Exposure to a uniform white background without visual stimuli was performed for 10 min, followed by moving the red line for 10 min and then returning to the white background for 10 min.Swim speed, percentage of larval edge preference, and percentage of larval avoidance[88]
MPs, Cu, and combined exposures of Cu and MPs14 dpf, 6-well platesProduced by Microsoft PowerPoint, static or bouncing two-dimensional red discs (1.35 cm diameter, RGB 255, 0, 0)Alternating 10 min of a white background and 10 min of a red bouncing ball appeared in the upper half of the well and moved from left-right-left on a 2 cm straight trajectory at 1 cm/sThe time spent in the lower half of the well[89]

4. Mechanism of Ocular Toxicity

4.1. Oxidative Stress and Inflammation

Environmental contaminants have the potential to impact the ocular immune system by inducing oxidative stress, resulting in abnormal production of cytokines and chemokines and altered immune cell function (Figure 4). Oxidative stress is a major cause of photoreceptor damage, and it can be directly caused by the accumulation of reactive oxygen species (ROS) and aberrant metabolic activities [90]. To counteract oxidative damage and maintain cellular redox homeostasis, the antioxidant defense system relies on a combination of antioxidant enzymes and non-enzymatic antioxidant molecules. Superoxide dismutase (SOD) and catalase (CAT) are key antioxidant enzymes that act as the first line of defense against oxidative toxicity, and elevated levels of malondialdehyde (MDA), a consequence of lipid peroxidation, serve as indicators of oxidative damage [91,92]. Previous studies have shown that environmental contaminants, including endocrine disruptors (e.g., TCC [93], TPhP [94]), antidepressants [91,95], the agricultural chemical boscalid [96], and MPs [97,98], induce oxidative stress in ocular tissues, leading to structural damage and dysfunction of ocular cells.
Oxidative stress also contributes to the production of pro-inflammatory cytokines, which may negatively impact the immune system. Cytokines and chemokines play critical roles in mediating the innate immune response. Pro-inflammatory cytokines such as IL-1β, IL-4, IL-8, TNF-α, and cxcl-clc are recognized as key regulators of the inflammatory process and serve as significant markers during the innate immune response [99]. Furthermore, changes in immune cells have a significant impact on the immunological response. Macrophages in zebrafish act as phagocytes and have an intrinsic immunological role in battling infectious pathogens [100]. Zebrafish exposed to TCC alone or co-exposed to MPs and F-53B exhibited increased transcript levels of pro-inflammatory cytokines and chemokines [99,101]. Notably, TCC directly inhibits macrophage proliferation, thereby reducing the resistance of ocular tissues to infection and increasing the likelihood of lesions [93,99].

4.2. Apoptosis

Apoptosis represents a prevalent mechanism underlying retinal ganglion and photoreceptor cell damage [56]. Environmental contaminants, including TPhP [94], F-53B [69], BPS [56], PCB1254 [102], Phe [46], crude oil [61], and BDE99 [16], have been demonstrated to induce cell apoptosis. Specifically, TPhP [94], F-53B [69], and BPS [56] have been shown to enhance cell apoptosis in the ocular region of zebrafish, ultimately causing visual impairment via the mitochondria-mediated cell death pathway.

4.3. Aberrant Gene Expression and Epigenetic Alterations

Multiple environmental contaminants have been demonstrated to have an impact on the structure and morphology of the retina, causing morphological defects and changes in the thickness of different retinal layers. These changes include disruptions in the arrangement of photoreceptor cells in the ONL of the retina [56], as well as decreases or increases in thickness and the loss of retinal ganglion cells [46]. Consequently, these changes can lead to a decrease or impairment in vision [16,69,92,103]. These alterations are likely attributed to abnormal gene expression, which affects the functionality and stability of ocular cells, consequently causing damage to ocular tissues. Previous studies have suggested that a decrease in ONL thickness may be associated with the down-regulation of the optin gene in optic cones [62]. Furthermore, the down-regulation of genes crucial for controlling eye morphology and retinal development can impact the structure and development of the eye and retina, ultimately resulting in the occurrence of ocular malformations [104].
Previous investigations have provided evidence indicating the influence of a diverse range of environmental contaminants on the genetic expression pertinent to retinal development and phototransduction in zebrafish. These contaminants encompass a range of EDCs such as PBDE-47 [77], DE-71 [63,103], BDE99 [16], Phe 6, crude oil [61], PCB1254 [15], BPS [56], TCC [62,105], and F-53B [69]. Additionally, the brominated flame retardant TPhP [44,94], the agricultural chemical boscalid [17], and PPCPs such as amitriptyline [80], venlafaxine [80], sertraline [80], and AVO [86,87], as well as microplastics [86,87,106] have demonstrated their ability to modulate the expression of genes closely associated with visual development in zebrafish. Consequently, these alterations in gene expression have been observed to induce notable changes in the visual behavior of fish. Table 7 summarizes the effects of the abovementioned environmental contaminants on genes related to visual development.

5. Challenges and Prospects

Zebrafish serve as a valuable model organism for investigating visual systems due to their resemblance to other vertebrates in terms of their visual system [21]. The assessment of zebrafish visual behavior through quantitative analysis provides insights into their response to visual stimuli, allowing for noninvasive detection of potential eye lesions, identification of toxic effects on vision, and rapid evaluation of the impacts of pollutants on the eye and visual system. However, the behavioral response does possess certain challenges in the evaluation of ocular toxicity in zebrafish. These techniques are unable to directly elucidate the underlying cellular and molecular processes, and there is a lack of standardized protocols for behavioral testing. Additionally, zebrafish models cannot entirely replace mammalian models. Therefore, it is crucial to consider and address these challenges fully when using zebrafish models for behavioral assessment of ocular toxicity.
(1)
The direct correlation between ocular toxicity and biological tissue levels, including genes, proteins, tissues, and physiology, cannot be readily determined when using behavioral responses to assess the potential toxicity of environmental contaminants [32]. Therefore, further quantitative investigations employing alternative methodologies are required to establish whether the observed damage to the visual system is directly attributed to the pollutants under scrutiny. In vision science research, the up-regulated or down-regulated trend of key visual pathways can be verified through molecular detection of gene and protein expression related to retinal development and function. Additionally, histological analysis using microscopy and physiological analysis through electroretinograms can serve to corroborate these trends. Further research is necessary to determine whether environmental contaminants have genetic effects on visual system toxicity in zebrafish;
(2)
Although many visually mediated behavior testing methods have been applied to zebrafish research, there is currently a lack of unified standards for each method in actual application. Therefore, it is crucial to establish zebrafish behavioral standards under the guidance of vision. For example, in behavioral experiments, it is essential to standardize key experimental parameters such as light intensity, pollutant co-incubation time, and recording time to improve the repeatability of research results across different teams and strengthen the persuasiveness of conclusions based on zebrafish behavioral data. This will promote the construction of zebrafish behavior test standards;
(3)
Considering the different routes of environmental pollution entry into zebrafish and mammalian visual systems is important when utilizing zebrafish as a predictive model. There is no doubt that environmental pollutants exist in a variety of environmental substrates, including water, sediments, soil, air, and biota. As a result, both zebrafish and human eyes are inevitably exposed to environmental pollutants, which can cause ocular damage [11,124,125,126]. This commonality indicates the potential shared effects of environmental pollutants on the visual system across different species. Researchers can take advantage of the benefits of zebrafish for rapid evaluation, drug screening, and preliminary studies of mechanisms. Nevertheless, multiple animal models, multiple tissue levels, and multi-angle studies are necessary to further clarify the mechanism of ocular toxicity caused by environmental contaminants.

6. Conclusions

Environmental contaminants have recently been the subject of increasing attention due to their potential influence on the visual system, which is a vital sensory organ for the survival, development, and learning activities of organisms. However, the currently used model organisms such as mice limit their usefulness for ocular toxic applications due to their rod-shaped vision, slow development of the visual system, and greater reliance on smell, touch, and hearing. In contrast, zebrafish, which possess cone vision similar to humans, have presented unprecedented advancements in visual system toxicity research. Observing physiological and behavioral changes in zebrafish enables the swift assessment of potential ocular toxicity caused by pollutants, revealing different levels of damage. Future research combining standard visually mediated behaviors with comprehensive quantitative investigations promises new insights into the mechanisms underlying the ocular toxicity of environmental contaminants.

Author Contributions

D.S., D.W. and L.J. contributed to the conception of this review. J.Y., Y.M. and J.M. analyzed the pieces of literature and wrote the manuscript. J.Y., Y.M., J.M. and H.Y. completed the figure drawings. D.S., L.J., D.W., K.Z. and Q.Y. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Zhejiang Medical Association Clinical Research Fund (2021ZYC-A213).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BDE992,2’,4,4’,5-pentabromodiphenyl ether
PBDE-472,2′,4,4′-tetrabromodiphenyl ether
AMIAmitriptyline
AVOAvobenzone
BPSBisphenol S
BFRsBrominated flame retardants
CATCatalase
CPCyclophosphamide
EDCsEndocrine disrupting chemicals
GCLGanglion cell layer
hpfHours post-fertilization
INLInner nuclear layer
IPLInner plexiform layer
MDAMalondialdehyde
MPsMicroplastics
NPsNanoplastics
OKROptokinetic reflex
OMROptomotor response
ONLOuter nuclear layer
OPLOuter plexiform layer
PPCPsPharmaceutical and personal care products
PhePhenanthrene polycyclic aromatic hydrocarbon
PBDEPolybrominated diphenyl ether
PAHsPolycyclic aromatic hydrocarbons
ROSReactive oxygen species
RGCRetinal ganglion cells
RPERetinal pigment epithelium
SERSertraline
SODSuperoxide dismutase
TCCTriclocarban
TPhPTriphenyl phosphate
UVUltraviolet
VENVenlafaxine

References

  1. Plaisancié, J.; Ceroni, F.; Holt, R.; Zazo Seco, C.; Calvas, P.; Chassaing, N.; Ragge, N.K. Genetics of Anophthalmia and Microphthalmia. Part 1: Non-Syndromic Anophthalmia/Microphthalmia. Hum. Genet. 2019, 138, 799–830. [Google Scholar] [CrossRef] [PubMed]
  2. Zhu, S.; Gong, L.; Li, Y.; Xu, H.; Gu, Z.; Zhao, Y. Safety Assessment of Nanomaterials to Eyes: An Important but Neglected Issue. Adv. Sci. 2019, 6, 1802289. [Google Scholar] [CrossRef] [PubMed]
  3. GBD 2019 Blindness and Vision Impairment Collaborators; Vision Loss Expert Group of the Global Burden of Disease Study. Trends in Prevalence of Blindness and Distance and near Vision Impairment over 30 Years: An Analysis for the Global Burden of Disease Study. Lancet Glob. Health 2021, 9, e130–e143. [Google Scholar] [CrossRef] [PubMed]
  4. Zona, A.; Iavarone, I.; Buzzoni, C.; Conti, S.; Santoro, M.; Fazzo, L.; Pasetto, R.; Pirastu, R.; Bruno, C.; Ancona, C.; et al. SENTIERI: Epidemiological Study of Residents in National Priority Contaminated Sites. Fifth Report. Epidemiol. Prev. 2019, 43, 1–208. [Google Scholar] [CrossRef] [PubMed]
  5. Baldacci, S.; Gorini, F.; Santoro, M.; Pierini, A.; Minichilli, F.; Bianchi, F. Environmental and Individual Exposure and the Risk of Congenital Anomalies: A Review of Recent Epidemiological Evidence. Epidemiol. Prev. 2018, 42, 1–34. [Google Scholar] [PubMed]
  6. Dolk, H.; Vrijheid, M. The Impact of Environmental Pollution on Congenital Anomalies. Br. Med. Bull. 2003, 68, 25–45. [Google Scholar] [CrossRef]
  7. Pellacani, C.; Tagliaferri, S.; Caglieri, A.; Goldoni, M.; Giordano, G.; Mutti, A.; Costa, L.G. Synergistic Interactions between PBDEs and PCBs in Human Neuroblastoma Cells. Environ. Toxicol. 2014, 29, 418–427. [Google Scholar] [CrossRef]
  8. Asante, K.A.; Adu-Kumi, S.; Nakahiro, K.; Takahashi, S.; Isobe, T.; Sudaryanto, A.; Devanathan, G.; Clarke, E.; Ansa-Asare, O.D.; Dapaah-Siakwan, S.; et al. Human Exposure to PCBs, PBDEs and HBCDs in Ghana: Temporal Variation, Sources of Exposure and Estimation of Daily Intakes by Infants. Environ. Int. 2011, 37, 921–928. [Google Scholar] [CrossRef]
  9. Polevoy, C.; Arbuckle, T.E.; Oulhote, Y.; Lanphear, B.P.; Cockell, K.A.; Muckle, G.; Saint-Amour, D. Prenatal Exposure to Legacy Contaminants and Visual Acuity in Canadian Infants: A Maternal-Infant Research on Environmental Chemicals Study (MIREC-ID). Environ. Health 2020, 19, 14. [Google Scholar] [CrossRef]
  10. Wigle, D.T.; Arbuckle, T.E.; Turner, M.C.; Bérubé, A.; Yang, Q.; Liu, S.; Krewski, D. Epidemiologic Evidence of Relationships between Reproductive and Child Health Outcomes and Environmental Chemical Contaminants. J. Toxicol. Environ. Health B Crit. Rev. 2008, 11, 373–517. [Google Scholar] [CrossRef]
  11. Chen, L. Visual System: An Understudied Target of Aquatic Toxicology. Aquat. Toxicol. 2020, 225, 105542. [Google Scholar] [CrossRef] [PubMed]
  12. Harada, M. Minamata Disease: Methylmercury Poisoning in Japan Caused by Environmental Pollution. Crit. Rev. Toxicol. 1995, 25, 1–24. [Google Scholar] [CrossRef] [PubMed]
  13. Fillion, M.; Lemire, M.; Philibert, A.; Frenette, B.; Weiler, H.A.; Deguire, J.R.; Guimarães, J.R.D.; Larribe, F.; Barbosa, F.; Mergler, D. Toxic Risks and Nutritional Benefits of Traditional Diet on near Visual Contrast Sensitivity and Color Vision in the Brazilian Amazon. Neurotoxicology 2013, 37, 173–181. [Google Scholar] [CrossRef] [PubMed]
  14. Altmann, L.; Sveinsson, K.; Krämer, U.; Weishoff-Houben, M.; Turfeld, M.; Winneke, G.; Wiegand, H. Visual Functions in 6-Year-Old Children in Relation to Lead and Mercury Levels. Neurotoxicol. Teratol. 1998, 20, 9–17. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, X.; Hong, Q.; Yang, L.; Zhang, M.; Guo, X.; Chi, X.; Tong, M. PCB1254 Exposure Contributes to the Abnormalities of Optomotor Responses and Influence of the Photoreceptor Cell Development in Zebrafish Larvae. Ecotoxicol. Environ. Saf. 2015, 118, 133–138. [Google Scholar] [CrossRef]
  16. Wei, S.; Chen, F.; Xu, T.; Cao, M.; Yang, X.; Zhang, B.; Guo, X.; Yin, D. BDE-99 Disrupts the Photoreceptor Patterning of Zebrafish Larvae via Transcription Factor Six7. Environ. Sci. Technol. 2022, 56, 5673–5683. [Google Scholar] [CrossRef]
  17. Qian, L.; Qi, S.; Wang, Z.; Magnuson, J.T.; Volz, D.C.; Schlenk, D.; Jiang, J.; Wang, C. Environmentally Relevant Concentrations of Boscalid Exposure Affects the Neurobehavioral Response of Zebrafish by Disrupting Visual and Nervous Systems. J. Hazard. Mater. 2021, 404, 124083. [Google Scholar] [CrossRef]
  18. Horzmann, K.A.; Freeman, J.L. Making Waves: New Developments in Toxicology with the Zebrafish. Toxicol. Sci. 2018, 163, 5–12. [Google Scholar] [CrossRef]
  19. Kalueff, A.V.; Stewart, A.M.; Gerlai, R. Zebrafish as an Emerging Model for Studying Complex Brain Disorders. Trends Pharmacol. Sci. 2014, 35, 63–75. [Google Scholar] [CrossRef]
  20. Howe, K.; Clark, M.D.; Torroja, C.F.; Torrance, J.; Berthelot, C.; Muffato, M.; Collins, J.E.; Humphray, S.; McLaren, K.; Matthews, L.; et al. The Zebrafish Reference Genome Sequence and Its Relationship to the Human Genome. Nature 2013, 496, 498–503. [Google Scholar] [CrossRef]
  21. Bilotta, J.; Saszik, S. The Zebrafish as a Model Visual System. Int. J. Dev. Neurosci. 2001, 19, 621–629. [Google Scholar] [CrossRef] [PubMed]
  22. Basnet, R.M.; Zizioli, D.; Taweedet, S.; Finazzi, D.; Memo, M. Zebrafish Larvae as a Behavioral Model in Neuropharmacology. Biomedicines 2019, 7, 23. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, L.; Ma, J.; Wu, W.; Fang, Y.; Liu, F.; Yang, Q.; Hu, X.; Gu, X.; He, Z.; Sun, D.; et al. Effect of Aerobic Exercise as a Treatment on Type 2 Diabetes Mellitus with Depression-like Behavior Zebrafish. Life Sci. 2022, 300, 120578. [Google Scholar] [CrossRef] [PubMed]
  24. Qu, L.; Liu, F.; Fang, Y.; Wang, L.; Chen, H.; Yang, Q.; Dong, H.; Jin, L.; Wu, W.; Sun, D. Improvement in Zebrafish with Diabetes and Alzheimer’s Disease Treated with Pasteurized Akkermansia Muciniphila. Microbiol. Spectr. 2023, 11, e0084923. [Google Scholar] [CrossRef]
  25. Ašmonaitė, G.; Boyer, S.; de Souza, K.B.; Wassmur, B.; Sturve, J. Behavioural Toxicity Assessment of Silver Ions and Nanoparticles on Zebrafish Using a Locomotion Profiling Approach. Aquat. Toxicol. 2016, 173, 143–153. [Google Scholar] [CrossRef]
  26. Legradi, J.; el Abdellaoui, N.; van Pomeren, M.; Legler, J. Comparability of Behavioural Assays Using Zebrafish Larvae to Assess Neurotoxicity. Environ. Sci. Pollut. Res. Int. 2015, 22, 16277–16289. [Google Scholar] [CrossRef]
  27. Shi, Q.; Tsui, M.M.P.; Hu, C.; Lam, J.C.W.; Zhou, B.; Chen, L. Acute Exposure to Triphenyl Phosphate (TPhP) Disturbs Ocular Development and Muscular Organization in Zebrafish Larvae. Ecotoxicol. Environ. Saf. 2019, 179, 119–126. [Google Scholar] [CrossRef]
  28. Sloman, K.A.; McNeil, P.L. Using Physiology and Behaviour to Understand the Responses of Fish Early Life Stages to Toxicants. J. Fish Biol. 2012, 81, 2175–2198. [Google Scholar] [CrossRef]
  29. Richardson, R.; Tracey-White, D.; Webster, A.; Moosajee, M. The Zebrafish Eye—A Paradigm for Investigating Human Ocular Genetics. Eye 2017, 31, 68–86. [Google Scholar] [CrossRef]
  30. Zhao, Y.; Yang, Q.; Liu, D.; Liu, T.; Xing, L. Neurotoxicity of Nanoparticles: Insight from Studies in Zebrafish. Ecotoxicol. Environ. Saf. 2022, 242, 113896. [Google Scholar] [CrossRef]
  31. Shen, C.; Zuo, Z. Zebrafish (Danio Rerio) as an Excellent Vertebrate Model for the Development, Reproductive, Cardiovascular, and Neural and Ocular Development Toxicity Study of Hazardous Chemicals. Environ. Sci. Pollut. Res. Int. 2020, 27, 43599–43614. [Google Scholar] [CrossRef] [PubMed]
  32. Cassar, S.; Dunn, C.; Ramos, M.F. Zebrafish as an Animal Model for Ocular Toxicity Testing: A Review of Ocular Anatomy and Functional Assays. Toxicol. Pathol. 2021, 49, 438–454. [Google Scholar] [CrossRef] [PubMed]
  33. Lei, P.; Zhang, W.; Ma, J.; Xia, Y.; Yu, H.; Du, J.; Fang, Y.; Wang, L.; Zhang, K.; Jin, L.; et al. Advances in the Utilization of Zebrafish for Assessing and Understanding the Mechanisms of Nano-/Microparticles Toxicity in Water. Toxics 2023, 11, 380. [Google Scholar] [CrossRef] [PubMed]
  34. Deeti, S.; O’Farrell, S.; Kennedy, B.N. Early Safety Assessment of Human Oculotoxic Drugs Using the Zebrafish Visualmotor Response. J. Pharmacol. Toxicol. Methods 2014, 69, 1–8. [Google Scholar] [CrossRef]
  35. Salvatore, L.; Stella, J.; Geathers, J.S.; Weber, S.R.; Grillo, M.A.; Barber, A.J.; Sundstrom, J.M.; Grillo, S.L. Neurodegeneration, Neuroprotection and Regeneration in the Zebrafish Retina. Cells 2021, 10, 633. [Google Scholar] [CrossRef]
  36. Noel, N.C.L.; Allison, W.T.; MacDonald, I.M.; Hocking, J.C. Zebrafish and Inherited Photoreceptor Disease: Models and Insights. Prog. Retin. Eye Res. 2022, 91, 101096. [Google Scholar] [CrossRef]
  37. Yoshimatsu, T.; Schröder, C.; Nevala, N.E.; Berens, P.; Baden, T. Fovea-like Photoreceptor Specializations Underlie Single UV Cone Driven Prey-Capture Behavior in Zebrafish. Neuron 2020, 107, 320–337.e6. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Y.J.; Cai, S.J.; Cui, J.L.; Chen, Y.; Tang, X.; Li, Y.H. Correlation between Photoreceptor Injury-Regeneration and Behavior in a Zebrafish Model. Neural Regen. Res. 2017, 12, 795–803. [Google Scholar] [CrossRef]
  39. Hong, Y.; Luo, Y. Zebrafish Model in Ophthalmology to Study Disease Mechanism and Drug Discovery. Pharmaceuticals 2021, 14, 716. [Google Scholar] [CrossRef]
  40. Blanco-Sánchez, B.; Clément, A.; Phillips, J.B.; Westerfield, M. Zebrafish Models of Human Eye and Inner Ear Diseases. Methods Cell Biol. 2017, 138, 415–467. [Google Scholar] [CrossRef]
  41. Bringmann, A.; Pannicke, T.; Grosche, J.; Francke, M.; Wiedemann, P.; Skatchkov, S.N.; Osborne, N.N.; Reichenbach, A. Müller Cells in the Healthy and Diseased Retina. Prog. Retin. Eye Res. 2006, 25, 397–424. [Google Scholar] [CrossRef] [PubMed]
  42. Zang, J.; Neuhauss, S.C.F. Biochemistry and Physiology of Zebrafish Photoreceptors. Pflug. Arch. 2021, 473, 1569–1585. [Google Scholar] [CrossRef]
  43. Harsing, L.G.; Szénási, G.; Zelles, T.; Köles, L. Purinergic–Glycinergic Interaction in Neurodegenerative and Neuroinflammatory Disorders of the Retina. Int. J. Mol. Sci. 2021, 22, 6209. [Google Scholar] [CrossRef]
  44. Shi, Q.; Wang, Z.; Chen, L.; Fu, J.; Han, J.; Hu, B.; Zhou, B. Optical Toxicity of Triphenyl Phosphate in Zebrafish Larvae. Aquat. Toxicol. 2019, 210, 139–147. [Google Scholar] [CrossRef]
  45. Bibliowicz, J.; Tittle, R.K.; Gross, J.M. Toward a Better Understanding of Human Eye Disease Insights from the Zebrafish, Danio Rerio. Prog. Mol. Biol. Transl. Sci. 2011, 100, 287–330. [Google Scholar] [CrossRef]
  46. Huang, L.; Wang, C.; Zhang, Y.; Wu, M.; Zuo, Z. Phenanthrene Causes Ocular Developmental Toxicity in Zebrafish Embryos and the Possible Mechanisms Involved. J. Hazard. Mater. 2013, 261, 172–180. [Google Scholar] [CrossRef]
  47. Zeng, H.; Yang, Q.; Chen, J.; Ding, L.; Rao, F.; Lv, J.; Xie, B.; Xiang, S.; Yu, H.; Chen, X.; et al. Image-Guided Optical Coherence Tomography to Assess Structural Changes in Rodent Retinas. J. Vis. Exp. 2023, 192, e64783. [Google Scholar] [CrossRef]
  48. Conedera, F.M.; Arendt, P.; Trepp, C.; Tschopp, M.; Enzmann, V. Müller Glia Cell Activation in a Laser-Induced Retinal Degeneration and Regeneration Model in Zebrafish. J. Vis. Exp. 2017, 128, 56249. [Google Scholar] [CrossRef]
  49. McNeil, P.L.; Nebot, C.; Cepeda, A.; Sloman, K.A. Environmental Concentrations of Prednisolone Alter Visually Mediated Responses during Early Life Stages of Zebrafish (Danio Rerio). Environ. Pollut. 2016, 218, 981–987. [Google Scholar] [CrossRef]
  50. Bhagat, J.; Zang, L.; Nishimura, N.; Shimada, Y. Zebrafish: An Emerging Model to Study Microplastic and Nanoplastic Toxicity. Sci. Total Environ. 2020, 728, 138707. [Google Scholar] [CrossRef]
  51. Chhetri, J.; Jacobson, G.; Gueven, N. Zebrafish—On the Move towards Ophthalmological Research. Eye 2014, 28, 367–380. [Google Scholar] [CrossRef] [PubMed]
  52. Tao, Y.; Li, Z.; Yang, Y.; Jiao, Y.; Qu, J.; Wang, Y.; Zhang, Y. Effects of Common Environmental Endocrine-Disrupting Chemicals on Zebrafish Behavior. Water Res. 2022, 208, 117826. [Google Scholar] [CrossRef] [PubMed]
  53. Hamilton, T.J.; Krook, J.; Szaszkiewicz, J.; Burggren, W. Shoaling, Boldness, Anxiety-like Behavior and Locomotion in Zebrafish (Danio Rerio) Are Altered by Acute Benzo[a]Pyrene Exposure. Sci. Total Environ. 2021, 774, 145702. [Google Scholar] [CrossRef] [PubMed]
  54. Zhou, S.; Chen, Q.; Di Paolo, C.; Shao, Y.; Hollert, H.; Seiler, T.B. Behavioral Profile Alterations in Zebrafish Larvae Exposed to Environmentally Relevant Concentrations of Eight Priority Pharmaceuticals. Sci. Total Environ. 2019, 664, 89–98. [Google Scholar] [CrossRef] [PubMed]
  55. Portugues, R.; Engert, F. The Neural Basis of Visual Behaviors in the Larval Zebrafish. Curr. Opin. Neurobiol. 2009, 19, 644–647. [Google Scholar] [CrossRef]
  56. Liu, W.; Zhang, X.; Wei, P.; Tian, H.; Wang, W.; Ru, S. Long-Term Exposure to Bisphenol S Damages the Visual System and Reduces the Tracking Capability of Male Zebrafish (Danio Rerio). J. Appl. Toxicol. 2018, 38, 248–258. [Google Scholar] [CrossRef]
  57. Fadool, J.M.; Dowling, J.E. Zebrafish: A Model System for the Study of Eye Genetics. Prog. Retin. Eye Res. 2008, 27, 89–110. [Google Scholar] [CrossRef]
  58. Easter, S.S.; Nicola, G.N. The Development of Eye Movements in the Zebrafish (Danio Rerio). Dev. Psychobiol. 1997, 31, 267–276. [Google Scholar] [CrossRef]
  59. Richards, F.M.; Alderton, W.K.; Kimber, G.M.; Liu, Z.; Strang, I.; Redfern, W.S.; Valentin, J.P.; Winter, M.J.; Hutchinson, T.H. Validation of the Use of Zebrafish Larvae in Visual Safety Assessment. J. Pharmacol. Toxicol. Methods 2008, 58, 50–58. [Google Scholar] [CrossRef]
  60. Tierney, K.B. Behavioural Assessments of Neurotoxic Effects and Neurodegeneration in Zebrafish. Biochim. Biophys. Acta 2011, 1812, 381–389. [Google Scholar] [CrossRef]
  61. Magnuson, J.T.; Bautista, N.M.; Lucero, J.; Lund, A.K.; Xu, E.G.; Schlenk, D.; Burggren, W.W.; Roberts, A.P. Exposure to Crude Oil Induces Retinal Apoptosis and Impairs Visual Function in Fish. Environ. Sci. Technol. 2020, 54, 2843–2850. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, X.F.; Chen, Z.F.; Lin, Z.C.; Liao, X.L.; Zou, T.; Qi, Z.; Cai, Z. Toxic Effects of Triclocarban on Larval Zebrafish: A Focus on Visual Dysfunction. Aquat. Toxicol. 2021, 241, 106013. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, L.; Huang, Y.; Huang, C.; Hu, B.; Hu, C.; Zhou, B. Acute Exposure to DE-71 Causes Alterations in Visual Behavior in Zebrafish Larvae. Environ. Toxicol. Chem. 2013, 32, 1370–1375. [Google Scholar] [CrossRef] [PubMed]
  64. Maaswinkel, H.; Li, L. Spatio-Temporal Frequency Characteristics of the Optomotor Response in Zebrafish. Vis. Res. 2003, 43, 21–30. [Google Scholar] [CrossRef]
  65. LeFauve, M.K.; Rowe, C.J.; Crowley-Perry, M.; Wiegand, J.L.; Shapiro, A.G.; Connaughton, V.P. Using a Variant of the Optomotor Response as a Visual Defect Detection Assay in Zebrafish. J. Biol. Methods 2021, 8, e144. [Google Scholar] [CrossRef] [PubMed]
  66. Zaluski, A.B.; Wiprich, M.T.; de Almeida, L.F.; de Azevedo, A.P.; Bonan, C.D.; Vianna, M.R.M. Atrazine and Diuron Effects on Survival, Embryo Development, and Behavior in Larvae and Adult Zebrafish. Front. Pharmacol. 2022, 13, 841826. [Google Scholar] [CrossRef]
  67. Huang, L.; Zuo, Z.; Zhang, Y.; Wu, M.; Lin, J.J.; Wang, C. Use of Toxicogenomics to Predict the Potential Toxic Effect of Benzo(a)Pyrene on Zebrafish Embryos: Ocular Developmental Toxicity. Chemosphere 2014, 108, 55–61. [Google Scholar] [CrossRef]
  68. Dehnert, G.K.; Karasov, W.H.; Wolman, M.A. 2,4-Dichlorophenoxyacetic Acid Containing Herbicide Impairs Essential Visually Guided Behaviors of Larval Fish. Aquat. Toxicol. 2019, 209, 1–12. [Google Scholar] [CrossRef]
  69. Wu, L.; Zeeshan, M.; Dang, Y.; Liang, L.Y.; Gong, Y.C.; Li, Q.Q.; Tan, Y.W.; Fan, Y.Y.; Lin, L.Z.; Zhou, Y.; et al. Environmentally Relevant Concentrations of F-53B Induce Eye Development Disorders-Mediated Locomotor Behavior in Zebrafish Larvae. Chemosphere 2022, 308, 136130. [Google Scholar] [CrossRef]
  70. Qiu, L.; Wei, S.; Yang, Y.; Zhang, R.; Ru, S.; Zhang, X. Mechanism of Bisphenol S Exposure on Color Sensitivity of Zebrafish Larvae. Environ. Pollut. 2023, 316, 120670. [Google Scholar] [CrossRef]
  71. Comai, S.; De Gregorio, D.; Posa, L.; Ochoa-Sanchez, R.; Bedini, A.; Gobbi, G. Dysfunction of Serotonergic Activity and Emotional Responses across the Light-Dark Cycle in Mice Lacking Melatonin MT2 Receptors. J. Pineal Res. 2020, 69, e12653. [Google Scholar] [CrossRef]
  72. Lahouel, A.; Kebieche, M.; Lakroun, Z.; Rouabhi, R.; Fetoui, H.; Chtourou, Y.; Djamila, Z.; Soulimani, R. Neurobehavioral Deficits and Brain Oxidative Stress Induced by Chronic Low Dose Exposure of Persistent Organic Pollutants Mixture in Adult Female Rat. Environ. Sci. Pollut. Res. Int. 2016, 23, 19030–19040. [Google Scholar] [CrossRef]
  73. Hwang, K.S.; Son, Y.; Kim, S.S.; Shin, D.S.; Lim, S.H.; Yang, J.Y.; Jeong, H.N.; Lee, B.H.; Bae, M.A. Size-Dependent Effects of Polystyrene Nanoparticles (PS-NPs) on Behaviors and Endogenous Neurochemicals in Zebrafish Larvae. Int. J. Mol. Sci. 2022, 23, 10682. [Google Scholar] [CrossRef]
  74. Tang, Y.; Mi, P.; Li, M.; Zhang, S.; Li, J.; Feng, X. Environmental Level of the Antidepressant Venlafaxine Induces Behavioral Disorders through Cortisol in Zebrafish Larvae (Danio Rerio). Neurotoxicol Teratol. 2021, 83, 106942. [Google Scholar] [CrossRef]
  75. Wang, X.H.; Souders, C.L.; Xavier, P.; Li, X.Y.; Yan, B.; Martyniuk, C.J. The Pyrethroid Esfenvalerate Induces Hypoactivity and Decreases Dopamine Transporter Expression in Embryonic/Larval Zebrafish (Danio Rerio). Chemosphere 2020, 243, 125416. [Google Scholar] [CrossRef]
  76. Maharaj, S.; El Ahmadie, N.; Rheingold, S.; El Chehouri, J.; Yang, L.; Souders, C.L.; Martyniuk, C.J. Sub-Lethal Toxicity Assessment of the Phenylurea Herbicide Linuron in Developing Zebrafish (Danio Rerio) Embryo/Larvae. Neurotoxicol. Teratol. 2020, 81, 106917. [Google Scholar] [CrossRef]
  77. Zhao, J.; Xu, T.; Yin, D.Q. Locomotor Activity Changes on Zebrafish Larvae with Different 2,2′,4,4′-Tetrabromodiphenyl Ether (PBDE-47) Embryonic Exposure Modes. Chemosphere 2014, 94, 53–61. [Google Scholar] [CrossRef]
  78. Wang, Y.; Chen, J.; Du, C.; Li, C.; Huang, C.; Dong, Q. Characterization of Retinoic Acid-Induced Neurobehavioral Effects in Developing Zebrafish. Environ. Toxicol. Chem. 2014, 33, 431–437. [Google Scholar] [CrossRef]
  79. Wei, S.; Qiu, L.; Ru, S.; Yang, Y.; Wang, J.; Zhang, X. Bisphenol S Disrupts Opsins Gene Expression and Impairs the Light-Sensing Function via Antagonizing TH-TRβ Signaling Pathway in Zebrafish Larvae. Food Chem. Toxicol. 2023, 172, 113588. [Google Scholar] [CrossRef]
  80. Sehonova, P.; Hodkovicova, N.; Urbanova, M.; Örn, S.; Blahova, J.; Svobodova, Z.; Faldyna, M.; Chloupek, P.; Briedikova, K.; Carlsson, G. Effects of Antidepressants with Different Modes of Action on Early Life Stages of Fish and Amphibians. Environ. Pollut. 2019, 254, 112999. [Google Scholar] [CrossRef]
  81. Li, D.; Sun, W.; Chen, H.; Lei, H.; Li, X.; Liu, H.; Huang, G.-Y.; Shi, W.J.; Ying, G.G.; Luo, Y.; et al. Cyclophosphamide Affects Eye Development and Locomotion in Zebrafish (Danio Rerio). Sci. Total Environ. 2022, 805, 150460. [Google Scholar] [CrossRef] [PubMed]
  82. Ivantsova, E.; Konig, I.; Souders, C.L.; McNabney, D.; Simmons, D.D.B.; Martyniuk, C.J. Lipidomic, Metabolomic, and Behavior Responses of Zebrafish (Danio Rerio) Exposed to Environmental Levels of the Beta Blocker Atenolol. Sci. Total Environ. 2023, 866, 161272. [Google Scholar] [CrossRef] [PubMed]
  83. Saint-Amant, L.; Drapeau, P. Time Course of the Development of Motor Behaviors in the Zebrafish Embryo. J. Neurobiol. 1998, 37, 622–632. [Google Scholar] [CrossRef]
  84. Thiagarajan, V.; Alex, S.A.; Seenivasan, R.; Chandrasekaran, N.; Mukherjee, A. Interactive Effects of Micro/Nanoplastics and Nanomaterials/Pharmaceuticals: Their Ecotoxicological Consequences in the Aquatic Systems. Aquat. Toxicol. 2021, 232, 105747. [Google Scholar] [CrossRef] [PubMed]
  85. Gaylarde, C.C.; Baptista Neto, J.A.; da Fonseca, E.M. Nanoplastics in Aquatic Systems—Are They More Hazardous than Microplastics? Environ. Pollut. 2021, 272, 115950. [Google Scholar] [CrossRef]
  86. Liu, Y.; Wang, Y.; Ling, X.; Yan, Z.; Wu, D.; Liu, J.; Lu, G. Effects of Nanoplastics and Butyl Methoxydibenzoylmethane on Early Zebrafish Embryos Identified by Single-Cell RNA Sequencing. Environ. Sci. Technol. 2021, 55, 1885–1896. [Google Scholar] [CrossRef]
  87. Liu, Y.; Wang, Y.; Li, N.; Jiang, S. Avobenzone and Nanoplastics Affect the Development of Zebrafish Nervous System and Retinal System and Inhibit Their Locomotor Behavior. Sci. Total Environ. 2022, 806, 150681. [Google Scholar] [CrossRef]
  88. Ranasinghe, P.; Thorn, R.J.; Seto, R.; Creton, R.; Bridges, W.C.; Chapman, S.C.; Lee, C.M. Embryonic Exposure to 2,2′,3,5′,6-Pentachlorobiphenyl (PCB-95) Causes Developmental Malformations in Zebrafish. Environ. Toxicol. Chem. 2020, 39, 162–170. [Google Scholar] [CrossRef]
  89. Santos, D.; Luzio, A.; Matos, C.; Bellas, J.; Monteiro, S.M.; Félix, L. Microplastics Alone or Co-Exposed with Copper Induce Neurotoxicity and Behavioral Alterations on Zebrafish Larvae after a Subchronic Exposure. Aquat. Toxicol. 2021, 235, 105814. [Google Scholar] [CrossRef]
  90. Alhasani, R.H.; Zhou, X.; Biswas, L.; Li, X.; Reilly, J.; Zeng, Z.; Shu, X. Gypenosides Attenuate Retinal Degeneration in a Zebrafish Retinitis Pigmentosa Model. Exp. Eye Res. 2020, 201, 108291. [Google Scholar] [CrossRef]
  91. Yang, H.; Gu, X.; Chen, H.; Zeng, Q.; Mao, Z.; Jin, M.; Li, H.; Ge, Y.; Zha, J.; Martyniuk, C.J. Transcriptome Profiling Reveals Toxicity Mechanisms Following Sertraline Exposure in the Brain of Juvenile Zebrafish (Danio Rerio). Ecotoxicol. Environ. Saf. 2022, 242, 113936. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, Y.P.; Hong, Q.; Qin, D.; Kou, C.Z.; Zhang, C.M.; Guo, M.; Guo, X.R.; Chi, X.; Tong, M.L. Effects of Embryonic Exposure to Polychlorinated Biphenyls on Zebrafish (Danio Rerio) Retinal Development. J. Appl. Toxicol. 2012, 32, 186–193. [Google Scholar] [CrossRef] [PubMed]
  93. Vimalkumar, K.; Seethappan, S.; Pugazhendhi, A. Fate of Triclocarban (TCC) in Aquatic and Terrestrial Systems and Human Exposure. Chemosphere 2019, 230, 201–209. [Google Scholar] [CrossRef]
  94. Zhang, Q.; Zheng, S.; Shi, X.; Luo, C.; Huang, W.; Lin, H.; Peng, J.; Tan, W.; Wu, K. Neurodevelopmental Toxicity of Organophosphate Flame Retardant Triphenyl Phosphate (TPhP) on Zebrafish (Danio Rerio) at Different Life Stages. Environ. Int. 2023, 172, 107745. [Google Scholar] [CrossRef]
  95. Yang, M.; Qiu, W.; Chen, J.; Zhan, J.; Pan, C.; Lei, X.; Wu, M. Growth Inhibition and Coordinated Physiological Regulation of Zebrafish (Danio Rerio) Embryos upon Sublethal Exposure to Antidepressant Amitriptyline. Aquat. Toxicol. 2014, 151, 68–76. [Google Scholar] [CrossRef]
  96. Wang, H.; Meng, Z.; Liu, F.; Zhou, L.; Su, M.; Meng, Y.; Zhang, S.; Liao, X.; Cao, Z.; Lu, H. Characterization of Boscalid-Induced Oxidative Stress and Neurodevelopmental Toxicity in Zebrafish Embryos. Chemosphere 2020, 238, 124753. [Google Scholar] [CrossRef]
  97. Pitt, J.A.; Kozal, J.S.; Jayasundara, N.; Massarsky, A.; Trevisan, R.; Geitner, N.; Wiesner, M.; Levin, E.D.; Di Giulio, R.T. Uptake, Tissue Distribution, and Toxicity of Polystyrene Nanoparticles in Developing Zebrafish (Danio Rerio). Aquat. Toxicol. 2018, 194, 185–194. [Google Scholar] [CrossRef]
  98. Sarasamma, S.; Audira, G.; Siregar, P.; Malhotra, N.; Lai, Y.-H.; Liang, S.-T.; Chen, J.-R.; Chen, K.H.-C.; Hsiao, C.-D. Nanoplastics Cause Neurobehavioral Impairments, Reproductive and Oxidative Damages, and Biomarker Responses in Zebrafish: Throwing up Alarms of Wide Spread Health Risk of Exposure. Int. J. Mol. Sci. 2020, 21, 1410. [Google Scholar] [CrossRef]
  99. Wei, J.; Zhou, T.; Hu, Z.; Li, Y.; Yuan, H.; Zhao, K.; Zhang, H.; Liu, C. Effects of Triclocarban on Oxidative Stress and Innate Immune Response in Zebrafish Embryos. Chemosphere 2018, 210, 93–101. [Google Scholar] [CrossRef]
  100. Xu, H.; Dong, X.; Zhang, Z.; Yang, M.; Wu, X.; Liu, H.; Lao, Q.; Li, C. Assessment of Immunotoxicity of Dibutyl Phthalate Using Live Zebrafish Embryos. Fish Shellfish. Immunol. 2015, 45, 286–292. [Google Scholar] [CrossRef]
  101. Yang, H.; Lai, H.; Huang, J.; Sun, L.; Mennigen, J.A.; Wang, Q.; Liu, Y.; Jin, Y.; Tu, W. Polystyrene Microplastics Decrease F–53B Bioaccumulation but Induce Inflammatory Stress in Larval Zebrafish. Chemosphere 2020, 255, 127040. [Google Scholar] [CrossRef]
  102. Zhang, X.; Zhang, Q.; Jiang, Y.; Zhang, S.; Hong, Q.; Guo, X.; Chi, X.; Tong, M. Expression and Significance of MiR—20b in Retinal Photoreceptor Cells Exposed to PCB1254. Aging 2019, 11, 8969–8981. [Google Scholar] [CrossRef]
  103. Xu, T.; Chen, L.; Hu, C.; Zhou, B. Effects of Acute Exposure to Polybrominated Diphenyl Ethers on Retinoid Signaling in Zebrafish Larvae. Environ. Toxicol. Pharmacol. 2013, 35, 13–20. [Google Scholar] [CrossRef]
  104. Roy, N.M.; Carneiro, B.; Ochs, J. Glyphosate Induces Neurotoxicity in Zebrafish. Environ. Toxicol. Pharmacol. 2016, 42, 45–54. [Google Scholar] [CrossRef]
  105. Caioni, G.; Merola, C.; Bertolucci, C.; Lucon-Xiccato, T.; Savaşçı, B.B.; Massimi, M.; Colasante, M.; Fioravanti, G.; Cacciola, N.A.; Ippoliti, R.; et al. Early-Life Exposure to Environmentally Relevant Concentrations of Triclocarban Impairs Ocular Development in Zebrafish Larvae. Chemosphere 2023, 324, 138348. [Google Scholar] [CrossRef]
  106. Torres-Ruiz, M.; De la Vieja, A.; de Alba Gonzalez, M.; Esteban Lopez, M.; Castaño Calvo, A.; Cañas Portilla, A.I. Toxicity of Nanoplastics for Zebrafish Embryos, What We Know and Where to Go Next. Sci. Total Environ. 2021, 797, 149125. [Google Scholar] [CrossRef]
  107. Xu, T.; Zhao, J.; Yin, D.; Zhao, Q.; Dong, B. High-Throughput RNA Sequencing Reveals the Effects of 2,2′,4,4′-Tetrabromodiphenyl Ether on Retina and Bone Development of Zebrafish Larvae. BMC Genom. 2015, 16, 23. [Google Scholar] [CrossRef]
  108. Palacio-Cortés, A.M.; Horton, A.A.; Newbold, L.; Spurgeon, D.; Lahive, E.; Pereira, M.G.; Grassi, M.T.; Moura, M.O.; Disner, G.R.; Cestari, M.M.; et al. Accumulation of Nylon Microplastics and Polybrominated Diphenyl Ethers and Effects on Gut Microbial Community of Chironomus Sancticaroli. Sci. Total Environ. 2022, 832, 155089. [Google Scholar] [CrossRef]
  109. PubChem 2,2′,4,4′,5-Pentabromodiphenyl Ether. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/36159 (accessed on 28 July 2023).
  110. PubChem Hazardous Substances Data Bank (HSDB): 2166. Available online: https://pubchem.ncbi.nlm.nih.gov/source/hsdb/2166#section=LogP (accessed on 28 July 2023).
  111. PubChem Hazardous Substances Data Bank (HSDB): 7836. Available online: https://pubchem.ncbi.nlm.nih.gov/source/hsdb/7836 (accessed on 28 July 2023).
  112. PubChem Concept 53623. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Polychlorinated-Biphenyls (accessed on 28 July 2023).
  113. PubChem 4,4′-Sulfonyldiphenol. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/6626 (accessed on 28 July 2023).
  114. PubChem Triclocarban. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/7547 (accessed on 28 July 2023).
  115. Wang, S.; Huang, J.; Yang, Y.; Hui, Y.; Ge, Y.; Larssen, T.; Yu, G.; Deng, S.; Wang, B.; Harman, C. First Report of a Chinese PFOS Alternative Overlooked for 30 Years: Its Toxicity, Persistence, and Presence in the Environment. Environ. Sci. Technol. 2013, 47, 10163–10170. [Google Scholar] [CrossRef]
  116. PubChem Triphenyl Phosphate. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/8289 (accessed on 28 July 2023).
  117. PubChem Boscalid. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/213013 (accessed on 28 July 2023).
  118. PubChem Amitriptyline. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/2160 (accessed on 28 July 2023).
  119. PubChem Venlafaxine. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/5656 (accessed on 28 July 2023).
  120. PubChem Sertraline. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/68617 (accessed on 28 July 2023).
  121. PubChem Avobenzone. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/51040 (accessed on 28 July 2023).
  122. Gigault, J.; ter Halle, A.; Baudrimont, M.; Pascal, P.-Y.; Gauffre, F.; Phi, T.-L.; El Hadri, H.; Grassl, B.; Reynaud, S. Current Opinion: What Is a Nanoplastic? Environ. Pollut. 2018, 235, 1030–1034. [Google Scholar] [CrossRef]
  123. Chen, Q.; Gundlach, M.; Yang, S.; Jiang, J.; Velki, M.; Yin, D.; Hollert, H. Quantitative Investigation of the Mechanisms of Microplastics and Nanoplastics toward Zebrafish Larvae Locomotor Activity. Sci. Total Environ. 2017, 584–585, 1022–1031. [Google Scholar] [CrossRef]
  124. Zhang, Z.N.; Yang, D.L.; Liu, H.; Bi, J.; Bao, Y.B.; Ma, J.Y.; Zheng, Q.X.; Cui, D.L.; Chen, W.; Xiang, P. Effects of TCPP and TCEP Exposure on Human Corneal Epithelial Cells: Oxidative Damage, Cell Cycle Arrest, and Pyroptosis. Chemosphere 2023, 331, 138817. [Google Scholar] [CrossRef] [PubMed]
  125. Boucher, O.; Burden, M.J.; Muckle, G.; Saint-Amour, D.; Ayotte, P.; Dewailly, É.; Nelson, C.A.; Jacobson, S.W.; Jacobson, J.L. Response Inhibition and Error Monitoring during a Visual Go/No-Go Task in Inuit Children Exposed to Lead, Polychlorinated Biphenyls, and Methylmercury. Environ. Health Perspect. 2012, 120, 608–615. [Google Scholar] [CrossRef] [PubMed]
  126. Zeeshan, M.; Yang, Y.; Zhou, Y.; Huang, W.; Wang, Z.; Zeng, X.Y.; Liu, R.Q.; Yang, B.Y.; Hu, L.W.; Zeng, X.W.; et al. Incidence of Ocular Conditions Associated with Perfluoroalkyl Substances Exposure: Isomers of C8 Health Project in China. Environ. Int. 2020, 137, 105555. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Assessing the ocular toxicity of environmental contaminants through visually mediated zebrafish behavioral studies. (Brominated flame retardants, BFRs; Microplastics, MPs; Pharmaceutical and personal care products, PPCPs; Endocrine disrupting chemicals, EDCs).
Figure 1. Assessing the ocular toxicity of environmental contaminants through visually mediated zebrafish behavioral studies. (Brominated flame retardants, BFRs; Microplastics, MPs; Pharmaceutical and personal care products, PPCPs; Endocrine disrupting chemicals, EDCs).
Toxics 11 00706 g001
Figure 2. Structural similarities between the human and zebrafish retina.
Figure 2. Structural similarities between the human and zebrafish retina.
Toxics 11 00706 g002
Figure 3. Visually mediated behavioral paradigm in zebrafish.
Figure 3. Visually mediated behavioral paradigm in zebrafish.
Toxics 11 00706 g003
Figure 4. Potential mechanisms of eye damage from environmental contaminants in zebrafish.
Figure 4. Potential mechanisms of eye damage from environmental contaminants in zebrafish.
Toxics 11 00706 g004
Table 7. The effects of the abovementioned environmental contaminants on genes related to visual development.
Table 7. The effects of the abovementioned environmental contaminants on genes related to visual development.
CategoriesCompoundsWater SolubilityExposure DoseExposure TimeChanges in Genes Associated with Visual DevelopmentRef.
EDCsPBDE-47 Insoluble in water500 μg/L/rx2, opn1sw1, opn1mw1, opn1sw2 ↓; opn1lw1 ↑[107,108]
DE-71water-soluble0.32, 3.58, and 31.0 μg/Lfrom 2 hpf to 15 dpfzfrho, zfgr1 ↑ (3.58, and 31.0 μg/L)[63]
BDE-999e-10 mg/mL5, 50 μg/Lfrom 3 hpf to 120 hpfopn1sw1, opn1sw2, opn1mw1, opn1mw2,
opn1lw2, rho
[16,109]
Phe1.10 mg/L3.56, 35.6 and 356 μg/Lfrom 2 hpf to 72 hpf Zeb1 ↓, Mitf[46,110]
Crude oil6.6e-3 mg/L12.8, 38.5, 64.2, and 89.8 μg/Lfrom 4 hpf to 72 hpf arr3b, gnat2, opn1mw, rgr, rho, rpe65a, sws1 ↓ (38.5, 64.2 and 89.8 μg/L); crx, pde6c, pde6h ↓ (64.2, 89.8 μg/L)[62,111]
PCB1254extremely low0.125, 0.25, 0.5 and 1 mg/L7 days of continuous exposureSWS2 ↓ (0.125,0.25,0.5 and 1 mg/L); CRX, RHO, SWS1 ↓ (0.5, 1 mg/L)[15,112]
BPSInsoluble in water1, 10, 100 and 1000 μg/Lfrom 2 hpf to 120 dpfzfrho ↑ (1,10 μg/L); zfblue ↑ (1000 μg/L); zfred, zfgr1 ↑ (1, 10, 100, 1000 μg/L)[57,113]
4400 nM from 2 hpf to 24 hpf opn1sw1, arr3a, pde6h, gk1b ↑, opn1mw2, opn1lw1, opn1lw2, rho, pde6a ↓ (4 nM); opn1sw1, opn1sw2, opn1mw1, opn1mw2, opn1mw3, opn1lw1, opn1lw2, arr3a, pde6h, gk1b ↓, rho, pde6a ↑ (400 nM)[79]
TCC0.11 mg/L0.20, 40.0 µg/Lfrom 2 hpf to 6 dpfopn1sw2, opn1mw1, opn1mw3, opn1lw1, opn1lw2, rho[62,114]
F-53B200 mg/L0.15, 1.5 and 15 μg/Lfrom 2 hpf to 120 hpfaldh1a2, cryaa, crybb, crygn, mipa, pax6, rx1, gant1, rho, opn1sw, opn1lw[69,115]
BFRsTPhPInsoluble in water4, 100 μg/Lfrom 2 hpf to 144 hpfzfrho, zfred, zfgr1, zfuv, zfblue, rhodopsin ↓ (100 μg/L); retinoschisin 1a ↓ (4 μg/L)[27,116]
0.1, 1, 10, and 30 μg/Lfrom 2 to 144 hpfzfrho, opn1sw1, opn1sw2, opn1mw1, opn1mw2, opn1mw3, opn1mw4, opn1lw1, opn1lw2 ↓[44]
Agricultural chemicalsBoscalid4.6 mg/L0.3, 0.6, and 1.2 mg/Lfrom 2 hpf to 4 dpeopn1sw1, opn1mw1, opn4.1, rho ↑ (0.3 mg/L); opn1sw1, opn1mw1, opn1lw2, opn4xb, opn4.1, rho, rhol ↓ (1.2 mg/L)[17,117]
from 2 hpf to 8 dpe opn1sw1, opn1lw2, opn4xb, rho ↑ (0.3 mg/L)[17]
PPCPsAMI9.71 mg/L0.3, 30 μg/Lto 144 hpfpax 6 ↓ (30 μg/L)[80,118]
VEN267 mg/Lpax 6, otx2[80,119]
SER3.8 mg/mL0.1, 10 μg/Lpax 6 ↓, otx2[80,120]
AVO2.2 mg/L10 μg/Lfrom 2 hpf to 144 hpfsix6 ↓; lhx9, pax2, pax6[87,121]
MPsNPsInsoluble in waterlhx9, six6 ↓; six3, pax2, pax6[87,122]
MPs Insoluble in water1 mg/Lfrom 3 hpf to 1120 hpfZfrho[108,123]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yi, J.; Ma, Y.; Ma, J.; Yu, H.; Zhang, K.; Jin, L.; Yang, Q.; Sun, D.; Wu, D. Rapid Assessment of Ocular Toxicity from Environmental Contaminants Based on Visually Mediated Zebrafish Behavior Studies. Toxics 2023, 11, 706. https://doi.org/10.3390/toxics11080706

AMA Style

Yi J, Ma Y, Ma J, Yu H, Zhang K, Jin L, Yang Q, Sun D, Wu D. Rapid Assessment of Ocular Toxicity from Environmental Contaminants Based on Visually Mediated Zebrafish Behavior Studies. Toxics. 2023; 11(8):706. https://doi.org/10.3390/toxics11080706

Chicago/Turabian Style

Yi, Jia, Yilei Ma, Jiahui Ma, Haiyang Yu, Kun Zhang, Libo Jin, Qinsi Yang, Da Sun, and Dejun Wu. 2023. "Rapid Assessment of Ocular Toxicity from Environmental Contaminants Based on Visually Mediated Zebrafish Behavior Studies" Toxics 11, no. 8: 706. https://doi.org/10.3390/toxics11080706

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop