Open access peer-reviewed chapter

Zebrafish as a High-Throughput In Vivo Model for Testing the Bioactivity of Cannabinoids

Written By

Lee Ellis

Submitted: 22 May 2018 Reviewed: 05 June 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.79321

From the Edited Volume

Recent Advances in Cannabinoid Research

Edited by Willard J Costain and Robert B Laprairie

Chapter metrics overview

1,442 Chapter Downloads

View Full Metrics

Abstract

Zebrafish represent an established vertebrate model system that helps to bridge the research gap between cell line/invertebrate studies and mammalian systems. While the initial testing of tetrahydrocannabinol (THC) using Zebrafish occurred in 1975, zebrafish are currently a burgeoning model for testing the bioactivity of cannabinoids. Zebrafish express both CB1 and CB2 receptors along with all of the other major endocannabinoid-related genes. Zebrafish endocannabinoid gene function has been associated with addiction, anxiety, development, energy homeostasis and food intake, immune system function, learning and memory. Both adult and larval zebrafish have been used to test the therapeutic potential of THC and cannabidiol (CBD) against various disease models such as models of nociception, epilepsy, stress/anxiety and addiction. This chapter will review recent studies that have used zebrafish as a model for testing the bioactivity of cannabinoids and provide insight on potential future work in this area.

Keywords

  • zebrafish
  • cannabinoid
  • pain
  • stress
  • addiction
  • epilepsy

1. Introduction

The use of zebrafish as a vertebrate model for biological research began in the late 1960s in the lab of George Streisinger at the University of Oregon. However, it was not until the middle of the 1980s that a community of researchers working on zebrafish began to emerge. Since that time the use of zebrafish as a model organism has continued to increase. Over the past 3 decades the use of zebrafish as a model species has contributed to our understanding of developmental biology, toxicology, drug efficacy and disease.

As a vertebrate, the zebrafish model provides more information than can be obtained from cell lines and invertebrate studies, while at the same time remaining low-cost and high-throughput compared with mammalian models. It has been estimated that screening 1 drug with rodent models costs approximately 50× more than through zebrafish assays and zebrafish testing can be done in days versus weeks to months for analogous rodent assays [1].

Another major advantage to using zebrafish as a model species is that they show high genetic homology to mammals. The sequencing of the zebrafish genome was begun in 2001 and the reference genome was published in 2013 [2]. This revealed that ~70% of human genes have at least 1 zebrafish ortholog and ~84% of genes known to be associated with human disease have a zebrafish counterpart. This then provides an important platform with which to begin to study genes linked to human disease. The initial studies that made use of zebrafish were largely entrenched in forward genetic screening, which revealed their genetic tractability and helped to lead the way to the generation of clonal lines [3]. While these original studies were begun nearly 40 years ago, since then an ever increasing number of genetic tools have been developed and used to alter the zebrafish genome such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR) [4, 5]. These tools along with a fully sequenced genome provide a stage for the creation of any number of informative transgenic and knockdown/knockout lines. Along with this, zebrafish reach maturity by 90 days post fertilization and produce hundreds of eggs per clutch on a weekly basis. As screening for germline transmission is the general bottle neck in the generation of transgenic lines, the high fecundity of zebrafish allows for more rapid screening and development of transgenic lines compared with mammalian models. The use of transgenic models has a broad applicability and can potentially contribute to all facets of zebrafish research.

One of the major advantages of zebrafish as a model species is that their embryos are fertilized and develop externally providing easy access to embryos and larvae. Importantly, all major organs are formed by 1 day post fertilization (dpf) and larvae hatch from their chorion and become free swimming by 3 dpf. Larvae can live off of the nutrition provided by their yolk sac until 5 dpf, at which time they begin to feed. During this period, larvae are largely transparent making the development of organs and body patterning visible. Their rapid development and transparency provides an ideal setting for testing the effects of various compounds on normal development along with their potential acute toxicity. Standard toxicity testing models exist, including the OECD recognized fish embryo toxicity assay (FET) that tests the effects of compound exposure on normal development from 6 to 72 hpf (OECD guideline 236, adopted July 2013). The general and behavioral toxicity (GBT) assay tests the effects of compound exposure on larvae from 72 to 120 hpf [6]. Additionally, the effect of compounds on the larval heart rate has been shown to be a predictive indicator of potential bradycardia related cardiotoxicity [7]. This is important when screening neuroactive compounds as the blockage of numerous ion channels, often the target of neuroactives, can lead to arrhythmias [8]. The toxicity profiling of potential therapeutics at early stages of development allows for the identification of off-target side effects as well as the potential to calculate a therapeutic window when the toxicity profile is compared with the level of compound required to have a positive effect on disease models.

The use of zebrafish in the field of neuroscience continues to increase and a number of recent reviews have highlighted both the strengths and weaknesses of using zebrafish to study neuroactive compounds and brain disorders [9, 10, 11, 12, 13, 14, 15]. The zebrafish brain has many analogous regions to those of higher vertebrates and the complexity of both juvenile and adult zebrafish brains has been well documented [16]. In addition to brain morphology, the neurochemistry and endocrine responses linked to zebrafish neuroactivity is highly homologous to other vertebrates including the same neurotransmitters, receptors, synthetic/metabolic enzymes and hypothalamo-pituitary hormones [9, 10, 11, 15, 17, 18, 19].

It has been demonstrated that zebrafish are sensitive to a large number of neurotropic drugs including: antipsychotics, mood stabilizers, anxiolytics, antidepressants, ethanol, hypnotics, stimulants, hallucinogens, antiepileptics, analgesics and cognitive enhancers [15, 16]. In addition, both adult and larval zebrafish can be used to model numerous neural disorders including pain/nociception, anxiety, stress, PTSD, ADHD, Autism, epilepsy, learning & memory deficits, psychiatric disorders, Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), Parkinson’s disease, schizophrenia, bi-polar disorder, addiction and brain cancer [6, 9, 11, 14, 15, 16, 20, 21, 22, 23, 24, 25]. This provides in vivo models with which to test not only the bioactivity of various neuroactive compounds, but also allows for the testing of their potential efficacy against numerous models of disease. Use of these models can provide an indication of the level of compound required to oppose a disease phenotype, which is required for the calculation of a therapeutic window for new drugs. The disease models also provide a platform for the testing and potential re-purposing of neuroactive compounds currently on the market. Finding an effective treatment for the disease models may help to provide clues to the etiology of human disease and insights into additional therapeutic targets.

Many of the neuronal disease models developed using zebrafish are centered on the assessment of aberrant behavior in both larvae and adults, which each provide their own distinct advantages [9, 11, 15, 16]. One of the major advantages of using larvae over adults stems from their reproducible patterns of behavior and potential to be screened in a high throughput fashion. Activity patterns can be assessed in multi-well plates allowing for up to 96 larvae to be tested simultaneously using benchtop tracking systems. As mentioned, larvae become free swimming between 3 and 5 dpf and develop stereotypical behavioral and stimulus response patterns. These include their response to startling stimuli such as noise, light–dark transitions and touch. Importantly the behavioral activity patterns are highly quantifiable and can be altered by neuroactive compounds with various targets. The assessment of adult behavior, while much lower throughput, does have some advantages over larval testing as it can often provide more intricate behavioral paradigms than can be obtained with larvae. Specifically, adult behavior can be tracked in 3 dimensions and various models of learning and memory, conspecific interactions and place preference exist that are not found for larvae. Many of these models are analogous to rodent behavioral models [16].

In addition to models of behavior, as previously mentioned, larval zebrafish are nearly transparent for their first week of development and a number of transgenic lines exist that completely lack pigment. This provides unparalleled access to an intact vertebrate brain. Numerous studies have used in situ hybridization and immunohistochemistry to map and profile neural activity using indicators such as c-Fos. Recent technical advances have allowed for a more in depth assessment of neuronal activity than is possible in mammalian systems. The assessment of neural activity has been accomplished using genetically encoded calcium indicators and whole brain imaging in immobilized larvae [26]. More recently the assessment of neural activity in freely behaving larvae at near cellular resolution has become possible [27]. The development of this new technology now allows for links to be made between localized brain activity and various behaviors and stimulus response patterns that is currently not possible with mammalian models.

While the original use of zebrafish as a model species was focused on genetics, they are currently contributing ever evolving models to the fields of developmental biology, neuroscience, molecular biology and pharmacology research.

Advertisement

2. Functions of the zebrafish endocannabinoid system

The initial use of zebrafish for testing the toxicity of THC occurred in 1975 [28]. However, it has only been the last 10–15 years that interest in the study of the zebrafish endocannabinoid system (ECS) has begun to grow. As outlined below, the zebrafish ECS shows genetic homology to mammalian systems and is involved in many of the same physiological processes. Importantly, the route of administration for cannabinoids to zebrafish is relatively straight forward as they can be added to the bath solution with either methanol or dimethyl sulfoxide (DMSO) as a solvent.

2.1. Gene expression patterns

The initial sequencing and mapping of the expression pattern of the CB1 receptor (CB1R) in both larvae and adults found that the zebrafish CB1R showed a 69% nucleotide identity and a 73.6% amino acid identity with the human CB1R [29]. Larvae begin to express the CB1R by the 3 somite stage of development [30] and, as expected, show a widespread and distinct expression pattern throughout the CNS (preoptic area, dorsal telencephalon, periventricular hypothalamus, tegmentum and anterior hindbrain) by 48 hpf that continues into adulthood [29, 31]. The general pattern of expression for the CB1R in the adult zebrafish brain appears to be homologous to that of mammals.

Shortly after the cloning of the CB1R zebrafish were found to express two CB2 receptor (CB2R) orthologs that showed 98% genetic identity with each other and a 39% amino acid identity with the human CB2R [32]. Importantly, similar to the CB1R, the expression patterns of the CB2R were homologous to those found in mammals with low levels in the brain and higher levels in the intestine, retina, gills, heart, muscle, pituitary and spleen.

Zebrafish also express the transient receptor potential vanilloid type 1 cation channel (TrpV1) and the G-protein coupled receptor 55 (Gpr55) early in development. Both receptors are known to bind endocannabinoids [33]. The cannabinoid receptor interacting protein (CRIP1A) is also expressed early in development [31].

In addition to the cannabinoid receptor genes, the genes responsible for the synthesis and catabolism of the endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG) begin to become expressed between 1 and 12 hpf and their expression levels continue to increase throughout development. These include the AEA biosynthetic enzymes N-acylphosphatidylethanolamine-selective phospholipase D (NAPE-pld) and αβ-hydrolase domain containing 4 (Abhd4), the AEA catabolic enzyme fatty acid amide hydrolase (faah), the 2-AG biosynthetic enzyme diacylglycerol lipase α (DAGL α) and the 2-AG catabolic enzymes monoglyceride lipase (mgll) and prostaglandin-endoperoxide synthase 2 (ptgs2b) [31, 33]. Importantly the expression of these biosynthetic enzymes is accompanied by an increase in the protein levels of AEA and 2-AG [33]. The tissue distribution pattern of these enzymes is vast with high levels found in the brain, muscle, heart, intestine, eyes and reproductive organs (ovary and testis) of adults (Table 1) [31]. In larvae mgll, dagla and cnr1 are expressed in different regions of the brain and all 3 show some overlap with the expression pattern of CB1R [31, 33].

Distribution
Protein name Abbreviation High levels Low levels
Cannabinoid receptor 1 CB1R Brain Eyes, testis
Cannabinoid receptor 2 CB2R Intestine, eyes, gills, heart, muscle, pituitary, kidney, spleen Brain, testis
Transient receptor potential vanilloid type 1 cation channel TrpV1 Sensory neurons
G protein-coupled receptor 55A GPR55A Brain, spleen, testis
Cannabinoid receptor interacting protein (CRIP1A) Brain, eyes, testis
N-acylphosphatidylethanolamine-selective phospholipase D NAPE-pld All organs
αβ-Hydrolase domain containing 4 Abhd4 Spleen, testis All organs
Fatty acid amide hydrolase faah Brain Skin, testis
Fatty acid amide hydrolase 2a faah2a Brain Intestine, eyes, testis
Diacylglycerol lipase α DAGL α Brain, muscle, kidney, eyes, testis, spleen
Diacylglycerol lipase β DAGL β Brain, muscle, kidney, eyes, testis Spleen
Monoglyceride lipase mgll Brain, kidney, spleen, eyes
Prostaglandin-endoperoxide synthase 2 ptgs2a Skin, spleen, eyes
αβ-Hydrolase domain containing 6b Abhd6b Not detectable
αβ-Hydrolase domain containing 6a Abhd6a Intestine, liver, testis
αβ-Hydrolase domain containing 12 abhd12 Brain, muscles, eyes, reproductive organs Kidney, heart, intestine
Glycerophosphodiester phosphodiesterase1 gde1 All organs
N-acylspihingosine amidohydrolase 1a naaa1a Reproductive organs
Peroxisome proliferator-activated receptor αβ pparab Muscles, spleen, brain, heart, eyes
Peroxisome proliferator-activated receptor γ pparg Muscles, spleen, testis

Table 1.

Organ distribution patterns of cannabinoid related proteins in adult zebrafish.

Adapted from Oltrabella et al. [31].

2.2. Growth and development

The CB1 receptor is present at early developmental stages in mammals and it has been suggested that the ECS may contribute to CNS development, such as axonal elongation, myelination, migration, cell proliferation and synaptogenesis [34].

In zebrafish it has been shown that the developmental expression of the CB1R and Dagl2α occurs at the same time during larval development, suggesting that larvae are able to both synthesize and respond to 2-AG during development [35]. The same study demonstrated that morpholino knockdown of the CB1R expression lead to aberrant patterns of axonal growth. It was subsequently shown that knockdown of Dagl2 alters axon formation in the midbrain-hindbrain region and alters different patterns of behavior which suggests that 2-AG plays a role in axon formation which subsequently affects the control of vision and movement in larvae [33]. Additionally, the highest level of CB1R expression in the developing larvae occurs at the time of hatching, which may suggest that the proper expression of the CB1R is necessary for the increase in movement that is required for the hatching process [30].

In addition to neuronal development, it has also been shown that endocannabinoid signaling is required for normal embryonic liver development and function [36]. Alteration of this normal development appears to impact the structure and function of the adult liver and may impact metabolic homeostasis. It has also been shown that the CB2R plays a role in the production, expansion and migration of hematopoietic stem cells suggesting that it may play a role hematopoiesis during development [37].

2.3. Feeding and lipid metabolism

The consumption of cannabis is well known to stimulate appetite and numerous animal and human studies have detailed the role that the endocannabinoid system plays in appetite regulation, weight gain, energy balance, and lipid metabolism. Rodent models have shown that both the endocannabinoids as well as THC stimulate appetite and can produce hyperphagia [38, 39, 40, 41], while CB1R antagonists can suppress appetite [42, 43]. In humans genetic variations in the CB1 receptor and a dysregulation of the endocannabinoid system have been linked to obesity [44, 45]. The first therapeutic targeting this system that was brought to market was the inverse agonist for the CB1 receptor rimonabant. It was shown to lead to weight loss in overweight subjects and was marketed as a therapeutic for the treatment of obesity [46]. In line with this, it has been shown that CB1 receptor knockout animals are thinner than controls and have less adipose tissue, which is thought to relate to both decreased caloric intake as well as changes in metabolic factors [47]. Less adipose tissue may also be linked to the therapeutic potential of targeting the CB1 receptor in the treatment of obesity, as obesity in humans is linked to hepatic stenosis, which was shown to be reduced by treatment with rimonabant [44]. Since the initial development of rimonabant, the endocannabinoid system has been of interest for the potential role its dysregulation may play in obesity [44]. Unfortunately, the side effect profile of rimonabant resulted in its withdrawal from the marketplace.

Zebrafish provide a model with which to study the role of the endocannabinoid system in appetite regulation and lipid bioaccumulation. Similar to what was found for mammals, rimonabant led to the suppression of feeding in juvenile fish [48]. In larvae it was found that rimonabant exposure led to larger yolk sacs during development, suggesting a decrease in the use of fat stores, which may be related to a decreased appetite. Exposure of adult zebrafish to melatonin, a known regulator of energy homeostasis, suppressed appetite through the downregulation of the CB1R gene expression [49]. It then appears that similar to rodents, modulation of the zebrafish endocannabinoid system can regulate appetite.

Zebrafish are also an established model for the study of lipid biology [50, 51, 52]. With respect to the endocannabinoid system, it has been shown that overexpression of the CB1R in liver leads to hepatic lipid accumulation, while suppression leads to a loss of lipid accumulation during hepatogenesis [53]. It has also been found that bisphenol A exposure produces hepatostenosis in adult zebrafish liver by increasing the liver levels of 2-AG and AEA [33]. Stimulation of the endocannabinoid system through CB1 and CB2 receptor activation can influence lipid deposition during embryogenesis through an up-regulation of the lipoprotein lipase gene [54]. Additionally, exposure to two non-psychoactive cannabinoids, namely CBD and tetrahydrocannabivarin (THCV), can lead to a decrease in intracellular lipid levels in zebrafish yolk along with human hepatocytes and adipocytes [55]. This activity does not appear to be linked to CB1R or TRPV1-R activation, but it may suggest a use for both cannabinoids in the treatment of obesity.

2.4. Learning and memory

The effects of cannabinoids on learning and memory in mammalian models is complex and often depends on the model employed and the neural pathways that are activated. However, cannabinoid exposure has been shown to lead to memory impairments for numerous rodent learning paradigms [56].

Zebrafish also have a number of different learning paradigms that include habituation learning, conditioned place preference, avoidance learning, associative learning and spatial memory tests. These learning paradigms are largely based on appetitive and/or fear conditioning [57]. Importantly, a number of these training models have been used to test the cognitive effects of various psychoactive drugs [58]. As many of these models involve the activation of different neuronal pathways, only some of which express cannabinoid receptors, the role of cannabinoid exposure on the development, retention and recall of memories can vary. One such example is a model of fear learning where adult fish were taught to associate the presentation of the alarm pheromone known as the Schreckstoff substance [59] with the presentation of a red light [60]. The response to Schreckstoff substance typically resulted in an increase in bottom dwelling and an increase in erratic movements, both of which are linked to stressful stimuli. Following training, the fish then respond to the red light stimulus, a previously inert stimulus, by showing a similar pattern of behavior. Pre-exposure to THC reduced, but did not eliminate the bottom dwelling and had only a minor effect on erratic movements [60]. A previous study from the same group evaluating spatial memory and found that THC exposure did not affect associative memory but did impair spatial cognition and memory retrieval [60]. In addition to THC, high levels of CBD also appear to reduce memory retention in a spatial memory test [61]. While the number of studies testing the role of cannabinoids on learning and memory using zebrafish is currently limited, it appears the model has great potential in assessing the role of the endocannabinoid system in multiple aspects of learning and how this can be influenced by various cannabinoids.

Advertisement

3. Developing models

3.1. Pain

The treatment of chronic pain is the largest indication for medical cannabis [62, 63, 64, 65, 66, 67]. This is not surprising given that endocannabinoids are known to act as retrograde transmitters blocking the transmission of pain signals at both GABAergic and glutamatergic synapses [68]. Unfortunately the etiology of pain is vast and thus there is not an all-encompassing treatment for pain. Often, an analgesic is only effective for a subset of patients or can only partially reduce pain, but cannot eliminate it [69, 70]. This often leads to multiple drugs being used in combination, which opens the door to various drug interactions that can lead to a number of potential adverse effects and an increased side effect profile.

It is now widely accepted that zebrafish have similar somatosensory systems to higher vertebrates and they can detect painful stimuli (nociception) [71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83]. The models that have been developed vary and include thermal and chemical stimuli that is either bath applied or focally by injection. The models also make use of both acute and chronic nociceptive stimuli and have been developed using larvae and adults. This then provides a number of platforms with which to test potential analgesics that may have links to different disease etiologies.

Recently, a novel model of nociception has been developed and used to test and compare a number of known therapeutics with THC and CBD. The model made use of a short-term exposure to acetic acid which led to tissue damage on the surface of zebrafish larvae and a distinct, reproducible, activity pattern that appears to indicate a multifaceted nociceptive response [83]. The study revealed that THC and CBD had different effects on the behavioral response pattern that varied from those of the known analgesics. Interestingly, of the compounds tested CBD had the most unique effect increasing the rate at which the larval activity pattern returned to that of controls. This would seem to suggest that CBD shortened the recovery from the nociceptive stimulus. This is consistent with literature that has suggested CBD is a strong candidate for pain management [84, 85, 86]. Importantly, this activity was found at a concentration of CBD that had no effect on baseline activity for controls, suggesting that there would be a low potential for side effects. One of the major issues surrounding the therapeutics currently used for pain management lies in their side effect profile, which is often vast and can range from relatively minor (constipation) to severe (addiction). This is especially evident for opioids, which are the most commonly used therapeutic for chronic pain, but have one of the highest addictive potentials [87]. While more work is required to test the effect of other cannabinoids and extracts on the various zebrafish models of nociception, the initial indications are that zebrafish will be valuable for assessing the efficacy of potential therapeutics for pain management.

3.2. Addiction

Recent data suggests that approximately 9% of individuals that use cannabis show symptoms associated with addiction, including tolerance and withdrawal [88]. Comparatively the rate of dependence for tobacco is 67.5% and for alcohol is 22.7% [89]. Zebrafish represent an underutilized model with which to study the addictive properties of cannabinoids. While it has been demonstrated that zebrafish can be used to study the pathology of addiction to numerous drugs of abuse, including, alcohol, cocaine, morphine, nicotine, amphetamine, diazepam and salvinorin A [90, 91, 92, 93, 94, 95], thus far their use to study the addictive properties of cannabinoids has been minimal. Changes in both larval and adult zebrafish behavior can be linked to numerous phenotypes associated with addiction that include conditioned place preference for drugs of abuse, relapse, changes in social behavior, along with symptoms indicating the development of tolerance and withdrawal [90, 93, 96, 97, 98, 99]. It has also been found that the genetic pathways linked to addiction are highly conserved in zebrafish [100]. Currently, with respect to cannabinoids, only one study has shown that zebrafish larvae develop tolerance to the effects of cannabinoids after chronic exposure [101]. As the levels of THC in cannabis plant strains is varied and the refinement and extraction processes allow for other cannabinoids to be used at higher levels both medicinally and recreationally, there is a need to develop models with which to test the addictive properties of both pure cannabinoids on their own, in combination and as part of a complex mixture or extract. Zebrafish have the potential to be such a model.

3.3. Stress and anxiety

One of the known difficulties in using cannabinoids as therapeutics lies in their effect on stress and anxiety. A sought after symptom of cannabis use is the euphoric feeling that often leads to it being considered an anxiolytic. However, it has been broadly shown that as the levels of cannabinoids (specifically THC) are elevated there can be an increase in anxiety-related effects [102]. This is important not only from a side effect perspective, but also becomes an issue when cannabinoids are used to treat anxiety related disorders such as PTSD.

Zebrafish provide numerous models with which to assess stress responses in both larvae and adults. Measurements such as scototaxis (light-dark preference), thigmotaxis (wall hugging), shoaling and the amount of time spent in the bottom of a tank are used as standard measures of stress. Induction of stress can occur by chemical means such as neuro-hyperactive compounds or exposure to the alarm substance. Stress can also be induced physically by touch or following the placement of a fish in a novel setting (novel tank response). Various visible stimuli can also lead to stress responses such as changes in back ground light/dark levels or the appearance of an image of prey. All of these models seem to activate both unique and overlapping neural pathways and thus could provide insight into the mechanism of action of any potential anxiolytic effect [103, 104, 105]. An example of the use of zebrafish stress models for testing cannabinoids was outlined in a recent paper that evaluated the acute effects of both THC and CBD on larval behavior [106]. Zebrafish larvae show a preference for light and a transition from a light to a dark setting results in an increase in activity in the form of darting type movements which are thought to be a stress response. It was found that while THC reduced the baseline activity in the light, the response to a light-dark transition was still evident. Exposure to CBD had a much different response with almost no effect on the baseline activity in the light accompanied by a concentration-dependant reduction in the light-dark transition until it was eliminated. This may suggest that CBD is showing anxiolytic effects at the levels tested [107].

It is currently felt that there is insufficient evidence to support the use of cannabis for the treatment more complex stress disorders such as PTSD [108]. Recently work has begun to establish zebrafish models of complex disorders such as PTSD [109, 110]. The development of these models will provide additional systems with which to test the efficacy of various cannabinoids and combinations thereof in the treatment of anxiety related disorders and may help provide insight into their etiology.

3.4. Uptake and metabolism

The adsorption and bioavailability of cannabinoids provides a challenge for their use as therapeutics. This is particularly true for orally ingested cannabinoids, which show low and, at times, unpredictable bioavailability [111]. The interaction between various cannabinoids along with their interaction with other therapeutics can affect their bioavailability. This is important since the effects of various cannabinoids can be bimodal (hyperactivity at low concentrations and sedation at higher concentrations). Having the ability to measure their uptake, bioaccumulation and excretion will provide insights into the exact levels found within the fish. Knowing the true concentration response profile based on the amount of compound found within zebrafish may also allow for comparisons to be made to the dose-response patterns found for mammals.

Previous work has shown that testing the uptake, metabolism and secretion of cannabinoids is possible using zebrafish larvae [106]. A number of important findings came from this study. First it was found that common pharmacokinetic cannabinoid metabolites are produced by the zebrafish larvae including the phase 1 and phase 2 metabolites hydroxylated THC (11-hyrdoxy-THC, 8-hydroxy-THC), 11-nor-9-carboxy THC, THC-glucuronide, hydroxyl-CBD and CBD-glucuronide. Both the cannabinoids and their metabolites were found to accumulate in the larvae with the metabolites eventually excreted into the bath. It was also shown that there appeared to be bioaccumulation of the cannabinoids in the larvae and a non-linear increase in the amount found in the larvae compared with the bath levels. The same study also revealed that when THC and CBD were co-administered the levels of metabolites that were produced was altered compared to when they were administered alone. This suggests that the complex chemical composition of various cannabis plant strains will also affect the normal metabolism of the individual cannabinoids. It then appears that it will be important to evaluate the uptake kinetics and metabolism of various cannabis derived compounds both alone and in combination.

3.5. Seizures

Approximately 1% of the world’s population is purported to have epilepsy with 30% of those affected having multi-drug resistant epilepsy. This often leads to the requirement for strong anti-seizure medications or drug cocktails. In general this leads to an ever increasing side effect profile that is often debilitating in and of itself. The treatment of seizures is one of the oldest reported uses of cannabis and it has recently garnered attention along with the use of pure cannabinoids (CBD) for their ability to treat severe forms of refractory childhood epilepsy (i.e. Dravet syndrome [112]). However, to date there still remains some controversy regarding its efficacy, with some groups suggesting there is no concrete evidence that it is effective [113]. While it has been purported that cannabinoids, in particular CBD, can mitigate, to some degree, epileptic seizures, unfortunately, with the exception of the childhood epilepsy study [112], the numerous human studies that have evaluated the effect of cannabinoids on seizures have either been from small sample groups, had insufficient controls or were not blinded, which confounds any potential outcomes of the studies [114]. The study was able to show that there was a reduction in seizure frequency in patients with Dravet syndrome following the addition of CBD to their current prescription regime. The one question that does remain is whether the reduction in seizures was due to the direct effect of CBD or if the effect was due to the effect of CBD on the patient’s current medication.

The lack of high-quality human trials for testing anti-epileptics stem from the difficulty in properly designing and/or interpreting the results of human studies. This is partially due to the fact that most study participants are already on another anti-epileptic drug, which often varies between participants in either the drug target or the dosage. During the course of the clinical trials often the levels of either the cannabinoid or the existing therapeutic have to be modified for an individual in order to resolve issues relating to side effects. This makes the proper grouping of different treatment regimens difficult.

There are currently a number of zebrafish models of epilepsy that have been generated to provide a platform for identifying new seizure medications and potentially to understand the etiology of the disease [115]. For instance, a number of small molecules that target different receptors or ion channels can be used to induce seizures or neural hyperactivity in larvae [103116, 117]. These platforms provide high throughput testing models with multiple etiologies. While CBD has been shown to be effective in the treatment of some forms of epilepsy, the mechanism of action is still largely unknown. The existing zebrafish seizure models provide multiple platforms with which to evaluate both the efficacy and potentially the mechanism of action for cannabinoids in the treatment of epilepsy. The further development of these models will be of great benefit for discerning the true therapeutic potential of various cannabinoids for the treatment of epilepsy.

3.6. Smoke toxicity

Currently the main delivery method for cannabinoids both medicinally and recreationally is by the inhalation of smoke from marijuana cigarettes. This is generally because of the rapid onset of effects compared with other delivery methods, which is beneficial from the perspective of symptom relief and also allows for a level of self-regulated dose control that is not possible with other delivery methods such as edibles, which can often take up to 90 min to reach peak effect [111]. Unfortunately, some of the major caveats to an inhaled product are that dosing is often inconsistent and difficult to titrate and they have the potential to have similar health risks as are found for smoked tobacco [118]. It has been suggested that high doses of THC containing products are associated with an increased risk of developing respiratory infections [119]. However, it has been difficult to establish a clear relationship between smoked cannabis and more severe lung disorders, such as cancer, since tobacco use is often co-morbid with cannabis. While in general the number of smoked cannabis cigarettes is lower on a per day basis, this risk cannot be overlooked. Additionally, the inhalation delivery methods for cannabis smoke are somewhat more diverse than for tobacco and include pipes, water pipes, burning on metal and vaporizers. All of these delivery methods produce smoke with its own set of chemical characteristics that depend on the temperature at which the smoke was created and any filtering that occurred before the smoke was inhaled. Processing of the plant material into oils or resins before combustion adds another level of complexity to the potential chemical diversity of the smoke that is inhaled.

Zebrafish larvae are an established model for testing vertebrate toxicity, teratogenicity and environmental risk assessment [120, 121, 122]. The use of these models has proven to be a valuable resource for testing the toxicity of various extracts and condensates obtained from tobacco cigarette smoke [6, 123, 124]. It was found that smoke from tobacco cigarettes was more toxic and produced different phenotypes than that of nicotine alone, suggesting, that the other toxic components found in tobacco smoke are having an effect. The use of the previously validated smoke testing models for testing cannabis smoke has the potential to provide information on developmental, cardiac, behavioral/neural and acute toxicity.

3.7. Multi-drug interactions and polypharmacology

One of the major complexities of working with cannabis is the fact that it is currently known to be comprised of 500+ constituents and more than 100 cannabinoid molecules [125]. The potential of interactions between many of these compounds is high and has been widely demonstrated for THC and CBD. It has been shown that the zebrafish can be used to assess the interaction of THC and CBD with respect to their effects on locomotor activity along with the uptake and metabolism of each compound [106]. Many of the aforementioned zebrafish models have the potential to be used to assess the potential interaction of THC and CBD and likely other cannabinoids. This is important as one of the next steps in the use of the zebrafish models for testing cannabinoids is to begin to test various extracts and isolates from cannabis for their bioactivity. Having an understanding of how the pure compounds interact in an in vivo system and how this relates to their activity in complex mixtures derived from plant material will be extremely valuable. In addition to the interaction of the various compounds found within cannabis, the use of cannabis or cannabinoids as therapeutics is also complicated by the fact that many patients are already taking a prescription drug for their particular indication. The zebrafish testing platforms appear to have the potential to characterize some of these interactions as well.

Understanding the pharmacokinetics and pharmacodynamics of cannabis is complicated by the fact that CBD (and potentially other cannabinoids) has numerous targets and mechanisms of action that contribute to its various biological effects. Similar to CBD a high percentage of neuroactive compounds have multiple targets and act on them within similar concentration ranges. This polypharmacology has both advantages and disadvantages. As many disease etiologies are not entirely known and may be multifactorial, there may be a substantial benefit of having activity on multiple targets. However, this may also increase the side effect potential and the potential of interacting with other therapeutics. It has been suggested that large-scale zebrafish behavioral testing models can be used to help discern the polypharmacological mechanisms of neuroactive compounds [126]. This provides an ideal platform with which to test cannabis derivatives.

Advertisement

4. Discussion

One of the unique characteristics of researching the effects of cannabis and cannabinoids using various models and for various disease indications is that often there is already clinical data on the effects in humans. While much of the data is often anecdotal in nature, it does allow for animal model testing to be used to back validate the findings of the clinical trials. By designing top-down translational research studies we can begin to elucidate the biological basis of the clinical findings and potentially provide information on the mechanism of action of therapeutic compounds. This is particularly true for cannabis uses where the cannabinoid mechanism of action is often difficult to discern. The use of animal models of disease may help to elucidate these mechanisms and further define the etiology of the disease.

As outlined in this chapter, zebrafish have an established endocannabinoid system that is highly analogous to that of humans. Additionally, as a model system both adults and larval zebrafish provide numerous models of disease that have been shown to be efficacious for testing the therapeutic potential of novel compounds. Importantly the response patterns in these various disease models following exposure to different cannabinoids reveal unique characteristics for each cannabinoid. Thus far, only a limited number of these models have been used to test the efficacy of cannabinoids. However, the framework is in place for an expansion of the use of zebrafish in this field.

References

  1. 1. Steenbergen PJ, Richardson MK, Champagne DL. The use of the zebrafish model in stress research. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2011;35(6):1432-1451
  2. 2. Howe K et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature. 2013;496(7446):498-503
  3. 3. Grunwald DJ, Eisen JS. Headwaters of the zebrafish—Emergence of a new model vertebrate. Nature Reviews. Genetics. 2002;3(9):717-724
  4. 4. Hoshijima K, Jurynec MJ, Grunwald DJ. Precise editing of the Zebrafish genome made simple and efficient. Developmental Cell. 2016;36(6):654-667
  5. 5. Sertori R et al. Genome editing in zebrafish: A practical overview. Briefings in Functional Genomics. 2016;15(4):322-330
  6. 6. Ellis LD et al. Use of the zebrafish larvae as a model to study cigarette smoke condensate toxicity. PLoS One. 2014;9(12):e115305
  7. 7. Park MJ et al. Predicted drug-induced bradycardia related cardio toxicity using a zebrafish in vivo model is highly correlated with results from in vitro tests. Toxicology Letters. 2013;216(1):9-15
  8. 8. Langheinrich U, Vacun G, Wagner T. Zebrafish embryos express an orthologue of HERG and are sensitive toward a range of QT-prolonging drugs inducing severe arrhythmia. Toxicology and Applied Pharmacology. 2003;193(3):370-382
  9. 9. Kalueff AV, Echevarria DJ, Stewart AM. Gaining translational momentum: More zebrafish models for neuroscience research. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2014;55:1-6
  10. 10. Kalueff AV, Nguyen M. Testing anxiolytic drugs in the C57BL/6J mouse strain. Journal of Pharmacological and Toxicological Methods. 2014;69(2):205-207
  11. 11. Stewart AM et al. Zebrafish models for translational neuroscience research: From tank to bedside. Trends in Neurosciences. 2014;37(5):264-278
  12. 12. Gerlai R. Reproducibility and replicability in zebrafish behavioral neuroscience research. Pharmacology, Biochemistry, and Behavior. 23 Feb 2018; pii: S0091-3057(17)30608-1. DOI: 10.1016/j.pbb.2018.02.005
  13. 13. Fontana BD et al. The developing utility of zebrafish models of neurological and neuropsychiatric disorders: A critical review. Experimental Neurology. 2018;299(Pt A):157-171
  14. 14. Shams S et al. The zebrafish as a promising tool for modeling human brain disorders: A review based upon an IBNS symposium. Neuroscience and Biobehavioral Reviews. 2018;85:176-190
  15. 15. Khan KM et al. Zebrafish models in neuropsychopharmacology and CNS drug discovery. British Journal of Pharmacology. 2017;174(13):1925-1944
  16. 16. Kalueff AV, Stewart AM, Gerlai R. Zebrafish as an emerging model for studying complex brain disorders. Trends in Pharmacological Sciences. 2014;35(2):63-75
  17. 17. Piato AL et al. Acute restraint stress in zebrafish: Behavioral parameters and purinergic signaling. Neurochemical Research. 2011;36(10):1876-1886
  18. 18. Panula P et al. The comparative neuroanatomy and neurochemistry of zebrafish CNS systems of relevance to human neuropsychiatric diseases. Neurobiology of Disease. 2010;40(1):46-57
  19. 19. Anichtchik O et al. Distinct structure and activity of monoamine oxidase in the brain of zebrafish (Danio rerio). The Journal of Comparative Neurology. 2006;498(5):593-610
  20. 20. Baraban SC et al. Pentylenetetrazole induced changes in zebrafish behavior, neural activity and c-fos expression. Neuroscience. 2005;131(3):759-768
  21. 21. Fonseka TM et al. Zebrafish models of major depressive disorders. Journal of Neuroscience Research. 2016;94(1):3-14
  22. 22. Giacomotto J et al. Developmental suppression of schizophrenia-associated miR-137 alters sensorimotor function in zebrafish. Translational Psychiatry. 2016;6:e818
  23. 23. Grone BP et al. Epilepsy, behavioral abnormalities, and physiological comorbidities in Syntaxin-binding protein 1 (STXBP1) mutant Zebrafish. PLoS One. 2016;11(3):e0151148
  24. 24. Lee J, Freeman JL. Embryonic exposure to 10 mug L(−1) lead results in female-specific expression changes in genes associated with nervous system development and function and Alzheimer’s disease in aged adult zebrafish brain. Metallomics. 2016;8(6):589-596
  25. 25. Wood JD et al. Disrupted-in-schizophrenia 1 and neuregulin 1 are required for the specification of oligodendrocytes and neurones in the zebrafish brain. Human Molecular Genetics. 2009;18(3):391-404
  26. 26. Ahrens MB et al. Whole-brain functional imaging at cellular resolution using light-sheet microscopy. Nature Methods. 2013;10(5):413-420
  27. 27. Vanwalleghem GC, Ahrens MB, Scott EK. Integrative whole-brain neuroscience in larval zebrafish. Current Opinion in Neurobiology. 2018;50:136-145
  28. 28. Thomas RJ. The toxicologic and teratologic effects of delta-9-tetrahydrocannabinol in the zebrafish embryo. Toxicology and Applied Pharmacology. 1975;32(1):184-190
  29. 29. Lam CS, Rastegar S, Strahle U. Distribution of cannabinoid receptor 1 in the CNS of zebrafish. Neuroscience. 2006;138(1):83-95
  30. 30. Migliarini B, Carnevali O. A novel role for the endocannabinoid system during zebrafish development. Molecular and Cellular Endocrinology. 2009;299(2):172-177
  31. 31. Oltrabella F et al. Role of the endocannabinoid system in vertebrates: Emphasis on the zebrafish model. Development, Growth & Differentiation. 2017;59(4):194-210
  32. 32. Rodriguez-Martin I et al. Characterization of two duplicate zebrafish Cb2-like cannabinoid receptors. Gene. 2007;389(1):36-44
  33. 33. Martella A et al. Important role of endocannabinoid signaling in the development of functional vision and locomotion in zebrafish. The FASEB Journal. 2016;30(12):4275-4288
  34. 34. Fernandez-Ruiz J et al. The endogenous cannabinoid system and brain development. Trends in Neurosciences. 2000;23(1):14-20
  35. 35. Watson S et al. The endocannabinoid receptor, CB1, is required for normal axonal growth and fasciculation. Molecular and Cellular Neurosciences. 2008;38(1):89-97
  36. 36. Liu LY et al. Cannabinoid receptor signaling regulates liver development and metabolism. Development. 2016;143(4):609-622
  37. 37. Esain V et al. Cannabinoid receptor-2 regulates embryonic hematopoietic stem cell development via prostaglandin E2 and P-selectin activity. Stem Cells. 2015;33(8):2596-2612
  38. 38. Kirkham TC et al. Endocannabinoid levels in rat limbic forebrain and hypothalamus in relation to fasting, feeding and satiation: Stimulation of eating by 2-arachidonoyl glycerol. British Journal of Pharmacology. 2002;136(4):550-557
  39. 39. Williams CM, Kirkham TC. Anandamide induces overeating: Mediation by central cannabinoid (CB1) receptors. Psychopharmacology. 1999;143(3):315-317
  40. 40. Williams CM, Kirkham TC. Reversal of delta 9-THC hyperphagia by SR141716 and naloxone but not dexfenfluramine. Pharmacology, Biochemistry, and Behavior. 2002;71(1-2):333-340
  41. 41. Wiley JL et al. CB1 cannabinoid receptor-mediated modulation of food intake in mice. British Journal of Pharmacology. 2005;145(3):293-300
  42. 42. Arias Horcajadas F. Cannabinoids in eating disorders and obesity. Molecular Neurobiology. 2007;36(1):113-128
  43. 43. Ravinet Trillou C et al. Anti-obesity effect of SR141716, a CB1 receptor antagonist, in diet-induced obese mice. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2003;284(2):R345-R353
  44. 44. Di Marzo V. CB(1) receptor antagonism: Biological basis for metabolic effects. Drug Discovery Today. 2008;13(23-24):1026-1041
  45. 45. Russo P et al. Genetic variations at the endocannabinoid type 1 receptor gene (CNR1) are associated with obesity phenotypes in men. The Journal of Clinical Endocrinology and Metabolism. 2007;92(6):2382-2386
  46. 46. Scheen AJ. CB1 receptor blockade and its impact on cardiometabolic risk factors: Overview of the RIO programme with rimonabant. Journal of Neuroendocrinology. 2008;20(Suppl 1):139-146
  47. 47. Ravinet Trillou C et al. CB1 cannabinoid receptor knockout in mice leads to leanness, resistance to diet-induced obesity and enhanced leptin sensitivity. International Journal of Obesity and Related Metabolic Disorders. 2004;28(4):640-648
  48. 48. Shimada Y et al. A high-throughput fluorescence-based assay system for appetite-regulating gene and drug screening. PLoS One. 2012;7(12):e52549
  49. 49. Piccinetti CC et al. Appetite regulation: The central role of melatonin in Danio rerio. Hormones and Behavior. 2010;58(5):780-785
  50. 50. Flynn EJ 3rd, Trent CM, Rawls JF. Ontogeny and nutritional control of adipogenesis in zebrafish (Danio rerio). Journal of Lipid Research. 2009;50(8):1641-1652
  51. 51. Miyares RL, de Rezende VB, Farber SA. Zebrafish yolk lipid processing: A tractable tool for the study of vertebrate lipid transport and metabolism. Disease Models & Mechanisms. 2014;7(7):915-927
  52. 52. Schlegel A, Stainier DY. Microsomal triglyceride transfer protein is required for yolk lipid utilization and absorption of dietary lipids in zebrafish larvae. Biochemistry. 2006;45(51):15179-15187
  53. 53. Pai WY et al. Cannabinoid receptor 1 promotes hepatic lipid accumulation and lipotoxicity through the induction of SREBP-1c expression in zebrafish. Transgenic Research. 2013;22(4):823-838
  54. 54. Fraher D et al. Lipid abundance in Zebrafish embryos is regulated by complementary actions of the endocannabinoid system and retinoic acid pathway. Endocrinology. 2015;156(10):3596-3609
  55. 55. Silvestri C et al. Two non-psychoactive cannabinoids reduce intracellular lipid levels and inhibit hepatosteatosis. Journal of Hepatology. 2015;62(6):1382-1390
  56. 56. Zanettini C et al. Effects of endocannabinoid system modulation on cognitive and emotional behavior. Frontiers in Behavioral Neuroscience. 2011;5:57
  57. 57. Maximino C et al. Measuring anxiety in zebrafish: A critical review. Behavioural Brain Research. 2010;214(2):157-171
  58. 58. Bailey JM, Oliveri AN, Levin ED. Pharmacological analyses of learning and memory in zebrafish (Danio rerio). Pharmacology, Biochemistry, and Behavior. 2015;139(Pt B):103-111
  59. 59. K vF. Uber einen Schreckstoff der Fischhaut und seine biologisehe Bedeutung seine biologisehe Bedeutung. Zeitschrift für Vergleichende Physiologie. 1941;29:46-145
  60. 60. Ruhl T et al. Acute administration of THC impairs spatial but not associative memory function in zebrafish. Psychopharmacology. 2014;231(19):3829-3842
  61. 61. Nazario LR et al. Caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio). Pharmacology, Biochemistry, and Behavior. 2015;135:210-216
  62. 62. Lynch ME. Cannabinoids in the management of chronic pain: A front line clinical perspective. Journal of Basic and Clinical Physiology and Pharmacology. 2016;27(3):189-191
  63. 63. Lynch ME, Ware MA. Cannabinoids for the treatment of chronic non-Cancer pain: An updated systematic review of randomized controlled trials. Journal of Neuroimmune Pharmacology. 2015;10(2):293-301
  64. 64. Savage SR et al. Cannabis in pain treatment: Clinical and research considerations. The Journal of Pain. 2016;17(6):654-668
  65. 65. Kondrad E. Medical marijuana for chronic pain. North Carolina Medical Journal. 2013;74(3):210-211
  66. 66. Kondrad E, Reid A. Colorado family physicians’ attitudes toward medical marijuana. Journal of American Board of Family Medicine. 2013;26(1):52-60
  67. 67. Romero-Sandoval EA, Kolano AL, Alvarado-Vazquez PA. Cannabis and cannabinoids for chronic pain. Current Rheumatology Reports. 2017;19(11):67
  68. 68. Zogopoulos P et al. The role of endocannabinoids in pain modulation. Fundamental & Clinical Pharmacology. 2013;27(1):64-80
  69. 69. Finnerup NB et al. Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis. Lancet Neurology. 2015;14(2):162-173
  70. 70. Volkow ND, McLellan AT. Opioid abuse in chronic pain—Misconceptions and mitigation strategies. The New England Journal of Medicine. 2016;374(13):1253-1263
  71. 71. Malafoglia V et al. The zebrafish as a model for nociception studies. Journal of Cellular Physiology. 2013;228(10):1956-1966
  72. 72. Correia AD et al. A novel behavioral fish model of nociception for testing analgesics. Pharmaceuticals (Basel, Switzerland). 2011;4(4):665-680
  73. 73. Curtright A et al. Modeling nociception in zebrafish: A way forward for unbiased analgesic discovery. PLoS One. 2015;10(1):e0116766
  74. 74. Gau P et al. The zebrafish ortholog of TRPV1 is required for heat-induced locomotion. The Journal of Neuroscience. 2013;33(12):5249-5260
  75. 75. Malafoglia V et al. Extreme thermal noxious stimuli induce pain responses in zebrafish larvae. Journal of Cellular Physiology. 2014;229(3):300-308
  76. 76. Maximino C. Modulation of nocicpetive-like behavior in zebrafish by environmental stressors. Psychology & Neuroscience. 2011;4(1):149-155
  77. 77. Prober DA et al. Zebrafish TRPA1 channels are required for chemosensation but not for thermosensation or mechanosensory hair cell function. The Journal of Neuroscience. 2008;28(40):10102-10110
  78. 78. Reilly SC et al. Behavioural analysis of a nociceptive event in fish: Comparisons between three species demonstrate specific responses. Applied Animal Behaviour Science. 2008;114(1-2):248-259
  79. 79. Steenbergen PJ, Bardine N. Antinociceptive effects of buprenorphine in zebrafish larvae: An alternative for rodent models to study pain and nociception? Applied Animal Behaviour Science. 2014;152:92-99
  80. 80. Lima MG et al. Nocifensive behavior in adult and larval zebrafish. In: Kalueff AV, Stewart AM, editors. Zebrafish Protocols for Neurobehavioral Research. Totowa, NJ: Humana Press; 2012. pp. 153-166
  81. 81. Taylor JC et al. A novel zebrafish-based model of nociception. Physiology & Behavior. 2017;174:83-88
  82. 82. Lopez-Luna J et al. Reduction in activity by noxious chemical stimulation is ameliorated by immersion in analgesic drugs in zebrafish. The Journal of Experimental Biology. 2017;220(Pt 8):1451-1458
  83. 83. Ellis LD et al. Comparison of cannabinoids with known analgesics using a novel high throughput zebrafish larval model of nociception. Behavioural Brain Research. 30 Jan 2018;337:151-159
  84. 84. Costa B et al. The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. European Journal of Pharmacology. 2007;556(1-3):75-83
  85. 85. Hammell DC et al. Transdermal cannabidiol reduces inflammation and pain-related behaviours in a rat model of arthritis. European Journal of Pain. 2016;20(6):936-948
  86. 86. Boychuk DG et al. The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: A systematic review. Journal of Oral & Facial Pain and Headache. 2015;29(1):7-14
  87. 87. Voon P, Karamouzian M, Kerr T. Chronic pain and opioid misuse: A review of reviews. Substance Abuse Treatment, Prevention, and Policy. 2017;12(1):36
  88. 88. Cressey D. The cannabis experiment. Nature. 2015;524(7565):280-283
  89. 89. Lopez-Quintero C et al. Probability and predictors of transition from first use to dependence on nicotine, alcohol, cannabis, and cocaine: Results of the National Epidemiologic Survey on alcohol and related conditions (NESARC). Drug and Alcohol Dependence. 2011;115(1-2):120-130
  90. 90. Mathur P, Guo S. Use of zebrafish as a model to understand mechanisms of addiction and complex neurobehavioral phenotypes. Neurobiology of Disease. 2010;40(1):66-72
  91. 91. Gerlai R et al. Acute and chronic alcohol dose: Population differences in behavior and neurochemistry of zebrafish. Genes, Brain, and Behavior. 2009;8(6):586-599
  92. 92. Petzold AM et al. Nicotine response genetics in the zebrafish. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(44):18662-18667
  93. 93. Cachat J et al. Modeling withdrawal syndrome in zebrafish. Behavioural Brain Research. 2010;208(2):371-376
  94. 94. Braida D et al. Hallucinatory and rewarding effect of salvinorin a in zebrafish: Kappa-opioid and CB1-cannabinoid receptor involvement. Psychopharmacology. 2007;190(4):441-448
  95. 95. Bretaud S et al. A choice behavior for morphine reveals experience-dependent drug preference and underlying neural substrates in developing larval zebrafish. Neuroscience. 2007;146(3):1109-1116
  96. 96. Stewart AM et al. Molecular psychiatry of zebrafish. Molecular Psychiatry. 2015;20(1):2-17
  97. 97. Kily LJ et al. Gene expression changes in a zebrafish model of drug dependency suggest conservation of neuro-adaptation pathways. The Journal of Experimental Biology. 2008;211(Pt 10):1623-1634
  98. 98. Lopez Patino MA et al. Gender differences in zebrafish responses to cocaine withdrawal. Physiology & Behavior. 2008;95(1-2):36-47
  99. 99. Mathur P, Lau B, Guo S. Conditioned place preference behavior in zebrafish. Nature Protocols. 2011;6(3):338-345
  100. 100. Klee EW et al. Zebrafish: A model for the study of addiction genetics. Human Genetics. Jun 2012;131(6):977-1008
  101. 101. Akhtar MT et al. Developmental effects of cannabinoids on zebrafish larvae. Zebrafish. 2013;10(3):283-293
  102. 102. Viveros MP, Marco EM, File SE. Endocannabinoid system and stress and anxiety responses. Pharmacology, Biochemistry, and Behavior. 2005;81(2):331-342
  103. 103. Ellis LD, Seibert J, Soanes KH. Distinct models of induced hyperactivity in zebrafish larvae. Brain Research. 17 Apr 2012;1449:46-59
  104. 104. Stewart A et al. Modeling anxiety using adult zebrafish: A conceptual review. Neuropharmacology. 2012;62(1):135-143
  105. 105. Stewart A et al. Pharmacological modulation of anxiety-like phenotypes in adult zebrafish behavioral models. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 1 Aug 2011;35(6):1421-1431
  106. 106. Achenbach JC et al. Analysis of the uptake, metabolism, and behavioral effects of cannabinoids on Zebrafish larvae. Zebrafish. 10 Apr 2018. DOI: 10.1089/zeb.2017.1541
  107. 107. Maximino C et al. Scototaxis as anxiety-like behavior in fish. Nature Protocols. 2010;5(2):209-216
  108. 108. O’Neil ME et al. Benefits and harms of plant-based Cannabis for posttraumatic stress disorder: A systematic review. Annals of Internal Medicine. 2017;167(5):332-340
  109. 109. Caramillo EM et al. Modeling PTSD in the zebrafish: Are we there yet? Behavioural Brain Research. 2015;276:151-160
  110. 110. Stewart AM et al. Developing zebrafish models relevant to PTSD and other trauma- and stressor-related disorders. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2014;55:67-79
  111. 111. Grotenhermen F. Pharmacokinetics and pharmacodynamics of cannabinoids. Clinical Pharmacokinetics. 2003;42(4):327-360
  112. 112. Devinsky O et al. Trial of Cannabidiol for drug-resistant seizures in the Dravet syndrome. The New England Journal of Medicine. 2017;376(21):2011-2020
  113. 113. Abrams DI. The therapeutic effects of Cannabis and cannabinoids: An update from the National Academies of sciences, engineering and medicine report. European Journal of Internal Medicine. 2018;49:7-11
  114. 114. Perucca E. Cannabinoids in the treatment of epilepsy: Hard evidence at last? Journal of Epilepsy Research. 2017;7(2):61-76
  115. 115. Cunliffe VT. Building a zebrafish toolkit for investigating the pathobiology of epilepsy and identifying new treatments for epileptic seizures. Journal of Neuroscience Methods. 2016;260:91-95
  116. 116. Burgess HA, Granato M. Modulation of locomotor activity in larval zebrafish during light adaptation. The Journal of Experimental Biology. 2007;210(Pt 14):2526-2539
  117. 117. Kim YH et al. Reduced neuronal proliferation by proconvulsant drugs in the developing zebrafish brain. Neurotoxicology and Teratology. 2010;32(5):551-557
  118. 118. Joshi M, Joshi A, Bartter T. Marijuana and lung diseases. Current Opinion in Pulmonary Medicine. 2014;20(2):173-179
  119. 119. Ware MA et al. Cannabis for the management of pain: Assessment of safety study (COMPASS). The Journal of Pain. 2015;16(12):1233-1242
  120. 120. Padilla S et al. Zebrafish developmental screening of the ToxCast phase I chemical library. Reproductive Toxicology. 2011;33(2):174-187
  121. 121. Sipes NS, Padilla S, Knudsen TB. Zebrafish: As an integrative model for twenty-first century toxicity testing. Birth Defects Research. Part C, Embryo Today. 2011;93(3):256-267
  122. 122. Peterson RT, Macrae CA. Systematic approaches to toxicology in the zebrafish. Annual Review of Pharmacology and Toxicology. 2012;52:433-453
  123. 123. Massarsky A et al. Teratogenic, bioenergetic, and behavioral effects of exposure to total particulate matter on early development of zebrafish (Danio rerio) are not mimicked by nicotine. Neurotoxicology and Teratology. 2015;51:77-88
  124. 124. Folkesson M et al. Differences in cardiovascular toxicities associated with cigarette smoking and snuff use revealed using novel zebrafish models. Biology Open. 2016;5(7):970-978
  125. 125. Mehmedic Z et al. Potency trends of Delta9-THC and other cannabinoids in confiscated cannabis preparations from 1993 to 2008. Journal of Forensic Sciences. 2010;55(5):1209-1217
  126. 126. McCarroll MN et al. Leveraging large-scale behavioral profiling in Zebrafish to explore neuroactive polypharmacology. ACS Chemical Biology. 2016;11(4):842-849

Written By

Lee Ellis

Submitted: 22 May 2018 Reviewed: 05 June 2018 Published: 05 November 2018