Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

Contents lists available at ScienceDirect

Comparative Biochemistry and Physiology, Part C


journal homepage: www.elsevier.com/locate/cbpc

Review

Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of T


xenobiotics in humans?

Carina de Souza Anselmoa,b, , Vinicius Figueiredo Sardelaa,c, Valeria Pereira de Sousab,
Henrique Marcelo Gualberto Pereiraa
a
Federal University of Rio de Janeiro, Institute of Chemistry, LBCD-LADETEC, Av Horácio Macedo, 1281, 21941-598, Polo de Química, bloco C, Cidade Universitária,
Rio de Janeiro, RJ, Brazil
b
Federal University of Rio de Janeiro, Department of Drugs and Pharmaceutics, Faculty of Pharmacy, LabCQ, Av Carlos Chagas Filho, 373, 21941-902, Bss36, Cidade
Universitária, Rio de Janeiro, RJ, Brazil
c
Federal University of Rio de Janeiro, Institute of Chemistry, LPDI-LADETEC, Av Horácio Macedo, 1281, 21941-598, Polo de Química, bloco C, Cidade Universitária, Rio
de Janeiro, RJ, Brazil

A R T I C LE I N FO A B S T R A C T

Keywords: Zebrafish has become a popular model organism in several lines of biological research sharing physiological,
Zebrafish (Danio rerio) morphological and histological similarities with mammals. In fact, many human cytochrome P450 (CYP) en-
Xenobiotic metabolism zymes have direct orthologs in zebrafish, suggesting that zebrafish xenobiotic metabolic profiles may be similar
Cytochrome P450 to those in mammals. The focus of the review is to analyse the studies that have evaluated the metabolite
Non-human model
production in zebrafish over the years, either of the drugs themselves or xenobiotics in general (environmental
Drug discovery
pollutants, natural products, etc.), bringing a vision of how these works were performed and comparing, where
Toxicology
possible, with human metabolism. Early studies that observed metabolic production by zebrafish focused on
environmental toxicology, and in recent years the main focus has been on toxicity screening of pharmaceuticals
and drug candidates. Nevertheless, there is still a lack of standardization of the model and the knowledge of the
extent of similarity with human metabolism. Zebrafish screenings are performed at different life stages, typically
being carried out in adult fish through in vivo assays, followed by early larval stages and embryos. Studies
comparing metabolism at the different zebrafish life stages are also common. As with any non-human model, the
zebrafish presents similarities and differences in relation to the profile of generated metabolites compared to that
observed in humans. Although more studies are still needed to assess the degree to which zebrafish metabolism
can be compared to human metabolism, the facts presented indicate that the zebrafish is an excellent potential
model for assessing xenobiotic metabolism.

1. Introduction et al., 2003), toxicology (Thompson et al., 2010; Weigt et al., 2011;
Zhang et al., 2016), drug discovery (Fleming et al., 2005; Kithcart and
Fish are the most numerous and phylogenetically diverse group of MacRae, 2017), disease models (Brugman, 2016; Capiotti et al., 2014)
vertebrates and are useful in the study of fundamental processes in and neurobiology (Pinho et al., 2016; Shams et al., 2017). Therefore,
vertebrate evolution, development, toxicology and disease processes zebrafish represents a viable alternative to the classical mammalian
(Garcia et al., 2016; Langheinrich, 2003; Spitsbergen and Kent, 2003). models currently used in biological research because it has some unique
In particular, zebrafish (Danio rerio), which is a tropical fish of the properties: it is an intact organism with similar development, anatomy
Cyprinidae family, has been a prominent genetic model since 1981 and physiology to higher vertebrates (Otte et al., 2017); it is easier and
(Streisinger et al., 1981), when its genetic amenability allowed the first less expensive than popular rodent models; it is a genetically tractable
genetic screening for mutations that affect organ development in a vertebrate model that can be easily injected with modified genes and
vertebrate (Kamel and Ninov, 2017). Since then, zebrafish has become can absorb chemical mutagens through water; embryos are transparent
a popular model organism in several lines of biological research beyond and develop externally, allowing the use of noninvasive imaging tech-
genetics, including developmental biology (Carten et al., 2011; Ho niques; embryos develop very rapidly compared to mammalian models,


Corresponding author at: LBCD-LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Av. Horácio Macedo, 1281, bloco C - Cidade Universitária, RJ, 21941-598 Rio
de Janeiro, Brazil.
E-mail address: carinaanselmo@iq.ufrj.br (C. de Souza Anselmo).

https://doi.org/10.1016/j.cbpc.2018.06.005
Received 8 May 2018; Received in revised form 25 June 2018; Accepted 27 June 2018
Available online 30 June 2018
1532-0456/ © 2018 Elsevier Inc. All rights reserved.
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

potentially reducing the experimentation time (Garcia et al., 2016); and et al., 1995). The chorion is an acellular envelope containing pores that
it has a high reproduction capacity, where a pair of zebrafish can are approximately 0.5 μm in diameter with 2 μm spacing (Lee et al.,
generate hundreds of fertilized eggs, which develop rapidly into larvae 2009). A limitation to using the zebrafish embryo is its need to take up
with functional metabolic organs (Kamel and Ninov, 2017). substances through the chorion and other membranes, because the gills
Although there are some major differences resulting from adapta- of the adult fish are not yet developed (Vallverdú-Queralt et al., 2015).
tion to aquatic life, most zebrafish organs perform the same functions as Therefore, the chorion can significantly confound the early life zebra-
their human counterparts and exhibit well-conserved physiology fish toxicity assay by leading to false positives because of size-depen-
(MacRae and Peterson, 2015). In fact, zebrafish share physiological, dent limitations in the uptake of large compounds (Langheinrich et al.,
morphological and histological similarities with mammals (Diekmann 2003; Mandrell et al., 2012). The chorion may be removed but not
and Hill, 2013) and its genome has been sequenced, revealing that without potentially affecting embryo integrity and behavior (Mandrell
~70% of human genes have a zebrafish orthologue and approximately et al., 2012; Sipes et al., 2011).
82% of potential human disease-related genes have at least one obvious The morphogenesis of the liver, which is an organ that plays a key
zebrafish orthologue (Howe et al., 2013). Not surprisingly, in terms of role in the biotransformation of xenobiotics, starts at approximately
publication, zebrafish are one of the fastest growing model organisms 24 hpf (hours post-fertilization), and liver-specific markers occur even
(Garcia et al., 2016). Furthermore, chemical-genetic screens in zebra- earlier (16 hpf) (Tao and Peng, 2009). By 5 dpf, the heart, liver, brain,
fish have already rendered the drug ProHema, which is undergoing pancreas, and other organs are developed (Garcia et al., 2016; Kimmel
phase II clinical trials for improving hematopoietic stem cell engraft- et al., 1995). The zebrafish larvae begin independent feeding at ap-
ment and expansion (Kamel and Ninov, 2017; North et al., 2007). As- proximately 6 dpf, growing rapidly and developing into juveniles at
sessing the toxicity of a compound is a critical step in the discovery of approximately 30 dpf, and subsequently into adults at approximately
new drugs. In forensic toxicology, it is also very important to assess the 90 dpf (Kamel and Ninov, 2017). Similar to the mammalian liver, the
toxicity of a compound, since the knowledge of its metabolism will be zebrafish liver is an essential organ in the body and performs several
fundamental for determining the analytical targets to be monitored as vital activities, including metabolism, detoxification and homeostasis
the emergence of ever newer designer drugs is an ongoing challenge for (Menke et al., 2011; Tao and Peng, 2009). However, zebrafish have a
analytical toxicologists in forensic as well as clinical toxicology (Peters unique hepatic anatomy and cellular architecture, despite the high
and Martinez-Ramirez, 2010). To understand or predict the toxic po- conservation of cell types within the liver, compared with mammals
tential of a compound, knowledge of the absorption, distribution, me- (Goessling and Sadler, 2015; Tao and Peng, 2009).
tabolism, and elimination of the compound is required (Garcia et al.,
2016). 3. Enzymes involved in the metabolism of xenobiotics
Notably, extensive metabolism of a xenobiotic, an external and
foreign compound to the organism, could lead to reduced activity, Drug-metabolizing enzymes play central roles in the metabolism,
whereas creation of new active metabolites could lead to increased elimination and/or detoxification of xenobiotics introduced into the
toxicity (Diekmann and Hill, 2013). Zebrafish embryos represent an organism (Xu et al., 2005). Zebrafish have the ability to perform both
attractive model for studies of developmental toxicity of xenobiotics phase I (oxidation, N-demethylation, O-demethylation and N-deal-
both for human and environmental risk assessment (Carlsson et al., kylation) and phase II (sulfation and glucuronidation) metabolism re-
2013). The reason for the preference in the use of embryos instead of actions, and the metabolic enzymes responsible for these reactions are
adult fish is because in Europe, zebrafish embryos are not considered highly conserved in relation to mammals (Brox et al., 2016; Chng et al.,
laboratory animals until the independent feeding stage (European 2012; Diekmann and Hill, 2013). Phase I enzymes consist primarily of
Commission, 2016), which makes them ideal candidates for several the cytochrome P450 (CYP) superfamily of microsomal enzymes, which
chemical screenings. On the other hand, in the United States of America are found abundantly in the liver, gastrointestinal tract, lung and
fish are not protected for research use, while in Brazil, China and India, kidney, catalyzing the oxidative and reductive metabolism of many
all animals are protected for use in research (Sneddon et al., 2017). In chemicals and endogenous compounds (Tseng et al., 2005; Xu et al.,
fact, the zebrafish embryo has already been accepted as a validated 2005). In humans, it is believed that five CYP gene families, namely,
alternative for the acute fish toxicity test (OECD TG236) (OECD, 2013). CYP1, CYP2, CYP3, CYP4 and CYP7, play crucial roles in hepatic as well
In addition, toxicology may be the most prevalent use of zebrafish in as extra-hepatic metabolism and elimination of xenobiotics and drugs,
the industry, with the majority of large pharmaceutical companies re- as CYP1A2, CYP2C9, CYP2D6 and CYP3A4/5 are responsible for the
porting some use of zebrafish for toxicology (MacRae and Peterson, oxidative biotransformation of approximately 70% of most clinically
2015). The metabolism of the substance as a critical step in the tox- used drugs (Saad et al., 2017; Xu et al., 2005). Therefore, CYP families
icological evaluation. Therefore, the purpose of this review is to analyse 1-3 are the main metabolizing enzymes responsible for xenobiotic
publications involving xenobiotic metabolism studies at various stages metabolism (Guengerich, 2008; Saad et al., 2016).
in the zebrafish's evolution over the years. Considerations will also be Many human CYP enzymes have direct orthologs in zebrafish
made on the development of zebrafish, enzymes involved in the me- (Goldstone et al., 2010), suggesting that zebrafish metabolic profiles
tabolism of xenobiotics, and analytical methods for the detection of may be similar to those of mammals. In addition, there are other CYPs
metabolites, highlighting advantages and limitations of zebrafish as a described in fish that lack orthologs in humans, such as CYP1C, CYP2AE
model for the evaluation of xenobiotic metabolism. and CYP2X (Table 1) (Goldstone et al., 2010). Nevertheless, as CYP
activity is not necessarily correlated with its gene expression or even
2. Development protein levels, more focus is needed on CYP activity (Saad et al., 2017).
Like the human CYP3A genes, CYP3A65 transcription in the foregut
The genetic signals and responses that drive early embryonic de- region was enhanced by treatment of the zebrafish larvae with dex-
velopment are fundamentally similar between zebrafish and mamma- amethasone and rifampicin (Tseng et al., 2005). On the other hand, 7-
lian embryogenesis (Sipes et al., 2011). The formation of different benzyloxy-4-(trifluoromethyl)coumarin, a vertebrate CYP3A substrate
organ systems can be followed under the microscope during the de- not usually associated with CYP1 activity, was metabolized more effi-
velopmental period, offering many opportunities for the study of or- ciently by CYP1A than CYP3A65 in zebrafish (Scornaienchi et al.,
ganogenesis (Dawid, 2004). The zebrafish has a rapid development, 2010). That is, a zebrafish CYP that has an ortholog in human will not
progressing from zygote to larvae in 72 h. During the early embryonic necessarily have the same behavior on a certain substrate. Another
stages, zebrafish are covered by a chorionic membrane (egg shell) interesting case is the metabolism of dextromethorphan in zebrafish;
which will be lost between 3 or 4 dpf (days post-fertilization) (Kimmel although no ortholog of human CYP2D6 has been found in the fish

35
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

Table 1 abundant), showing close similarities between mammalian and zebra-


Synteny comparison between zebrafish and the major fish Ugt1 and Ugt2 families (Huang and Wu, 2010; Christen and Fent,
human CYPs involved in the metabolism of xenobiotics. 2014). All the isoforms of UGTs have tissue-, sex- and developmental-
Source: Adapted from Goldstone et al. (2010). specific expression patterns. UGTs expression level dropped at 2 dpf and
Zebrafish Human increased again from 3 to 5 dpf and features high expression of Ugt1a
and Ugt1b in the liver and intestine of female and male zebrafish
CYP1A CYP1A1/1A2
(Christen and Fent, 2014).
CYP1B1 CYP1B1
CYP1C1,2 –
SULTs have also been identified in zebrafish being divided into five
CYP1D1 CYP1D1P families SULT 1, SULT 2, SULT 3 and SULT 6. Similar to the human
CYP2Ks CYP2W1 SULTs, the majority of the zebrafish SULTs that have been cloned be-
CYP2N13 CYP2J2 long to the SULT1 gene family (Liu et al., 2010; Mohammed et al.,
CYP2Ps CYP2J2
2012). The amino acid sequence alignment demonstrates that the pu-
CYP2R1 CYP2R1
CYP2U1 CYP2U1 tative zebrafish Naa10 (catalytic subunit Nα-acetyltransferase from
CYP2V1 CYP2J2 NAT) is highly similar to human Naa10 (Ree et al., 2015). Phylogenetic
CYP2X1-10 – analysis revealed a great diversity of fish GSTs, with 27 members found
CYP2Y3,4 CYP2A/B/F/S
in zebrafish (Glisic et al., 2015). Glisic et al. (2015) suggest involve-
CYP2AA1-12 –
– CYP2D
ment of GST Pi class, Gstt1a, Gstz1, Gstr1, Mgst3a and Mgst3b in the
– CYP2C biotransformation of xenobiotics on their functional similarity to the
CYP2AD2,3,6 CYP2J2 human orthologs in respect to the xenobiotic metabolism. Other work
CYP2AE1,2 – showed further the considerable conservation with regard to the critical
CYP3A65 CYP3A-se1,-se2a
amino acid residues between the zebrafish and mammalian COMTs
CYP3C1-4 CYP3A3,4,7
CYP4F43 CYP4F (Alazizi et al., 2011). In fact, many studies have revealed the presence
CYP4V7,8 CYP4V2 of phase II metabolites produced by zebrafish at different life stages
CYP4T8 – (Alderton et al., 2010; Anselmo et al., 2017; Brox et al., 2016; Shen
CYP5A1 CYP5A1 et al., 2017).
CYP7A1 CYP7A1
CYP7B1 CYP7B1
CYP7C1 – 4. Metabolism of xenobiotics

The zebrafish is increasingly used as a vertebrate model for several


(Table 1) (Goldstone et al., 2010). Saad et al. (2017) showed that studies, including in vivo toxicological screenings, because it combines
zebrafish do metabolize dextromethorphan to dextrorphan, in addition the scale and throughput of in vitro systems with the physiological
to 3-methoxymorphinan, but the ratio between both metabolite con- complexity of a vertebrate whole animal (Garcia et al., 2016). Several
centrations was different than in man. This indicates that not having an studies using zebrafish have observed the formation of metabolites of
ortholog of human CYP in the zebrafish does not necessarily mean that the substances administered in the fish, with several goals in several
there will be no biotransformation of xenobiotics. areas of interest, such as pollutants of the environment, drug discovery
In several mammalian species including man, CYP-related drug and toxicological evaluation. However, there is still a lot to be under-
metabolism tends to be significantly lower during early life stages, and stood about the model and several particularities to be discussed.
the same was observed in vitro in zebrafish (Saad et al., 2017), despite Through an advanced search in the Science Direct database (https://
several studies having already observed the production of metabolites www.sciencedirect.com/science/search), searching for “zebrafish and
in the early larval stages (2 to 7 dpf) (Alderton et al., 2010; Chng et al., metabolism or metabolite” in the abstract, title and keywords fields, the
2012; Hu et al., 2012; Pardal et al., 2014) and embryos (fertilized eggs result presented in Fig. 1 was obtained. As can be observed, the subject
exposed right after being collected) (Brox et al., 2016; Vallverdú- in question presented a considerable growth in the number of pub-
Queralt et al., 2015; Wiegand et al., 2001). Regarding the embryos, the lications overtime, especially after 2005. After evaluating the results of
biotransformation capability was five-fold higher in the older fish em- the search and withdrawing the studies that did not focus on the ob-
bryos (50 hpf) than in the younger embryos (2 to 26 hpf) (Kühnert servation of the metabolism of exogenous substances by the fish, the
et al., 2017). Concerning CYP3A65, the strength of transcription in- data presented in Table 2 were obtained, which included studies that,
creased with stages of development; CYP3A65 mRNA first appeared at with different aims, evaluated the production of xenobiotic metabolites
the 72 hpf stage, and in adult fish, it was intensively transcribed in liver in zebrafish.
and intestine (Chang et al., 2013). The same is true for zebrafish Most of the studies evaluated the metabolism of xenobiotics through
CYP1A, where expression is low during the early stages of development in vivo assays, four used in vitro assays, and two compared in vitro with
and increases dramatically after hatching at approximately 72 hpf (Saad in vivo (Table 2; Fig. 2a). Zebrafish screenings involving metabolite
et al., 2016). Overall, the expression of zebrafish CYPs appears to analysis are performed at different life stages, typically being carried
broadly increase throughout normal embryonic development, with out in adult fish, followed by early larvae stages and embryos (Table 2;
higher expression observed in larvae posthatching (Jones et al., 2010). Fig. 2b). Studies comparing metabolism at the different zebrafish life
During phase II drug metabolism, the drugs or metabolites from stages are also common (Chng et al., 2012; Hertl and Nagel, 1993; Saad
phase I pathways are enzymatically conjugated with a hydrophilic en- et al., 2017). In cases where adult fish were used, most studies used
dogenous compound by the most common transferase enzymes: UDP- both sexes, and some did not cite this type of information (Table 2). The
glucuronosyltransferases (UGTs), sulfotransferases (SULTs), N-acetyl- in vitro studies were performed using liver homogenate, liver micro-
transferases (NATs), glutathione S-transferases (GSTs), thiopurine S- somes or whole zebrafish, whereas in vivo assays generally directly
methyltransferases (TPMTs), and catechol O-methyltransferases exposed the fish to water or medium containing the substances, with
(COMTs) (Almazroo et al., 2017; Xu et al., 2005). Glucuronidation is two exceptions in the routes of administration: spiked food (Wen et al.,
the major phase II drug metabolism pathway, with approximately 40% 2015) and intraperitoneal injection (Troxel et al., 1997). The number of
to 70% of human endogenous and exogenous compounds conjugated to animals used in the studies was widely variable, as generally when
glucuronidated forms to be excreted from the body (Almazroo et al., using embryos or larvae the number tends to be higher (25 (Brox et al.,
2017). In zebrafish, a total of 45 UGT genes were identified that can be 2016), 60 (Vallverdú-Queralt et al., 2015) and 200 (Hertl and Nagel,
divided into three families consisting of Ugt1s, Ugt2s and Ugt5s (most 1993) embryos; 20 (Chng et al., 2012), 30 (Jones et al., 2012), 40

36
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

Fig. 1. Number of publications over the years mentioning the terms “zebrafish” and “metabolism” or “metabolite” in the title, abstract and/or keywords.
Source: https://www.sciencedirect.com/science/search.

(Alderton et al., 2010) and 100 (Hertl and Nagel, 1993) larvae) than cresol (Kasokat et al., 1987). Following this line of study, the metabo-
when using adult animals (6 (Shen et al., 2017), 12 (Anselmo et al., lism of chlorobenzene (CB) and hexachlorobenzene (HCB) was also
2017), 16 (Wen et al., 2015) and 20 animals (Kasokat et al., 1987, evaluated in adult zebrafish in a very similar way to the previous work
1989)), but the number of animals used is not always provided. (Kasokat et al., 1989). The metabolites found for CB were isomeric
Another factor that has been shown to be quite variable is the ex- chlorophenols (phase I products) and the corresponding glucuronide
posure time of the fish to the substances tested: in the in vitro assays the and sulfate conjugates (phase II products), and no metabolites were
exposure time tends to be shorter and less variable (from 45 min to detected for HCB. The results for CB were in agreement with those
24 h) than in the in vivo tests (from 3 h to 20 days) (Fig. 2c). The mean observed in other freshwater fish species. However, other fish species
fish maintenance temperature, in the case of the in vivo experiments and have already been shown to metabolize HCB, which may indicate that
the in vitro assays, ranged from 23 to 28.5 °C, except for an in vitro study failure to observe metabolites in this work may be due to the exposure
that used a temperature of 35 °C (Wiegand et al., 2001) (Table 2). time, mode of exposure or use of the organic solvent toluene.
Zebrafish are classified as eurythermal, as they exhibit a tolerance for Continuing to use the zebrafish for xenobiotic analysis with impact
wide temperature ranges, and usually a temperature of 28.5 °C is con- on the environment, Gorge and Nagel (1990) evaluated the hazards of
sidered optimal and recommended (Kimmel et al., 1995; Lawrence, the pesticides lindane and atrazine to know whether if and to what
2007), although OECD guidelines also suggest ranges between 21 and extent substances are incorporated, bioconcentrated and eliminated
25 °C (OECD, 1992) and 26 ± 1 °C (OECD, 2013). Overall, adult zeb- when exposure is stopped. None of the metabolites were identified, but
rafish were cultured in a 12 h:12 h (Shen et al., 2017; G. Wang et al., it was possible to observe that atrazine was metabolized to a small
2017; Zok et al., 1991) or 14 h:10 h (Saad et al., 2017; Troxel et al., extent (4%) and that only three metabolites were formed. In contrast,
1997) day/night cycle as recommended (12–16 h photoperiod) (OECD, 50% of lindane was converted to twelve more polar metabolites than
2013). Concerning the use of organic solvents to increase the solubility lindane, which must be dechlorination products of the compound. Zok
of tested compounds, several studies used DMSO at different con- et al. (1991) conducted an experiment to evaluate the toxicokinetics of
centrations (0.01 to 1%, v/v); two used methanol (0.1 or 1%, v/v), nine anilines in adult zebrafish following in vivo exposure in water. The
ethanol (0.01 or 0.0015%, v/v) or toluene (0.007%, v/v); and less than corresponding acetanilides were the only metabolites identified in
half of the studies did not use any type of solvent (Fig. 2d). It is im- water, and no biotransformation occurred at all for 2- and 4-nitroani-
portant to note that even low concentrations of organic solvents may line, suggesting that substitution in the ortho position of the aniline
influence the activity of zebrafish phase I and II enzymes (David et al., sterically hinders acetylation. The acetylation reaction is catalyzed by
2012), and some studies have used high concentrations of DMSO and an acetyltransferase that depends on acetyl-CoA, which exists in zeb-
methanol (1%, v/v) (Table 2). rafish (Ree et al., 2015). No hydroxylation of the aromatic ring was
The first publications that verified the generation of metabolites by observed for the anilines in zebrafish, which was not in agreement with
zebrafish were focused on environmental toxicology, since the knowl- results observed in humans, where N-acetyl-4-aminophenol was the
edge of biotransformation pathways of xenobiotics plays a fundamental predominant urinary aniline metabolite and acetanilides were found
role in environmental monitoring programs. To the best of our knowl- only in minor amounts (Modick et al., 2016).
edge, the pioneering work that evaluated xenobiotics metabolites in Hertl and Nagel (1993) evaluated the in vivo metabolism of 3,4-
zebrafish was conducted in 1987 (Kasokat et al., 1987) (Table 2), in dichloroaniline, which is an intermediate product of the degradation of
which the metabolism of phenol and substituted phenols was studied in several herbicides, at different life stages of zebrafish. All life stages of
adult zebrafish following in vivo exposure in the water. On this occa- zebrafish possess the capability to metabolize 3,4-dichloroaniline to
sion, phenyl glucuronide, phenyl sulfate, and quinol sulfate were 3,4-dichloroacetanilide, and acetanilide formation appeared to be
identified as metabolites of phenol; 2-cresyl glucuronide, 2-cresyl sul- higher in the more advanced stages of the fish. In this work, although
fate, and 2-hydroxybenzoic acid were determined to be metabolites of without confirmation, there is evidence of the presence of the hydro-
2-cresol; and only the glucuronide and sulfate conjugates were detected xylated metabolite for 3,4-dichloroaniline. Other studies with en-
as metabolites of 4-nitrophenol, 4-chlorophenol and penta- vironmental pollutants were performed, such as the evaluation of the
chlorophenol. Overall, no significant differences from related fresh- metabolism of atrazine and bisphenol A in zebrafish. Atrazine is a
water fish species were found, with the exception of the considerably widely used herbicide, as the induction of GST provides an effective
lower oxidation rates which were observed in the case of phenol and 2- resistance against atrazine in plants, and it was evaluated whether if the

37
Table 2
Publications involving the study of the metabolism of xenobiotics in zebrafish.
Author Zebrafish's life stage Brief Experimental design Analytical technique Results Observations

Kasokat et al., 1987 Adult (female; age not The metabolism of phenol and In vivo. Exposure in water with ethanol HPLC-UV Several phenol and substituted Overall, the metabolites recovered
informed) substituted phenols in zebrafish was 0.01% (v/v) for some compounds. phenols metabolites have been were higher for substituted phenols
studied Water analyzed. ETa: 48 h/35 h Tb: identified, with phase II being than or phenol itself indicating that
C. de Souza Anselmo et al.

26 °C more common than phase I some substituted phenols are more


metabolites. rapidly metabolized
Kasokat et al., 1989 Adult (female; age not The metabolism of chlorobenzene (CB) In vivo. Exposure in water with ethanol HPLC-UV Hydroxylate and conjugate Zebrafish is able to hydroxylate
informed) and hexachlorobenzene (HCB) in 0.01/toluene 0.007% (v/v). Water and metabolites were detected for aromatic compounds. The finding
zebrafish was studied fish analyzed. ETa: 48 h Tb: 26 °C CB. Turning to HCB, no was in contrast to published work
metabolites could be detected concerning the metabolism of HCB
under the experimental in fish.
conditions
Gorge and Nagel, Larvae (28 dpf) Study of the metabolism of two In vivo. Exposure in water. Water HPLC-UV Presence of 12 metabolites of Lindane more metabolized than
1990 radiolabelled pesticides, lindane and analyzed. ETa: 40 h/Tb: (26 ± 1)°C lindane and 4 metabolites of atrazine.
atrazine atrazine
Zok et al., 1991 Adult (both sexes; Biotransformation products of anilines In vivo. Exposure in water. Water and HPLC-UV Corresponding acetanilides Substitution in the ortho position of
12 weeks) in the zebrafish were analyzed by fish analyzed. ETa: 24 h Tb: were the only metabolites the aniline sterically hinders
HPLC-UV (26.5 ± 1)°C identified in water. No acetylation. No ring hydroxylation of
biotransformation for 2- and 4- the anilines was observed.
nitroaniline.
Hertl and Nagel, 1993 Embryos (4 hpf), larvae Metabolism of 3,4-dichloroaniline In vivo. Exposure in water. Fish HPLC-UV All life stages of zebrafish There is evidence of hydroxylation
(4 dpf and 17 dpf) and (3,4-DCA) analyzed. ETa: 48 h Tb: 26 °C possess the capability to product of the aniline.
adult (female; 24 weeks) generate the metabolite 3,4-
dichloroacetanilide
Troxel et al., 1997 Adult (sexually mature, Characterize the in vivo metabolism In vivo/in vitro. Intraperitoneal HPLC-UV The major metabolites of AFB1 The results demonstrate that
both sexes) and hepatic DNA adduction of the injection.. Water analyzed. ET: 24 h/T: recovered in the water were zebrafish have the capacity for both

38
carcinogenic aflatoxin B1 (AFB1) in the (26 ± 1)°C. Zebrafish liver identified as aflatoxicol and phase I and phase II metabolism of
zebrafish homogenate. ETa: 45 min/Tb: 28 °C aflatoxicol-glucuronide AFB1
Wiegand et al., 2001 Embryos (fertilized eggs; Characterize glutathione S-transferases In vitro. Enzyme assay with zebrafish's HPLC-UV-ESI-QqQ Zebrafish embryos conjugate Phase I metabolism has not been
prim 6 stage embryos) (GST)-formed atrazine metabolites embryos. ETa: 24 h/Tb: 35 °C atrazine to glutathione by the evaluated, although in animals,
GSTs in the same way that hydroxylation and N-dealkylation
plants do are described as the main
metabolism pathway.
Lindholst et al., 2003 Adult (both sexes; age Metabolism of bisphenol A in zebrafish In vivo. Exposure in water. Water and HPLC-ESI-MSD Zebrafish produced the The high concentration of BPAGA
not informed) (Danio rerio) tissue. ETa: 7 days Tb: (27 ± 1)°C metabolites bisphenol A found in the bile indicates that
glucuronic acid (BPAGA) and biliary excretion into the intestines is
bisphenol A sulfate a major route of elimination.
Li et al., 2009 Larvae (2 dpf) Zebrafish CYP3A4 and CYP2D6 In vitro. Microplate whole zebrafish CYP's specific CYP's functional activity was up These findings underscore the high
functional activity assays for assessing assay with 0.1% DMSO. ETa: 24 h Tb: chemiluminogenic and downregulated in zebrafish degree of CYP conservation in
drug metabolism Not informed substrate similar to the response in zebrafish.
humans.
Alderton et al., 2010 Larvae (7 dpf) Accumulation and metabolism of drugs In vivo. Exposure in medium with HPLC-ESI-MS/MS Cisapride, chlorpromazine, Metabolism of amodiaquine,
commonly used as probes for human DMSO 1% (v/v). Medium and larvae verapamil, testosterone, and midazolam, felodipine, and S-
cytochrome P450s (CYPs) in zebrafish analyzed. ETa: 4 h Tb: (28.5 ± 0.5)°C dextromethorphan showed that mephenytoin could not be detected.
larvae the zebrafish larvae catalyze a Both similarities and differences in
range of phase I and phase II the metabolism were observed in
reactions. zebrafish when compared to
mammals
Jones et al., 2010 Larvae (4 dpf) Identify genes homologous to In vivo Exposure in water with 0.1% of Fluorescence ECODc, Zebrafish larvae express genes The continued use of these model
mammalian CYP's and UDP-UGT genes DMSO ETa: 10 h Tb: (28 ± 1)°C ERODd and OOMRe similar to mammalian CYP and organisms in toxicity testing is
and assess the ability of zebrafish to assay UGT isoforms throughout early supported by this study.
metabolize substrates for these development and have
enzymes activities toward model CYP
substrates
Smith et al., 2010 HPLC-APCI-MS/MS
(continued on next page)
Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46
Table 2 (continued)

Author Zebrafish's life stage Brief Experimental design Analytical technique Results Observations

a b
Adult (sexually mature, In vitro hepatic fluoxetine metabolism In vitro. Liver microsomes ET : 1 h T : Fluoxetine metabolism in fish is The low level of metabolism is likely
both sexes) was determined in several model fish 25 °C much less than that seen in the reason that fish bioaccumulate
species: rainbow trout, goldfish, mammals. The major fluoxetine in vivo. After killifish,
zebrafish and killifish. mammalian metabolite, zebrafish was the fish model with
C. de Souza Anselmo et al.

norfluoxetine, is not the higher rates of metabolism of


primary metabolite in fish fluoxetine.
Chen et al., 2012 Embryos (2 hpf) Determine if zebrafish larvae can be In vivo. Exposure in BDE solution with GC–MS Bioconcentration of BDE-209 in The limited metabolites of BDE-209
used to evaluate thyroid endocrine 0.01% of DMSO. Larvae analyse. ETa: the larvae was evident, and detected compared with previous
disruption of BDE-209 and if it 14 days Tb: (28 ± 0.5)°C BDE-209 could be metabolized. studies suggest lower metabolic
bioaccumulates and is metabolized. Both T4 and T3 levels were capability in larvae,
changed by BDE-209 exposure
in the larvae.
Chng et al., 2012 Larvae (5 dpf) and adult Investigation of the bioactivation In vitro/in vivo. Human (HLM) and HPLC-MS/MS/HPLC- NAPQI (APAP's reactive The ZLM overall metabolite profile
(age not informed; potential and metabolism profile of adult zebrafish's (ZLM) liver QTOF/MS/MS metabolite) was generated by of testosterone had more
female) zebrafish versus human using microsomes; larvae exposure in ZLM at lower levels than HLM. hydroxylated metabolites compared
acetaminophen (APAP) and medium with methanol 0.1% (v/v) ETa Several metabolites for with HLM. The zebrafish larvae
testosterone (in vitro): 2 h/Tb: 28.5 °C. ETa (in vivo): testosterone were identified in produced only two hydroxilated
3 h/Tb: 28.5 °C. ZLM and larvae. metabolites
Hu et al., 2012 Larvae (2 dpf) Characterize the metabolic profile of In vivo. Exposure in medium with 0.1% HPLC-MS/MS A total of ten calycosin The predominant phase II
calycosin in a zebrafish model DMSO. Medium and larvae analyzed. metabolites formed from conjugation of calycosin observed in
ETa: 24 h Tb: 28.5 °C. glucuronidation, glucosylation, zebrafish larvae were similar to the
sulfation, oxidation, metabolic profiles of other
demonstrating existence of both flavonoids in mammals
phase I and phase II metabolic
activities.
Jones et al., 2012 Larvae (3 dpf) Determine if zebrafish larvae could In vivo. Exposure in water with ethanol HPLC-LTQ-orbitrap- Hydroxy-ibuprofen was also Zebrafish larvae (72–96 hpf) can

39
metabolize ibuprofen (CYP2C 0.0015% (v/v). Water and fish MS/MS identified in larvae extracts and absorb, oxidize and excrete
substrate) that has well characterised analyzed ETa: 24 h Tb: (28 ± 1)°C water ibuprofen, similar to that identified
metabolism in mammals in mammalian systems.
Carlsson et al., 2013 Embryos (fertilized eggs Potential risk to fish embryos of In vivo. Exposure in medium with 0.1% HPLC-ESI-MS/MS AL was metabolized efficiently The results also demonstrate the
15 min after collection) veterinary pharmaceuticals were DMSO. Medium analyzed. ETa: 6 days into albendazole sulfoxide, great value of including sublethal
investigated and metabolism by the Tb: (25.1 ± 0.4)°C resulting in reduced toxicity. endpoints and chemical analysis to
embryos was studied for albendazole The prodrug febantel was the zebrafish embryo test for hazard
(AL), febantel, fenbendazole and metabolized to fenbendazole identification and risk assessment
oxfendazole. which is further metabolized to
oxfendazole in various species.
Pardal et al., 2014 Larvae (3 dpf) Metabolism of resveratrol and its In vivo. Exposure in medium with 1% HPLC-UV-MS/MS The principal metabolites found Besides the metabolites found in this
glucoside (piceid) in zebrafish DMSO. Medium and larvae analyzed. were monoglucoronide and study for piceid, it's already been
ETa: 48 h Tb: 27 °C monosulfate forms of identified glucuronidated and
resveratrol. Other metabolites sulfated piceid in rat urine after oral
found in the literature were not administration.
detected in zebrafish larvae
Wen et al., 2015 Adults (20 weeks, both Evaluate potential risks posed by BDE- In vivo. Adult exposure by spiked food. GC–MS/MS Concentrations of compounds Transfer of residues from adult
sexes) and eggs 47 and their analogs in aquatic Adults and eggs analyzed. ETa: 20 days in the liver of males were females to eggs was thought to be
(collected every environment, distribution among Tb: (28 ± 1)°C higher than in females. one of the main factors for the sex-
morning before feeding) tissues of adult fish and transfer from Significant transfer of all dependent difference in
females to eggs compounds from adult females concentrations in liver.
to eggs was observed. Several
metabolites formed.
Vallverdú-Queralt Embryos (fertilized eggs; To provide accurate and In vivo. Exposure in medium. Embryos HPLC-LTQ-Orbitrap- Quercetin-O-glucoronide and This research constitutes the first
et al., 2015 24 hpf) comprehensive identification of the analyzed ETa: 24 h Tb: (27 ± 1)°C MS anthocyanin metabolites was comprehensive identification of
phenolic constituents found in wine detected in embryos. phenolic compounds in zebrafish by
extracts and zebrafish embryos HPLC-HRMS
Brox et al., 2016 HPLC-QTOF-MS
(continued on next page)
Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46
Table 2 (continued)

Author Zebrafish's life stage Brief Experimental design Analytical technique Results Observations

Embryos (fertilized eggs; Study of xenobiotic metabolism of In vivo. Exposure in medium. Medium Eighteen metabolites was The results of this study outline the
incubated until 72 hpf zebrafish embryo (ZE) using clofibric and embryos analyzed. ETa: 24 h Tb: detected formed by phase I and high metabolic potential of the ZE
before use) acid and comparing it with the (26 ± 1)°C phase II metabolism. with respect to the transformation of
metabolism in other primarily higher Transformation of clofibric acid xenobiotics. Similarities but also
C. de Souza Anselmo et al.

organisms. involves conjugation with differences to other test systems


sulfate or glucuronic acid, were observed.
carnitine, taurine, and
aminomethanesulfonic acid.
Anselmo et al., 2017 Adult (age and sex not The ability of adult zebrafish to In vivo. Exposure in water. Water HPLC-QExactive- Adult zebrafish could produce This study demonstrates that adult
informed) produce metabolites of sibutramine analyzed ETa: 7 days Tb: (26 ± 2)°C Orbitrap-MS several sibutramine and zebrafish can absorb, oxidize, and
and stanozolol, substances with a well- stanozolol metabolites, excrete several metabolites in a
known metabolism that are widely including demethylated, manner similar to humans.
used as doping agents in sports, was hydroxylated, dehydroxylated,
evaluated. and reduced derivatives, all of
which have already been
detected in human
Shen et al., 2017 Adult (24–40 weeks; Metabolite profiles of ginsenosides Rk1 In vivo. Exposure in water 0.5% DMSO. HPLC-QTOF-MS Several metabolites of Rk1 (4) Desugarization, glucuronidation,
both sexes) and Rg5 from red ginseng or red Medium and embryos analyzed. ETa: and Rg5 (7), were identified. sulfation, and dehydroxy-
notoginseng in zebrafish were 24 h Tb: (23 ± 1)°C There are glucuronidation, methylation was observed.
qualitatively analyzed sulfation, dihydroxylation and Dehydroxylation and loss of C-17
dehydroxy-methylation residue were also metabolic
products in metabolites of Rk1 pathways of ginsenoside Rg5
and/or Rg5.
Saad et al., 2017 Different developmental Drug metabolism human CYP-specific In vitro. Liver microsomes with < 0.5% HPLC-MS/MS Adult zebrafish produced the No sex-related differences were
stages (5 and 24 hpf, 2, substrates: dextromethorphan (DXM), (v/v) DMSO. ETa: 2 h Tb: 28.5 °C two major human metabolites detected, except for the higher TST
3, 4 and 5 dpf) and adult diclofenac (DIC), testosterone (TST) of DIC and DXM. For DIC the depletion rate in adult females.

40
(age not informed; both and midazolam (MDZ) was metabolite ratio was similar to Zebrafish embryos and larvae
sexes) investigated in adult (both sexes), that in man, whereas it was showed no or low biotransformation
embryos and larvae different for DXM. For TST, the capacity.
major human metabolite could
not be detected and MDZ was
not metabolized.
G. Wang et al., 2017 Adult (20 weeks; both The metabolism of six In vivo. Exposure in water 0.01% HPLC-QTOF-MS Twenty main metabolites were The reaction pathways involving
sexes) organophosphate (OP) flame DMSO. Water and fish analyzed ETa: detected in the liver of OPs- scission of the ester bond
retardants was investigated in adult 19 days Tb: (24 ± 1)°C exposed zebrafish using high (hydrolysis), cleavage of the ether
zebrafish resolution mass spectrometry bond, oxidative hydroxylation,
dechlorination, and coupling with
glucuronic acid
C. Wang et al., 2017 Larvae (7 dpf) Zebrafish as the biotransformation In vivo. Exposure in water 0.5% DMSO. HPLC-QTOF-MS 46 metabolites screened from 37 of the metabolites were identified
system for rapid identification of Water analyzed. ETa: 24 h Tb: 28.5 °C water samples of zebrafish and validated. 75% of the identified
metabolites derived from herbal treated with total Epimedium EFs metabolites were successfully
compounds flavonoids (EFs) could be detected in urine samples of rats
matched with their
corresponding parent
compounds.

a
Exposure time.
b
Temperature; dpf: days post-fertilization; hpf: hours post-fertilization. ECOD.
c
ECOD Assay: The metabolism of 7-ethoxycoumarin to 7-hydroxycoumarin as a general marker for CYP2 activities in mammals; EROD.
d
EROD Assay: The metabolism of 7-ethoxyresorufin to resorufin as a general marker for CYP1 activities in mammals; OOMR.
e
OOMR Assay: The conversion of octyloxymethylresorufin to resorufin was used as a substrate for CYP3 activities in mammals.
Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

Fig. 2. Overview of zebrafish experiments in relation to the type of experiment (a), life stage (b), exposure time (c) and use of organic solvents (d).

zebrafish was capable of performing this reaction (Wiegand et al., BDE, tetra-BDE, penta-BDE, hex-BDE, hepta-BDE and octa-BDE after
2001). In vitro experiments were performed with enzyme extracts from exposure for 5 months (He et al., 2011). The limited production of BDE-
zebrafish embryos and atrazine being showed that atrazine-glutathione 209 metabolites in this study (Chen et al., 2012) compared with He
conjugates products were formed. In this work, phase I metabolism had et al. (2011) can suggest lower metabolic capability in larvae, but may
not been evaluated, although in animals, hydroxylation and N-deal- also be due to the different exposure times and species used. Both T4
kylation are described as the main metabolic pathways. A study of the and T3 levels were changed by BDE-209 exposure in the larvae, sug-
metabolism of bisphenol A, an endocrine disrupter which is used in the gesting thyroid disruption (Chen et al., 2012). Other work evaluated
production of plastics, has shown that the zebrafish produced the phase maternal transfer, distribution, and metabolism of BDE-47 and its re-
II metabolites bisphenol A glucuronic acid and bisphenol A sulfate after lated hydroxylated, methoxylated analogs in zebrafish, because analogs
exposure of adult animals in water (Lindholst et al., 2003). Another may be more toxic than PBDEs (Wen et al., 2015). Unlike the other
toxicological analysis performed on zebrafish was with carcinogenic studies, adult fish were exposed by spiked food in this study. The results
agents. Zebrafish were used as alternative in vivo and in vitro models for indicated that the concentrations of all compounds in the liver of male
chemical carcinogenesis studies of aflatoxin B1 (AFB1) (Troxel et al., zebrafish were significantly greater than those in the liver of females,
1997). The major metabolites of AFB1 recovered in the water at various suggesting transfer of the residues from adult females to eggs, with
time points after adult intraperitoneal injection were identified as several metabolites formed in adult fish. More recently, G. Wang et al.
aflatoxicol (predominant metabolite, produced by a cytosolic reductase (2017) evaluated the in vivo metabolism of six typical organophosphate
reaction) and aflatoxicol-glucuronide. The in vitro assay assessing AFB1 (OP) flame retardants using adult zebrafish. In this work, they found
metabolism again demonstrated the proficiency of AFL formation in several phase I and phase II metabolites, including OP diesters, hy-
zebrafish and produced AFB1-8,9-epoxide, which is responsible for the droxylated OP triesters, hydroxylated OP diesters, and glucuronic acid
carcinogenic effects. Male zebrafish did appear to have lower activity conjugated metabolites, after hydroxylation.
toward the formation of the AFB1–epoxide than females. This pub- In 2009, the first study to evaluate the zebrafish CYP response to
lication demonstrated not only the ability of zebrafish to generate phase drugs used to treat humans was carried out, seeking an in vivo com-
I and II metabolites but also sex differences in relation to the metabo- parison between the species. Li et al. (2009) developed a microplate
lism of xenobiotics. whole zebrafish larvae (2 dpf) assay to evaluate zebrafish CYP3A4 and
Due to their low cost and high performance, polybrominated di- CYP2D6 functional activity with various specific substrates of these
phenyl ethers (PBDEs) have been widely used for many years as flame enzymes. The overall prediction success rate was 86% for CYP3A4 in-
retardants in various commercial products and are an environmental hibition and induction in zebrafish, 100% for CYP3A4 inhibition and
problem. As PBDEs have similar structures to those of thyroid hormones 71% for CYP3A4 induction (lovastatin and phenytoin did not induce
(THs), they have the potential to disrupt thyroid endocrine activities; CYP3A4 in zebrafish as they do in humans); zebrafish CYP2D6 was
thus, it is important to understand their in vivo metabolism (Chen et al., upregulated by treatment with dexamethasone, 2,4-di-
2012; Wen et al., 2015). After 14 days of exposure, the bioconcentra- chlorophenoxyacetic acid and ethanol. These results highlight the high
tion of BDE-209 in the larvae was evident and BDE-209 could also be degree of CYP conservation in zebrafish. After this work, several others
metabolized, with the main metabolite being nona-BDE. Other BDE have succeeded in evaluating drug metabolism in zebrafish.
metabolites have already been identified in zebrafish, including tri- Alderton et al. (2010) examined the accumulation and metabolism

41
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

of a number of drugs and commonly used probes for human CYPs in v) (Jones et al., 2012). Ibuprofen metabolism is well described in hu-
zebrafish larvae (7 dpf) under conditions relevant to pharmacological mans, involving oxidation of the parent compound to hydroxy-ibu-
and toxicological assays. Metabolism studies with cisapride, chlorpro- profen and carboxy-ibuprofen by CYP2C8/9, followed by conjugation
mazine, verapamil, testosterone, and dextromethorphan showed that with glucuronic acid. Although no CYP2C-corresponding orthologs
the zebrafish larvae catalyze several phase I (oxidation, N-demethyla- have been identified in zebrafish, it has been able to generate hydroxy-
tion, O-de-ethylation, hydroxylation and N-dealkylation) and phase II ibuprofen and other hydroxylated derivatives. However, the presence of
(sulfation and glucuronidation) reactions, where both similarities and carboxy-ibuprofen and glycoconjugate derivatives was not observed,
differences in the metabolic pathways were observed in zebrafish larvae revealing differences between zebrafish and human metabolism of
when compared to mammals. Additionally, the metabolism of phena- ibuprofen.
cetin to paracetamol and dextromethorphan to dextrorphan (metabolic Several other studies have observed the in vivo production of phase I
reactions catalyzed by CYP1A2 and CYP2D6 in humans, respectively) and II metabolites during different life stages of zebrafish using the
was observed in the zebrafish larvae. Metabolism of amodiaquine, following various substances, showing once again that the zebrafish is a
midazolam, felodipine, and S-mephenytoin could not be detected under promising model that shares similarities and differences with mammals:
the conditions used in these experiments. When zebrafish larvae were flavonoids (Hu et al., 2012), veterinary pharmaceuticals (albendazole,
exposed to cisapride, the only metabolite observed was cisapride N- febantel, fenbendazole and oxfendazole) (Carlsson et al., 2013), re-
sulfate, which is a minor metabolite in mammalian species observed sveratrol (Pardal et al., 2014), phenolic constituents found in wine
only in the dog. Although some hydroxylation of testosterone (very (Vallverdú-Queralt et al., 2015), clofibric acid (Brox et al., 2016) and
low) was observed in the current experiments, the predominant meta- ginsenosides Rk1 and Rg5 (Shen et al., 2017). Pardal et al. (2014)
bolic pathway of testosterone observed in zebrafish larvae was glu- evaluated the metabolism of the phenolic compounds resveratrol and
curonidation. In addition to that, the amount of any specific metabolite its glucoside derivative (piceid) in vivo after exposure of zebrafish
formed was always much lower than that of the parent compound. All larvae in 1% (v/v) DMSO medium. The principal metabolites found in
these factors may indicate that the high amount of DMSO used (1%) in zebrafish larvae were monoglucuronide and monosulfate forms of re-
this work may have influenced the results, since it has already been sveratrol (the most abundant metabolite), which is in agreement with
demonstrated that low concentrations of organic solvents may influence other mammals, including human. In the case of the piceid group, the
the activity of zebrafish phase I and II enzymes (David et al., 2012). most abundant metabolite was the sulfated form of resveratrol, even if
Jones et al. (2010) have assessed the expression of genes that have it appeared later. In this work, it was not possible to identify glucur-
been identified as similar to mammalian CYP1A, CYP2B6, CYP3A5, and onidated or sulfated piceid and other metabolites found in rat urine,
UGT1A1, which are considered key mammalian families responsible for possibly for one of the following reasons: inability of the zebrafish
the oxidative metabolism of a variety of pharmaceuticals. Overall, this model to generate these metabolites; minor compounds that cannot be
study provided further evidence that zebrafish larvae express genes and detected under the experimental conditions; high amount of DMSO
proteins with activities that are responsible for xenobiotic metabolism used that may have interfered in the metabolism.
with both oxidative and conjugative metabolism similar to those of Brox et al. (2016) evaluated the metabolism of clofibric acid using
mammalian systems. Another study evaluated the in vitro hepatic zebrafish embryos and compared it with the metabolism in other, pri-
fluoxetine metabolism of adult zebrafish compared with several others marily higher organisms. A total of 18 metabolites of clofibric acid were
model fish: rainbow trout, goldfish and killifish (Smith et al., 2010). detected, including reactions of hydroxylation, demethylation, hydro-
After killifish, zebrafish was the fish model with the highest rates of lysis, methylation, decarboxylation and conjugation with sulfate and
metabolism of fluoxetine. In humans, fluoxetine is primarily metabo- glucuronic acid. In addition, conjugation with taurine, aminometha-
lized into norfluoxetine (major metabolite) through demethylation by nesulfonic acid, and carnitine represents new phase II conjugates that
CYP2D6 (has no ortholog in zebrafish, Table 1) (Goldstone et al., 2010) have not been described in zebrafish before this work. In humans, the
and to a lesser extent, by CYPs 2C9, 3A4 and 2C19. In zebrafish, the loss acyl glucuronide of clofibric acid (also generated by zebrafish) re-
of fluoxetine is much greater than the production of the demethylated presents the major metabolite, and taurine-conjugated clofibric acid
metabolite, norfluoxetine, suggesting that there may be production of was previously detected in dogs, cats, and ferrets, and now in zebrafish
multiple fluoxetine metabolites in fish and that norfluoxetine is unlikely but not in humans.
to be the primary fluoxetine metabolite, as it is in humans (Smith et al., Anselmo et al. (2017) evaluated the ability of adult zebrafish to
2010). produce metabolites of sibutramine and stanozolol after in vivo ex-
Chng et al. (2012) progressed forward and investigated the bioac- posure, with the focus of using the model to search for analytical targets
tivation potential and metabolism profile of zebrafish (in vitro and in for the detection of doping abuse. Several sibutramine and stanozolol
vivo) versus human (in vitro) using acetaminophen (APAP) and testos- metabolites were identified, including demethylated, hydroxylated,
terone. The hepatotoxicity of APAP is attributed to its reactive meta- dehydroxylated, and reduced derivatives and stanozolol conjugates, all
bolite, NAPQI (N-acetyl-p-benzoquinone imine), that was generated by of which have already been detected in human urine. This study sug-
adult zebrafish liver microsomes (ZLM) (possibly by CYP3A65), albeit gests that adult zebrafish can absorb, oxidize, and excrete several me-
at lower levels than by human liver microsomes (HLM). Several hy- tabolites in a manner similar to humans.
droxylated metabolites of testosterone were identified, including 2α-, Saad et al. (2017) made an important contribution by comparing
6β-, and 16β hydroxytestosterone and three putative metabolites (M2, the in vitro drug metabolism of human CYP-specific substrates at dif-
M3, and M7) in the ZLM and only 6β-hydroxytestosterone, testosterone ferent life stages of zebrafish with humans. Several CYP-related reac-
glucuronide, and M8 in zebrafish larvae homogenate and media. Al- tions in adult zebrafish using human CYP2- and CYP3-specific sub-
though the main metabolite formed by zebrafish was 6β-hydro- strates were detected. No in vitro biotransformation of the tested
xytestosterone, the overall metabolite profile was distributed across substrates was observed during zebrafish embryonic development until
more hydroxylated metabolites compared with that of HLM, and a 96 hpf (end of organogenesis). Unlike other studies, the bio-
number of unique hydroxytestosterone metabolites were observed only transformation rates were predominantly higher in zebrafish compared
in zebrafish. This result suggests a difference in either the function or to humans, except for CYP2D6-like activity. Sex-related differences
the expression of the drug-metabolizing enzymes between zebrafish were only present for testosterone. Diclofenac biotransformation in
larvae and adult zebrafish, in addition to demonstrating differences zebrafish into 4- and 5-OH-diclofenac reveals similar biotransformation
between human and zebrafish in vitro testosterone metabolism. pathways compared to humans by the production of similar major
Another study evaluated the metabolism of ibuprofen in zebrafish metabolites with similar proportions in both species. However, dex-
larvae (3 dpf) after in vivo exposure in water with ethanol 0.0015% (v/ tromethorphan was also biotransformed into similar main metabolites

42
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

as in humans but with altered ratios as shown (dextrorphan/3-meth- et al., 2011). Thus, a major advantage of using the in vivo zebrafish
oxymorphinan). In this study, no production of the main testosterone model for predicting metabolism is that the matrix is cleaner than urine,
metabolite was observed, which was already reported in another work for example, widely used in other animal models, which reduces the
(Chng et al., 2012). For midazolam, neither consumption nor metabo- matrix effect.
lites could be detected in ZLM, whereas this substrate was largely me-
tabolized into 1-OH-midazolam and, to a much lesser extent, into 4-OH- 6. Zebrafish considerations for metabolite screening
midazolam in HLM. As a result, midazolam, a typical substrate for
CYP3A in man, appeared to have no affinity for any of the zebrafish In general, the studies that evaluated the production of xenobiotic
CYPs. Therefore, the in vitro CYP-mediated drug metabolism can be metabolites in zebrafish observed several different phase I and II me-
substantially different in zebrafish compared with man. tabolites, demonstrating the utility of the model in the prediction of the
C. Wang et al. (2017) proposed a rapid identification of metabolites metabolism of several substances. Therefore, there is further evidence
derived from herbal compounds after in vivo exposure of zebrafish that zebrafish express genes and proteins with activities responsible for
larvae (7 dpf), compared with metabolism in rats, a representative xenobiotic metabolism with both oxidative and conjugative metabolism
mammalian model. This work identified 46 potential metabolites that similar to mammalian systems. Regarding the zebrafish CYP orthologs
could be matched with their corresponding parent compounds, and 37 in humans, it is important to highlight that three cases were observed:
of them were identified and validated by the known metabolic path- no orthology with production of metabolites (fluoxetine (Smith et al.,
ways and fragmentation patterns. Among the 37 metabolites identified 2010), CYP2D6; and ibuprofen (Jones et al., 2012), CYP2C); presence of
in zebrafish, 28 were found in rat urine. This result encourages re- orthology without production of metabolites (midazolam (Saad et al.,
searchers to use zebrafish larvae as the biotransformation system for 2017), CYP3A) and presence of orthology with production of metabo-
preliminary metabolism study. lites (sibutramine and stanozolol (Anselmo et al., 2017), CYP2B6 and
CYP3A4, respectively), which is the ideal case and the most often
5. Metabolites analysis found. Even in cases where human orthologs of CYP are present in
zebrafish, divergences may occur in relation to the major metabolites
The first studies that carried out the detection of xenobiotic meta- generated or the production ratio of these metabolites in comparison
bolites in zebrafish used HPLC (high-performance liquid chromato- with humans (Alderton et al., 2010; Chng et al., 2012; Jones et al.,
graphy) with diode-array detection (DAD) analysis (Gorge and Nagel, 2012). So, further studies are needed to understand to what extent the
1990; Hertl and Nagel, 1993; Kasokat et al., 1987, 1989; Troxel et al., zebrafish is able to reproduce the metabolism observed in humans.
1997; Zok et al., 1991). HPLC-DAD provides a means for analysing Nevertheless, it is important to highlight that even the mouse, a classic
various compounds, but is limited by the non-specific nature of UV model for prediction of toxicity and evaluation of metabolites, presents
detection. On the other hand, gas chromatography–mass spectrometry some differences in relation to human metabolism (Lootens et al.,
(GC–MS), together with detailed GC–MS spectral libraries, is a very 2009). Therefore, this issue does not prevent zebrafish from being
useful tool for toxicological analysis, but non-volatile, polar and ther- considered a model with great potential for studying the metabolism of
mally labile compounds are difficult or impossible to analyse without a xenobiotics.
long derivatization procedure (Couchman and Morgan, 2011). Zebrafish larvae represent a very interesting alternative model to
An important contribution for analytical toxicology was the devel- animal experimentation, although it is important to remember that
opment of the electrospray ionization (ESI) (Fenn, 2002), which was early life stages (3 to 5 dpf) are still undergoing embryogenic processes
responsible for the coupling of liquid chromatography (LC) and mass and that their metabolic status is different from that of the young or
spectrometry (MS), expanding the detection range of new metabolites, adult zebrafish (Pardal et al., 2014). In addition, when using zebrafish
and increasing the detection capacity of substances even at low con- embryos, there is a need for them to take up substances through the
centrations. Triple quadrupole (QqQ) instruments operating in selected chorion and other membranes (Vallverdú-Queralt et al., 2015), which
reaction monitoring (SRM) mode are the most common mass analysers can generate results compromised by the differentiated absorption at
used in bioanalysis (Núñez et al., 2013), as can be observed in several this life stage. Moreover, it has already been observed that the activity
studies (Alderton et al., 2010; Carlsson et al., 2013; Chng et al., 2012; of most CYPs involved in xenobiotic metabolism is increased dramati-
Saad et al., 2017). However, for the detection by tandem-MS techni- cally after hatching approximately 3 dpf (Saad et al., 2016) and that the
ques, as the triple quadrupole, the potential metabolites must be pre- liver is fully formed at approximately 5 dpf (Kimmel et al., 1995). Al-
viously known or need to share a high degree of structural similarity though several studies have observed the production of metabolites in
with the parent compound to allow their determination by neutral loss the embryonic and larval stages of zebrafish (Table 2), Saad et al.
or precursor ion scan. However, these conditions are not necessarily (2017) observed no in vitro biotransformation during zebrafish em-
possible in metabolism studies (Brox et al., 2016). bryonic development until 96 hpf. Therefore, the age of the zebrafish is
An emerging approach for toxicological analysis is that of the de- a controversial issue and should be taken into account during experi-
termination of exact mass with high-resolution (HRMS). Through a full- mental design and when comparing the results with other mammals.
scan HRMS experiment at high mass accuracy (mass errors below In zebrafish in vivo screening models, the main route of exposure is
6 ppm), it is possible to very precisely filter the full-scan data and ex- dermal until approximately 5 dpf when the mouth is opened and both
tract analyte chromatograms with very low background noise, which dermal and enteral routes exist from 5 to 14 dpf while the embryo is
allows the identification of metabolites by knowledge of the elemental free-feeding (Garcia et al., 2016; Sipes et al., 2011). Usually, in con-
composition alone, without the need for reference material (Couchman ventional in vivo experiments, a known and measurable amount of drug
and Morgan, 2011). Time-of-flight (TOF) and Orbitrap-based technol- is administered to the animal. However, in the zebrafish in vivo models,
ogies are currently the most common analysers used in LC–HRMS the uptake from the water into the zebrafish is non-uniform and un-
(Núñez et al., 2013). In fact, studies using LC-HRMS to search for xe- predictable, which can make it difficult to interpret screening results
nobiotic metabolites have been able to identify and sometimes char- and to make comparisons between compounds (Fleming and Alderton,
acterize a greater number of substances (Anselmo et al., 2017; Brox 2013).
et al., 2016; Shen et al., 2017; C. Wang et al., 2017) compared to other Another important issue to be discussed in relation to zebrafish
techniques. The major challenge in the identification of metabolites by screening is the use of organic solvents to increase the solubility of the
LC-MS is the detection and structural elucidation of trace levels of substances tested and their impact on metabolism enzymes. David et al.
unknown metabolites in the presence of large amounts of complex in- (2012) observed that metabolism of ethoxyresorufin in zebrafish larvae
terfering ions of endogenous components, called matrix effect (Zhu was significantly reduced by dimethylsulfoxide (DMSO) and methanol

43
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

(0.1% and 0.05% v/v, respectively) after 24 h of exposure. The de- and function using differential fatty acid metabolism in live zebrafish. Dev. Biol. 360,
creases in activity of ethoxyresorufin were accompanied by decreased 276–285. https://doi.org/10.1016/j.ydbio.2011.09.010.
Chang, C.T., Chung, H.Y., Su, H.T., Tseng, H.P., Tzou, W.S., Hu, C.H., 2013. Regulation of
expression of various genes coding for drug metabolizing enzymes zebrafish CYP3A65 transcription by AHR2. Toxicol. Appl. Pharmacol. 270, 174–184.
corresponding to CYP1, CYP2, CYP3 and UGT family enzymes (David https://doi.org/10.1016/j.taap.2013.04.010.
et al., 2012). Thus, relatively low concentrations of organic solvents Chen, Q., Yu, L., Yang, L., Zhou, B., 2012. Bioconcentration and metabolism of deca-
bromodiphenyl ether (BDE-209) result in thyroid endocrine disruption in zebrafish
may impact the biotransformation of certain xenobiotics in zebrafish larvae. Aquat. Toxicol. 110–111, 141–148. https://doi.org/10.1016/j.aquatox.2012.
larvae, and this deserves consideration when evaluating substances for 01.008.
the study of metabolism and toxicity in this species. This fact, together Chng, H.T., Ho, H.K., Yap, C.W., Lam, S.H., Chan, E.C.Y., 2012. An investigation of the
bioactivation potential and metabolism profile of zebrafish versus human. J. Biomol.
with the age of the fish, the amount of substance used in the experiment Screen. 17, 974–986. https://doi.org/10.1177/1087057112447305.
and the unpredictable uptake from the exposure water, may be a de- Christen, V., Fent, K., 2014. Tissue-, sex- and development-specific transcription profiles
terminant in the metabolic profile that will be found and should be of eight UDP-glucuronosyltransferase genes in zebrafish (Danio rerio) and their reg-
ulation by activator of aryl hydrocarbon receptor. Aquat. Toxicol. 150, 93–102.
taken into account when designing and interpreting results from me-
https://doi.org/10.1016/j.aquatox.2014.02.019.
tabolite screening. Couchman, L., Morgan, P.E., 2011. LC-MS in analytical toxicology: some practical con-
siderations. Biomed. Chromatogr. 25, 100–123. https://doi.org/10.1002/bmc.1566.
7. Conclusion David, R.M., Jones, H.S., Panter, G.H., Winter, M.J., Hutchinson, T.H., Kevin Chipman, J.,
2012. Interference with xenobiotic metabolic activity by the commonly used vehicle
solvents dimethylsulfoxide and methanol in zebrafish (Danio rerio) larvae but not
In the last 30 years, the zebrafish has been used to predict the me- Daphnia magna. Chemosphere 88, 912–917. https://doi.org/10.1016/j.
tabolism of xenobiotics, with different focuses. In the early years, stu- chemosphere.2012.03.018.
Dawid, I.B., 2004. Developmental biology of zebrafish. Ann. N. Y. Acad. Sci. 1038, 88–93.
dies focused on environmental toxicology, and in the possible impact of https://doi.org/10.1196/annals.1315.015.
the generated metabolites on increasing the toxicity of the con- Diekmann, H., Hill, A., 2013. ADMETox in zebrafish. Drug Discov. Today Dis. Model. 10,
taminants. Currently, several studies have evaluated the metabolism of e31–e35. https://doi.org/10.1016/j.ddmod.2012.02.005.
European Commission, 2016. Legislation for the Protection of Animals Used for Scientific
drugs aiming to establish zebrafish as a model for toxicological Purposes - Environment - European Commission 1–2.
screening, either for the development of new drugs or for analytical Fenn, J.B., 2002. Electrospray ionization mass spectrometry: how it all began. J. Biomol.
targets in anti-doping control. These studies have shown that the zeb- Tech. 13, 101–118. https://doi.org/10.1126/science.2675315.
Fleming, A., Alderton, W.K., 2013. Zebrafish in pharmaceutical industry research: finding
rafish enzymes responsible for metabolism are analogous with those in the best fit. Drug Discov. Today Dis. Model. 10, e43–e50. https://doi.org/10.1016/j.
humans, performing several phase I and II reactions. Some factors need ddmod.2012.02.006.
to be considered for the use of the model, since they can directly in- Fleming, A., Sato, M., Goldsmith, P., 2005. High-throughput in vivo screening for bone
anabolic compounds with zebrafish. J. Biomol. Screen. 10, 823–831. https://doi.org/
fluence the results, such as: fish age, impact of the use of organic sol-
10.1177/1087057105279952.
vents and time of exposure. In addition, a limitation of the in vivo model Garcia, G.R., Noyes, P.D., Tanguay, R.L., 2016. Advancements in zebrafish applications
of zebrafish is that it is not possible to control the amount of substance for 21st century toxicology. Pharmacol. Ther. 161, 11–21. https://doi.org/10.1016/j.
administered by each fish and can generate non-reproducible results. pharmthera.2016.03.009.
Glisic, B., Mihaljevic, I., Popovic, M., Zaja, R., Loncar, J., Fent, K., Kovacevic, R., Smital,
As with any non-human model, zebrafish model presents similarities T., 2015. Characterization of glutathione-S-transferases in zebrafish (Danio rerio).
and differences in relation to the profile of metabolites generated Aquat. Toxicol. 158, 50–62. https://doi.org/10.1016/j.aquatox.2014.10.013.
compared to that observed in humans. All the facts presented indicate Goessling, W., Sadler, K.C., 2015. Zebrafish: an important tool for liver disease research.
Gastroenterology 149, 1361–1377. https://doi.org/10.1053/j.gastro.2015.08.034.
that zebrafish is a valuable potential model for the evaluation of xe- Goldstone, J.V., G, M.A., Kubota, A., Zanette, J., Parente, T., Jönsson, M.E., Nelson, D.R.,
nobiotic metabolism, but further studies are still needed to assess the Stegeman, J.J., 2010. Identification and developmental expression of the full com-
degree to which it can be compared to human metabolism. plement of Cytochrome P450 genes in Zebrafish. 11, 643. https://doi.org/10.1186/
1471-2164-11-643.
Gorge, G., Nagel, R., 1990. Kinetics and metabolism of 14C-lindane and 14C-atrazine in
Conflict of interest statement early life stage of zebrafish (Danio rerio). Chemosphere 21, 1125–1137. https://doi.
org/10.1038/028031d0.
Guengerich, F.P., 2008. Cytochrome P450 and chemical toxicology cytochrome P450 and
The authors declare that there are no conflicts of interest.
chemical toxicology. Chem. Res. Toxicol. 21, 70–83. https://doi.org/10.1021/
tx700079z.
References He, J., Yang, D., Wang, C., Liu, W., Liao, J., Xu, T., Bai, C., Chen, J., Lin, K., Huang, C.,
Dong, Q., 2011. Chronic zebrafish low dose decabrominated diphenyl ether (BDE-
209) exposure affected parental gonad development and locomotion in F1 offspring.
Alazizi, A., Liu, M.Y., Williams, F.E., Kurogi, K., Sakakibara, Y., Suiko, M., Liu, M.C., Ecotoxicology 20, 1813–1822.
2011. Identification, characterization, and ontogenic study of a catechol O-methyl- Hertl, J., Nagel, R., 1993. Bioconcentration and metabolism of 3,4-dichloroaniline in
transferase from zebrafish. Aquat. Toxicol. 102, 18–23. https://doi.org/10.1016/j. different life stages of guppy and zebrafish. Chemosphere 27, 2225–2234.
aquatox.2010.12.016. Ho, S.Y., Pack, M., Farber, S.A., 2003. Analysis of small molecule metabolism in zebrafish.
Alderton, W., Berghmans, S., Butler, P., Chassaing, H., Fleming, A., Golder, Z., Richards, Methods Enzymol. 364, 408–426. https://doi.org/10.1016/S0076-6879(03)64023-1.
F., Gardner, I., 2010. Accumulation and metabolism of drugs and CYP probe sub- Howe, K., Clark, M.D., Torroja, C.F., Torrance, J., Berthelot, C., Muffato, M., Collins, J.E.,
strates in zebrafish larvae. Xenobiotica 40, 547–557. https://doi.org/10.3109/ Humphray, S., McLaren, K., Matthews, L., McLaren, S., Sealy, I., Caccamo, M.,
00498254.2010.493960. Churcher, C., Scott, C., Barrett, J.C., Koch, R., Rauch, G.J., White, S., Chow, W.,
Almazroo, O.A., Miah, M.K., Venkataramanan, R., 2017. Drug metabolism in the liver. Kilian, B., Quintais, L.T., Guerra-Assunção, J.A., Zhou, Y., Gu, Y., Yen, J., Vogel, J.H.,
Clin. Liver Dis. 21, 1–20. https://doi.org/10.1016/j.cld.2016.08.001. Eyre, T., Redmond, S., Banerjee, R., Chi, J., Fu, B., Langley, E., Maguire, S.F., Laird,
Anselmo, C.S., Sardela, V.F., Matias, B.F., de Carvalho, A.R., de Sousa, V.P., Pereira, G.K., Lloyd, D., Kenyon, E., Donaldson, S., Sehra, H., Almeida-King, J., Loveland, J.,
H.M.G., de Aquino Neto, F.R., 2017. Is zebrafish (Danio rerio) a tool for human-like Trevanion, S., Jones, M., Quail, M., Willey, D., Hunt, A., Burton, J., Sims, S., McLay,
metabolism study? Drug Test. Anal. 9 (11-12), 1685–1694. https://doi.org/10.1002/ K., Plumb, B., Davis, J., Clee, C., Oliver, K., Clark, R., Riddle, C., Eliott, D.,
dta.2318. Threadgold, G., Harden, G., Ware, D., Mortimer, B., Kerry, G., Heath, P., Phillimore,
Brox, S., Seiwert, B., Haase, N., Küster, E., Reemtsma, T., 2016. Metabolism of clofibric B., Tracey, A., Corby, N., Dunn, M., Johnson, C., Wood, J., Clark, S., Pelan, S.,
acid in zebrafish embryos (Danio rerio) as determined by liquid chromatography-high Griffiths, G., Smith, M., Glithero, R., Howden, P., Barker, N., Stevens, C., Harley, J.,
resolution-mass spectrometry. Comp. Biochem. Physiol. C Toxicol. Pharmacol. Holt, K., Panagiotidis, G., Lovell, J., Beasley, H., Henderson, C., Gordon, D., Auger,
185–186, 20–28. https://doi.org/10.1016/j.cbpc.2016.02.007. K., Wright, D., Collins, J., Raisen, C., Dyer, L., Leung, K., Robertson, L., Ambridge, K.,
Brugman, S., 2016. The zebrafish as a model to study intestinal inflammation. Dev. Comp. Leongamornlert, D., McGuire, S., Gilderthorp, R., Griffiths, C., Manthravadi, D.,
Immunol. 64, 82–92. https://doi.org/10.1016/j.dci.2016.02.020. Nichol, S., Barker, G., Whitehead, S., Kay, M., Brown, J., Murnane, C., Gray, E.,
Capiotti, K.M., Antonioli, R., Kist, L.W., Bogo, M.R., Bonan, C.D., Da Silva, R.S., 2014. Humphries, M., Sycamore, N., Barker, D., Saunders, D., Wallis, J., Babbage, A.,
Persistent impaired glucose metabolism in a zebrafish hyperglycemia model. Comp. Hammond, S., Mashreghi-Mohammadi, M., Barr, L., Martin, S., Wray, P., Ellington,
Biochem. Physiol. B Biochem. Mol. Biol. 171, 58–65. https://doi.org/10.1016/j.cbpb. A., Matthews, N., Ellwood, M., Woodmansey, R., Clark, G., Cooper, J., Tromans, A.,
2014.03.005. Grafham, D., Skuce, C., Pandian, R., Andrews, R., Harrison, E., Kimberley, A.,
Carlsson, G., Patring, J., Kreuger, J., Norrgren, L., Oskarsson, A., 2013. Toxicity of 15 Garnett, J., Fosker, N., Hall, R., Garner, P., Kelly, D., Bird, C., Palmer, S., Gehring, I.,
veterinary pharmaceuticals in zebrafish (Danio rerio) embryos. Aquat. Toxicol. 126, Berger, A., Dooley, C.M., Ersan-Ürün, Z., Eser, C., Geiger, H., Geisler, M., Karotki, L.,
30–41. https://doi.org/10.1016/j.aquatox.2012.10.008. Kirn, A., Konantz, J., Konantz, M., Oberländer, M., Rudolph-Geiger, S., Teucke, M.,
Carten, J.D., Bradford, M.K., Farber, S.A., 2011. Visualizing digestive organ morphology Osoegawa, K., Zhu, B., Rapp, A., Widaa, S., Langford, C., Yang, F., Carter, N.P.,

44
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

Harrow, J., Ning, Z., Herrero, J., Searle, S.M.J., Enright, A., Geisler, R., Plasterk, in fast liquid chromatography-mass spectrometry for bio-analytical applications. J.
R.H.A., Lee, C., Westerfield, M., De Jong, P.J., Zon, L.I., Postlethwait, J.H., Nüsslein- Chromatogr. B Anal. Technol. Biomed. Life Sci. 927, 3–21. https://doi.org/10.1016/
Volhard, C., Hubbard, T.J.P., Crollius, H.R., Rogers, J., Stemple, D.L., 2013. The j.jchromb.2012.12.031.
zebrafish reference genome sequence and its relationship to the human genome. OECD, 1992. Fish, Acute Toxicity test. OECD Guidel. Test. Chem. https://doi.org/10.
Nature 496, 498–503. https://doi.org/10.1038/nature12111. 1787/9789264069961-en.
Hu, G., Siu, S.O., Li, S., Chu, I.K., Kwan, Y.W., Chan, S.W., Leung, G.P.H., Yan, R., Lee, OECD, 2013. Test No. 236: Fish Embryo Acute Toxicity (FET) Test 1-22. https://doi.org/
S.M.Y., 2012. Metabolism of calycosin, an isoflavone from Astragali Radix, in zeb- 10.1787/9789264203709-en.
rafish larvae. Xenobiotica 42, 294–303. https://doi.org/10.3109/00498254.2011. Otte, J.C., Schultz, B., Fruth, D., Fabian, E., Van, B.R., Hidding, B., Salinas, E.R., 2017.
617015. Intrinsic xenobiotic metabolizing enzyme activities in early life stages of zebrafish
Huang, H., Wu, Q., 2010. Cloning and comparative analyses of the zebrafish Ugt re- (Danio rerio). Toxicol. Sci. 159, 86–93.
pertoire reveal its evolutionary diversity. PLoS One 5. https://doi.org/10.1371/ Pardal, D., Caro, M., Tueros, I., Barranco, A., Navarro, V., 2014. Resveratrol and piceid
journal.pone.0009144. metabolites and their fat-reduction effects in zebrafish larvae. Zebrafish 11, 32–40.
Jones, H.S., Panter, G.H., Hutchinson, T.H., Chipman, J.K., 2010. Oxidative and con- https://doi.org/10.1089/zeb.2013.0893.
jugative xenobiotic metabolism in zebrafish larvae in vivo. Zebrafish 7, 23–30. Peters, F.T., Martinez-Ramirez, Jorge A., 2010. Analytical toxicology of emerging drugs of
https://doi.org/10.1089/zeb.2009.0630. abuse. Ther. Drug Monit. 32, 532–539. https://doi.org/0.1097/FTD.
Jones, H.S., Trollope, H.T., Hutchinson, T.H., Panter, G.H., Chipman, J.K., 2012. 0b013e31826d0915.
Metabolism of ibuprofen in zebrafish larvae. Xenobiotica 42, 1069–1075. https://doi. Pinho, B.R., Reis, S.D., Guedes-Dias, P., Leitão-Rocha, A., Quintas, C., Valentão, P.,
org/10.3109/00498254.2012.684410. Andrade, P.B., Santos, M.M., Oliveira, J.M.A., 2016. Pharmacological modulation of
Kamel, M., Ninov, N., 2017. Catching new targets in metabolic disease with a zebrafish. HDAC1 and HDAC6 in vivo in a zebrafish model: therapeutic implications for
Curr. Opin. Pharmacol. 37, 41–50. https://doi.org/10.1016/j.coph.2017.08.007. Parkinson's disease. Pharmacol. Res. 103, 328–339. https://doi.org/10.1016/j.phrs.
Kasokat, T., Nagel, R., Urich, K., 1987. The metabolism of phenol and substituted phenols 2015.11.024.
in zebra fish. Xenobiotica 17, 1215–1221. https://doi.org/10.3109/ Ree, R., Myklebust, L.M., Thiel, P., Foyn, H., Fladmark, K.E., Arnesen, T., 2015. The N-
00498258709047175. terminal acetyltransferase Naa10 is essential for zebrafish development. Biosci. Rep.
Kasokat, T., Nagel, R., Urich, K., 1989. Metabolism of chlorobenzene and hexa- 35, 1–10. https://doi.org/10.1042/BSR20150168.
chlorobenzene by the zebra fish, Brachydanio rerio. Bull. Environ. Contam. Toxicol. Saad, M., Cavanaugh, K., Verbueken, E., Pype, C., Casteleyn, C., Van Ginneken, C., Van
42, 254–261. https://doi.org/10.1007/BF01699408. Cruchten, S., 2016. Xenobiotic metabolism in the zebrafish: a review of the spatio-
Kimmel, C.B., Ballard, W.W., Kimmel, S.R., Ullmann, B., Schilling, T.F., 1995. Stages of temporal distribution, modulation and activity of Cytochrome P450 families 1 to 3. J.
embryonic development of the zebrafish. Dev. Dyn. 203, 253–310. https://doi.org/ Toxicol. Sci. 41, 1–11. https://doi.org/10.2131/jts.41.1.
10.1002/aja.1002030302. Saad, M., Matheeussen, A., Bijttebier, S., Verbueken, E., Pype, C., Casteleyn, C., Van
Kithcart, A., MacRae, C.A., 2017. Using zebrafish for high-throughput screening of novel Ginneken, C., Apers, S., Maes, L., Cos, P., Van Cruchten, S., 2017. In vitro CYP-
cardiovascular drugs. JACC Basic Transl. Sci. 2, 1–12. https://doi.org/10.1016/j. mediated drug metabolism in the zebrafish (embryo) using human reference com-
jacbts.2017.01.004. pounds. Toxicol. in Vitro 42, 329–336. https://doi.org/10.1016/j.tiv.2017.05.009.
Kühnert, A., Vogs, C., Aulhorn, S., Altenburger, R., Küster, E., Busch, W., Kühnert, A., Scornaienchi, M.L., Thornton, C., Willett, K.L., Wilson, J.Y., 2010. Functional differences
Vogs, C., Altenburger, R., Hollert, H., Seiwert, B., 2017. Biotransformation in the in the cytochrome P450 1 family enzymes from Zebrafish (Danio rerio) using het-
zebrafish embryo –temporal gene transcription changes of cytochrome P450 enzymes erologously expressed proteins. Arch. Biochem. Biophys. 502, 17–22. https://doi.
and internal exposure dynamics of the AhR binding xenobiotic benz[a]anthracene. org/10.1016/j.abb.2010.06.018.
Environ. Pollut. 230, 1–11. https://doi.org/10.1016/j.envpol.2017.04.083. Shams, S., Rihel, J., Ortiz, J.G., Gerlai, R., 2017. The zebrafish as a promising tool for
Langheinrich, U., 2003. Zebrafish: a new model on the pharmaceutical catwalk. BioEssays modeling human brain disorders: a review based upon an IBNS Symposium. Neurosci.
25, 904–912. https://doi.org/10.1002/bies.10326. Biobehav. Rev. 0–1. https://doi.org/10.1016/j.neubiorev.2017.09.002.
Langheinrich, U., Vacun, G., Wagner, T., 2003. Zebrafish embryos express an orthologue Shen, W., Wei, Y., Tang, D., Jia, X., Chen, B., 2017. Metabolite profiles of ginsenosides
of HERG and are sensitive toward a range of QT-prolonging drugs inducing severe Rk1 and Rg5 in zebrafish using ultraperformance liquid chromatography/quad-
arrhythmia. Toxicol. Appl. Pharmacol. 193, 370–382. https://doi.org/10.1016/j. rupoleetime-of-flight MS. J. Ginseng Res. 41, 78–84. https://doi.org/10.1016/j.jgr.
taap.2003.07.012. 2015.12.010.
Lawrence, C., 2007. The husbandry of zebrafish (Danio rerio): a review. Aquaculture 269, Sipes, N.S., Padilla, S., Knudsen, T.B., 2011. Zebrafish-as an integrative model for twenty-
1–20. https://doi.org/10.1016/j.aquaculture.2007.04.077. first century toxicity testing. Birth Defects Res. C Embryo Today Rev. 93, 256–267.
Lee, K.J., Nallathamby, P.D., Browning, L.M., Osgood, C.J., Xu, X.N., 2007. In Vivo https://doi.org/10.1002/bdrc.20214.
imaging of transport and biocompatibility of single silver nanoparticles in early de- Smith, E.M., Chu, S., Paterson, G., Metcalfe, C.D., Wilson, J.Y., 2010. Cross-species
velopment of zebrafish embryos. ACS Nano 1 (2), 133–143. http://dx.doi.org/10. comparison of fluoxetine metabolism with fish liver microsomes. Chemosphere 79,
1021/nn700048y. 26–32. https://doi.org/10.1016/j.chemosphere.2010.01.058.
Li, C., Lin, W.S., McGrath, P., 2009. Whole zebrafish cytochrome P450 microplate assays Sneddon, L.U., Halsey, L.G., Bury, N.R., 2017. Considering aspects of the 3Rs principles
for assessing drug metabolism and drug safety. J. Pharmacol. Toxicol. Methods 60, within experimental animal biology. J. Exp. Biol. 220, 3007–3016. https://doi.org/
233. https://doi.org/10.1016/j.vascn.2009.04.103. 10.1242/jeb.147058.
Lindholst, C., Wynne, P.M., Marriott, P., Pedersen, S.N., Bjerregaard, P., 2003. Spitsbergen, J.M., Kent, M.L., 2003. The state of the art of the zebrafish model for tox-
Metabolism of bisphenol A in zebrafish (Danio rerio) and rainbow trout (Oncorhynchus icology and toxicologic pathology research—advantages and current limitations.
mykiss) in relation to estrogenic response. Comp. Biochem. Physiol. C 135, 169–177. Toxicol. Pathol. 31, 62–87. https://doi.org/10.1080/01926230390174959.
https://doi.org/10.1016/S1532-0456. Streisinger, George, Walker, Charline, Dower, Nancy, Knauber, Donna, Singer, F., 1981.
Liu, T.-A., Bhuiyan, S., Liu, M.-Y., Sugahara, T., Sakakibara, Y., Suiko, M., Yasuda, S., Production of clones of homozygous diploid zebra fish (Brachydanio rerio). Nature
Kakuta, Y., Kimura, M., Williams, F.E., Liu, M.-C., 2010. Zebrafish as a model for the 291, 293–296. https://doi.org/10.1016/0044-8486(93)90155-R.
study of the phase II cytosolic sulfotransferases. Curr. Drug Metab. 11, 538–546. Tao, T., Peng, J., 2009. Liver development in zebrafish (Danio rerio). J. Genet. Genom. 36,
https://doi.org/10.2174/138920010791636158. 325–334. https://doi.org/10.1016/S1673-8527(08)60121-6.
Lootens, L., Meuleman, P., Pozo, O.J., Van Eenoo, P., Leroux-Roels, G., Delbeke, F.T., Thompson, E.D., Burwinkel, K.E., Chava, A.K., Notch, E.G., Mayer, G.D., 2010. Activity of
2009. uPA+/+-SCID mouse with humanized liver as a model for in vivo metabolism Phase I and Phase II enzymes of the benzo[a]pyrene transformation pathway in
of exogenous steroids: methandienone as a case study. Clin. Chem. 55, 1783–1793. zebrafish (Danio rerio) following waterborne exposure to arsenite. Comp. Biochem.
https://doi.org/10.1373/clinchem.2008.119396. Physiol. C Toxicol. Pharmacol. 152, 371–378. https://doi.org/10.1016/j.cbpc.2010.
MacRae, C.A., Peterson, R.T., 2015. Zebrafish as tools for drug discovery. Nat. Rev. Drug 06.004.
Discov. 14, 721–731. https://doi.org/10.1038/nrd4627. Troxel, C.M., Reddy, A.P., O'Neal, P.E., Hendricks, J.D., Bailey, G.S., 1997. In vivo afla-
Mandrell, D., Truong, L., Jephson, C., Sarker, M.R., Moore, A., Lang, C., Simonich, M.T., toxin B1 metabolism and hepatic DNA adduction in zebrafish (Danio rerio). Toxicol.
Tanguay, R.L., 2012. Automated zebrafish chorion removal and single embryo pla- Appl. Pharmacol. 78, 213–220. https://doi.org/10.1006/taap.1996.8058.
cement: optimizing throughput of zebrafish developmental toxicity screens. J Lab Tseng, H.P., Hseu, T.H., Buhler, D.R., Der Wang, W., Hu, C.H., 2005. Constitutive and
Autom. 17, 66–74. http://dx.doi.org/10.1177/2211068211432197. xenobiotics-induced expression of a novel CYP3A gene from zebrafish larva. Toxicol.
Menke, A.L., Spitsbergen, J.M., Wolterbeek, A.P.M., Woutersen, R.A., 2011. Normal Appl. Pharmacol. 205, 247–258. https://doi.org/10.1016/j.taap.2004.10.019.
anatomy and histology of the adult zebrafish. Toxicol. Pathol. 39, 759–775. https:// Vallverdú-Queralt, A., Boix, N., Piqué, E., Gómez-Catalan, J., Medina-Remon, A., Sasot,
doi.org/10.1177/0192623311409597. G., Mercader-Martí, M., Llobet, J.M., Lamuela-Raventos, R.M., 2015. Identification of
Modick, H., Weiss, T., Dierkes, G., Koslitz, S., Käfferlein, H.U., Brüning, T., Koch, H.M., phenolic compounds in red wine extract samples and zebrafish embryos by HPLC-ESI-
2016. Human metabolism and excretion kinetics of aniline after a single oral dose. LTQ-Orbitrap-MS. Food Chem. 181, 146–151. https://doi.org/10.1016/j.foodchem.
Arch. Toxicol. 90, 1325–1333. https://doi.org/10.1007/s00204-015-1566-x. 2015.02.098.
Mohammed, Y.I., Kurogi, K., Al Shaban, A., Xu, Z., Liu, M.Y., Williams, F.E., Sakakibara, Wang, G., Chen, H., Du, Z., Li, J., Wang, Z., Gao, S., 2017a. In vivo metabolism of or-
Y., Suiko, M., Bhuiyan, S., Liu, M.C., 2012. Identification and characterization of ganophosphate flame retardants and distribution of their main metabolites in adult
zebrafish SULT1 ST9, SULT3 ST4, and SULT3 ST5. Aquat. Toxicol. 112–113, 11–18. zebrafish. Sci. Total Environ. 590–591, 50–59. https://doi.org/10.1016/j.scitotenv.
https://doi.org/10.1016/j.aquatox.2012.01.015. 2017.03.038.
North, T.E., Goessling, W., Walkley, C.R., Lengerke, C., Kopani, K.R., Lord, A.M., Weber, Wang, C., Yin, Y.-H., Wei, Y.-J., Shi, Z.-Q., Liu, J.-Q., Liu, L.-F., Xin, G.-Z., 2017b. Rapid
G.J., Bowman, T.V., Jang, I., Grosser, T., Fitzgerald, G.A., Daley, G.Q., Orkin, S.H., identification of herbal compounds derived metabolites using zebrafish larvae as the
Zon, L.I., 2007. Prostaglandin E2 regulates vertebrate haematopoietic stem cell biotransformation system. J. Chromatogr. A 1515, 100–108. https://doi.org/10.
homeostasis. Nature 447 (7147), 1007–1011. http://dx.doi.org/10.1038/ 1016/j.chroma.2017.07.076.
nature05883. Weigt, S., Huebler, N., Strecker, R., Braunbeck, T., Broschard, T.H., 2011. Zebrafish
Núñez, O., Gallart-Ayala, H., Martins, C.P.B., Lucci, P., Busquets, R., 2013. State-of-the-art (Danio rerio) embryos as a model for testing proteratogens. Toxicology 281, 25–36.

45
C. de Souza Anselmo et al. Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46

https://doi.org/10.1016/j.tox.2011.01.004. Zhang, Q., Ji, C., Yan, L., Lu, M., Lu, C., Zhao, M., 2016. The identification of the me-
Wen, Q., Liu, H. ling, Zhu, Y. ting, Zheng, X. mei, Su, G. yong, Zhang, X. wei, Yu, H. xia, tabolites of chlorothalonil in zebrafish (Danio rerio) and their embryo toxicity and
Giesy, J.P., Lam, M.H.W., 2015. Maternal transfer, distribution, and metabolism of endocrine effects at environmentally relevant levels. Environ. Pollut. 218, 8–15.
BDE-47 and its related hydroxylated, methoxylated analogs in zebrafish (Danio rerio). https://doi.org/10.1016/j.envpol.2016.08.026.
Chemosphere 120, 31–36. https://doi.org/10.1016/j.chemosphere.2014.05.050. Zhu, M., Zhang, H., Humphreys, W.G., 2011. Drug metabolite profiling and identification
Wiegand, C., Krause, E., Steinberg, C., Pflugmacher, S., 2001. Toxicokinetics of atrazine in by high-resolution mass spectrometry. J. Biol. Chem. 286, 25419–25425. https://doi.
embryos of the zebrafish (Danio rerio). Ecotoxicol. Environ. Saf. 49, 199–205. https:// org/10.1074/jbc.R110.200055.
doi.org/10.1006/eesa.2001.2073. Zok, S., Görge, G., Kalsch, W., Nagel, R., 1991. Bioconcentration, metabolism and toxicity
Xu, C., Li, C.Y.-T., Kong, A.-N.T., 2005. Induction of phase I, II and III drug metabolism/ of substituted anilines in the zebrafish (Brachydanio rerio). Sci. Total Environ. 109,
transport by xenobiotics. Arch. Pharm. Res. 28, 249–268. https://doi.org/10.1007/ 411–421.
BF02977789.

46

You might also like