Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Environ Sci Pollut Res (2015) 22:16262–16276

DOI 10.1007/s11356-014-3466-7

DANIO RERIO AS A MODEL IN AQUATIC TOXICOLOGY AND SEDIMENT RESEARCH

Zebrafish as a model to study the role of DNA methylation


in environmental toxicology
Jorke H. Kamstra & Peter Aleström & Jan M. Kooter &
Juliette Legler

Received: 28 May 2014 / Accepted: 14 August 2014 / Published online: 31 August 2014
# Springer-Verlag Berlin Heidelberg 2014

Abstract Environmental epigenetics is a rapidly growing identified so far in mammals are present in zebra fish, as well
field which studies the effects of environmental factors such as a number of major demethylation pathways. However,
as nutrition, stress, and exposure to compounds on epigenetic zebra fish appear to lack some methylation pathways present
gene regulation. Recent studies have shown that exposure to in mammals, such as parental imprinting. Several studies
toxicants in vertebrates is associated with changes in DNA report effects on DNA methylation in zebra fish following
methylation, a major epigenetic mechanism affecting gene exposure to environmental contaminants, such as arsenic,
transcription. Zebra fish, a well-known model in toxicology benzo[a]pyrene, and tris(1,3-dichloro-2-propyl)phosphate.
and developmental biology, are emerging as a model species Though more research is needed to examine heritable effects
in environmental epigenetics despite their evolutionary dis- of contaminant exposure on DNA methylation, recent data
tance to rodents and humans. In this review, recent insights in suggests the usefulness of the zebra fish as a model in envi-
DNA methylation during zebra fish development are ronmental epigenetics.
discussed and compared to mammalian models in order to
evaluate zebra fish as a model to study the role of DNA Keywords Environmental epigenetics . Zebra fish .
methylation in environmental toxicology. Differences exist 5-Methylcytosine . 5-Hydroxymethylcytosine .
in DNA methylation reprogramming during early develop- Environmental toxicology . Development . Transgenerational
ment, whereas in later developmental stages, tissue distribu- effects
tion of both 5-methylcytosine and 5-hydroxymethylcytosine
seems more conserved between species, as well as basic DNA
(de)methylation mechanisms. All DNA methyl transferases Introduction

As shown by a multitude of studies over the past decades,


Responsible editor: Philippe Garrigues
epigenetic modifications alter the expression of underlying
J. H. Kamstra (*) : J. Legler genes and are critically important in the development and
Institute for Environmental Studies, VU University Amsterdam,
homeostasis during life (Bird 2007). Epigenetic marks include
1081 HV Amsterdam, The Netherlands
e-mail: jorke.kamstra@vu.nl covalent DNA base (cytosine methylation), histone modifica-
J. Legler
tions (e.g., lysine and arginine methylation, lysine acetylation
e-mail: juliette.legler@vu.nl and ubiquitination, serine phosphorylation), noncoding
RNAs, and chromatin structure organization of the DNA itself
P. Aleström and are embodied in the field of epigenetics (Bird 2007).
Faculty of Veterinary Medicine and Biosciences, Dept. of Basic
Importantly, DNA and histone modifications can be inherited
Science and Aquatic Medicine, Norwegian University of Life
Sciences, 0033 Oslo, Norway during cell division and to some extent over generations
e-mail: peter.alestrom@nmbu.no (Probst et al. 2009). Epigenetic modifications are known
to be highly dynamic and are affected by external stimuli,
J. M. Kooter
such as changes in internal and external environment,
Department of Molecular Cell Biology, Section Genetics, VU
University Amsterdam, 1081 HV Amsterdam, The Netherlands nutrition, and exposure to compounds. The effects of these
e-mail: j.m.kooter@vu.nl stimuli on epigenetic gene regulation are described as
Environ Sci Pollut Res (2015) 22:16262–16276 16263

environmental epigenetics (Jirtle and Skinner 2007). These The advantages of using zebra fish in epigenetic research
changes may lead to a variety of either beneficial or adverse are numerous. For instance, due to techniques like zinc finger
phenotypical outcomes, as well as disease-related effects, and nucleases and the more recently developed transcription
in some cases even over generations (Fukata and Mori 2004; activator-like effector nucleases (TALENs) (Cermak et al.
Bollati and Baccarelli 2010; Baylin and Jones 2011; Wang 2011) and clustered, regularly interspaced, short palindromic
et al. 2012). Transgenerational effects have been linked to repeats (CRISPR/Cas9) (Hwang et al. 2013; Wang et al.
aberrant DNA methylation patterns, emphasizing the impor- 2013), the design and generation of mutants of proteins in-
tance of this epigenetic mark (Skinner 2013; Vandegehuchte volved in epigenetic processes have become possible to
and Janssen 2014). achieve for many laboratories. Moreover, since many genes
So far, research in DNA methylation has focused pre- involved in epigenetic processes in the early embryo are
dominantly on mice and human, especially in the fields of maternally transferred, prolonged survival is possible in these
medicine and environmental epigenetics (Skinner 2013). mutants during the first stages of development. Furthermore,
Many studies report aberrant DNA methylation levels and if needed mRNAs from these maternal genes can be silenced
patterns in virtually all cancers (Gokul and Khosla 2013). by antisense morpholinos (MOs) (Landsverk et al. 2010;
Due to drawbacks in using rodent models for epigenetic Pikulkaew et al. 2011). Recent research in zebra fish DNA
research such as economical and ethical considerations, methylation has focused mainly on processes during early
DNA methylation studies in other species, including zebra zebra fish development and the basic enzyme systems in-
fish, are emerging rapidly in the field of epigenetics volved in DNA methylation (e.g., methyl transferases, de-
(Williams et al. 2014). Despite the evolutionary distance methylation mechanisms, methylcytosine-binding proteins).
to mammals, zebra fish share 70 % of their genes with Since DNA methylation patterns during embryonic develop-
human and have proven to be a suitable model organism in ment as well as the (de) methylation pathways in mammals are
many studies, including epigenetics. The transparent na- extensively studied (Law and Jacobsen 2010; Smallwood and
ture of the early embryo, its well-characterized develop- Kelsey 2012; Wu and Zhang 2014), comparisons with recent
mental stages, its rapid development, and the ease of cul- findings in the zebra fish are timely. The purpose of this
ture make this species an extremely useful experimental review is to describe and compare features of DNA methyla-
model (Strähle et al. 2012). tion in the zebra fish with those of mammals in order to
During zebra fish embryogenesis, several developmental evaluate their applicability as a model to study the effects of
stages can be distinguished (Kimmel et al. 1995), and dur- environmental toxicants in humans.
ing development, changes in the epigenetic landscape are
highly dynamic (Mhanni and McGowan 2004; MacKay
et al. 2007; Lindeman et al. 2010b; Andersen et al. 2012a, DNA methylation
b; Jiang et al. 2013; Potok et al. 2013), leading to tightly
controlled gene expression (Aanes et al. 2011). During the DNA methylation, a covalent modification of a methyl group
zygotic stage (0.25 h postfertilization (hpf)), maternal on the 5 position of cytosine (mC), is an epigenetic mark
mRNAs are present and determine the fate of cells during involved in gene regulation and genome maintenance. In
the first hours of embryonic development. Many maternal vertebrates, DNA methylation is predominantly located at a
mRNAs encode for chromatin remodeling factors during cytosine-guanine dinucleotide (CpG). Non-CpG methylation
this stage, and no apparent zygotic transcription is observed has also been found, for example, embryonic stem cells, but
(Aanes et al. 2011). This period of embryonic cell divisions the levels are much lower, and the significance of this mark is
in the absence of de novo transcription is relatively long, ten as yet unknown (Lister et al. 2009). Although silencing of
cell cycles, or ∼3 h, after which zygotic genome activation genes is an important feature of promoter methylation, as
(ZGA) takes place during the midblastula transition (MBT, shown by the inactive X chromosomes and transposons
3.3 hpf, 1,000 cells) (Aanes et al. 2011). The majority of and imprinted and tissue-specific genes, recent studies
maternal mRNAs are degraded by the aid of zygotic indicate that DNA methylation is more dynamic and
miRNAs (miR-430) during this stage (Schier 2007). At plays a more complex role in gene regulation (reviewed
gastrula, when the germline is formed (50 % epiboly, by Jones 2012).
5.3 hpf), genes involved in cell cycle, cell regulation, and Until recently, distinct differences in CpG islands (CGIs)
early development are expressed (Mathavan et al. 2005; occurrence between mammals and cold blooded vertebrates
Aanes et al. 2011), and the epigenetic landscape will be- were observed. A CGI is commonly defined as a DNA region
come more complex due to the formation of differentiated of more than 200 bp long with a relatively high guanosine and
cells (Andersen et al. 2012a). During segmentation and later cytosine (GC) content in a CpG dense region. In mammals,
stages, genes involved in organogenesis are expressed CGIs are often found at transcription start sites (TSSs) of
(Mathavan et al. 2005). genes (>70 %) and remain mostly unmethylated and are
16264 Environ Sci Pollut Res (2015) 22:16262–16276

important for gene regulation (Deaton and Bird 2011). CGIs DNA methyltransferases in zebra fish
are identified using various algorithms; however, a definite
definition of a CGI has not yet been reported (Gardiner- DNA methyltransferases (DNMTs) methylate cytosines at the
Garden and Frommer 1987; Takai and Jones 2002; Wu et al. C5 position with S-adenosyl methionine (SAM) as a methyl
2010). In cold blooded vertebrates, these algorithms generally donor (Hermann et al. 2004). The DNMT family consists of
calculate a much lower CGI occupancy at TSSs compared to different members which can be roughly divided into mainte-
mammals, indicating an evolutionary difference between nance and de novo methyltransferases. In mammals, five
mammals and cold blooded vertebrates in CGI occurrence different isoforms of DNMTs are known: DNMT1,
and perhaps in function (Long et al. 2013). For example, using DNMT2, DNMT3a, DNMT3b, and DNMT3L (Table 1).
the algorithm of Gardiner-Garden and Frommer (1987) within DNMT1 is involved in maintenance methylation, whereas
the UCSC genome browser, Potok and colleagues (2013) DNMT3a and DNMT3b are involved in de novo methylation.
found a total amount of 12,683 CGIs in zebra fish, a lower DNMT3L lacks the catalytic subunit and has a supporting role
number than in mouse and human (16,026 and 28,691, re- in de novo methylation (Hermann et al. 2004). DNMT2,
spectively) (Table 2). Zebra fish CGIs were located at TSSs, which harbors only the catalytic domain of a DNA methyl-
repeat sequences, and gene bodies of metabolic and develop- transferase, is thought to be involved in RNA cytosine meth-
mental genes, but only 16 % actually spanned TSSs compared ylation (Smith et al. 2011). Zebra fish have eight different
to >50 % in mouse and humans (Potok et al. 2013). Therefore, dnmt genes, of which six are most likely de novo Dnmts as
these algorithms need to be adapted to the specific genomic they are most similar to the mammalian DNMT3 members. It
features of zebra fish. For example, Andersen and coworkers appears that zebra fish lack a Dnmt3L protein, since it has not
(2012b) used the Takai and Jones algorithm with modified been located in the zebra fish genome up to this date (Smith
parameters for observed to expected (o/e) ratio of CpG sites, et al. 2011) (Table 1). All zebra fish Dnmts are maternally
i.e., the observed number of CpG sites versus theoretical transferred to the zygote, and mRNA levels decline during the
number of CpG sites in a given sequence. By determining first stages of development and increase again after ZGA
the percent GC and CpG content at regions 1 kb upstream of (Martin et al. 1999; Smith et al. 2011). Gene expression of
the transcription start sites of zebra fish genes, they the Dnmt3b orthologs (dnmt3, dnmt4, and dnmt7) increases
could divide the promoters analyzed into 65 % high up to 6 hpf and gradually declines again towards undetectable
and 35 % low CpG promoters, suggesting that this levels at 72 hpf, whereas expression of DNMT3a orthologs
approach is more conserved between vertebrate taxa (dnmt6 and dnmt8) steadily increases after 6 hpf. All DNMT3
and comparable to the >70 % CGIs found in mouse orthologs are also detectable in muscle and brain tissue, but
and human promoters using the Gardiner-Garden and are consistently higher in brain (Smith et al. 2011).
Frommer algorithm. DNMTs are evolutionarily conserved proteins, as shown
Another abundant modification of cytosine is 5- by the overall sequence similarity of DNMTs from different
hydroxymethylcytosine (hmC). In mammals, this cytosine species (Mhanni et al. 2001) and the observation that ectopic
modification has been observed in the early embryo inner cell expression of the human DNMT1 and DNMT3b in zebra fish
mass and embryonic stem cells, and high levels have been can take over the function of the zebra fish orthologs (Rai et al.
found in brain neurons and bone marrow (Pfeifer et al. 2013; 2006, 2010). Additional evidence of evolutionary conserva-
Wu a n d Z h a n g 2 0 1 4 ) . T h e e x a c t f u n c t i o n s o f tion is found in the interactions of DNMTs with other proteins.
hydroxymethylation are not known up to this date, but one In mice, UHRF1 (DNMT1-recruiting proteins, ubiquitin-like,
is an intermediate step during DNA demethylation, together
with the less abundant 5-formylcytosine (fC) and 5- Table 1 DNMTs isoforms in mice and zebra fish
carboxylcytosine (cC) (Wu and Zhang 2010). Since many
Mice Zebra fish
tissues have a higher hmC content than expected, it might
serve as an epigenetic mark during stem cell and neuronal Maintenance methylation DNMT1 Dnmt1
development (Pfeifer et al. 2013). hmC is enriched in genic RNA methylation DNMT2 Dnmt2
regions and is present in gene bodies of transcribed genes, De novo methylation DNMT3a Dnmt6
indicating its specific functional role (Nestor et al. 2012). Dnmt8
Also, studies hint to the existence of specific hmC interacting De novo methylation DNMT3b Dnmt3
proteins, which are known to be involved in epigenetic pro- Dnmt4
cesses (Yildirim et al. 2011; Mellén et al. 2012; Wu and Zhang Dnmt5
2014). Despite the increasing knowledge about DNA Dnmt7
(hydroxy)methylation, still many aspects of these epigenetic Cofactor DNMT3L N/A
marks remain unanswered (Vastenhouw and Schier 2012;
Jones 2012; Wu and Zhang 2014). From Shimoda et al. (2005) and Law and Jacobsen (2010)
Environ Sci Pollut Res (2015) 22:16262–16276 16265

containing PHD and RING finger domains) and proliferating reported in mammals (Law and Jacobsen 2010). The first
cell nuclear antigen (PCNA) recruit DNMT1 to evidence of similar interactions in zebra fish was obtained
hemimethylated CpG sites after DNA replication (Law and with Dnmt3, an ortholog of DNMT3B, which was shown to
Jacobsen 2010). Similarly, in zebra fish, interaction of Dnmt1 interact with zebra fish G9a (Rai et al. 2010). G9a
with Uhrf1 has been observed (Tittle et al. 2011). Both Uhrf1 morpholinos exhibited similar deficiencies in brain develop-
and Dnmt1 mutants show similar phenotypical defects in ment as with Dnmt3 knockouts. Interestingly, phenotypes
zebra fish lens development along with a lower global mC could be rescued by overexpression of both zebra fish and
content, indicating that Uhrf1 is essential for maintenance human G9a, thereby supporting the evolutionary conservation
methylation, though a direct interaction between Uhrf1 and of this pathway (Rai et al. 2010). In the same study, it was
Dnmt1 was not reported (Tittle et al. 2011). Further evidence shown that both Dnmt1 and Dnmt3 are essential for retinal
of Uhrf1 conservation was found in a genome-wide methyla- development, with Dnmt3 being vital for early stages of
tion study where both zebra fish and mice Uhrf1 mutants differentiation and Dnmt1 for terminal differentiation (Rai
showed similar demethylation patterns (Feng et al. 2010). et al. 2010). Recently, expression of all de novo Dnmts in
Interaction of Dnmt1 with H3K9 methyltransferase suppres- the developing eye of zebra fish has been observed, with
sor of variegation 3-9 homologue 1 (Suv39h1) and euchro- specific functions of different Dnmts in different regions.
matic histone-lysine N-methyltransferase 2 (Ehmt2 or G9a) However, there is an overlap in expression of the various
has also been observed in zebra fish (Rai et al. 2006, 2010). Dnmts (Seritrakul and Gross 2014).
Interestingly, similar interactions between DNMTs and these In summary, these data show that similar interactions be-
proteins have also been found in mammalian in vitro and tween DNA methyltransferases and histone methyltransfer-
in vivo studies (Rai et al. 2006). ases occur between species, indicating strong conservation of
In zebra fish, Dnmt3 to Dnmt8 are the main contributors to these epigenetic pathways. However, some interactions ob-
de novo methylation, similar to DNMT3A and DNMT3B in served in mice, such as Suv39h1-DNMT3b, have not been
mammals (Table 1). In mammals, a DNMT3A/DNMT3L observed in zebra fish up to this date.
tetramer is thought to be involved in methylating imprinted
genes (Yokomine et al. 2006; Law and Jacobsen 2010). Since
no DNMT3L ortholog seems to be present in zebra fish, it is Demethylation pathways in zebra fish
unlikely that zebra fish harbor a similar imprinting mechanism
(Wu et al. 2011). The fact that parthogenesis and gynogenesis In mammals, demethylation of cytosines occurs both passive-
result in the normal development of zebra fish demonstrates a ly and actively during development and life. Passive demeth-
fundamental difference between fish and mammals in this ylation occurs during DNA replication and cell division when
mechanism of epigenetic control of development (Corley- hemimethylated DNA is not methylated by the maintenance
Smith et al. 1996). In mice, the DNMT3A/DNMT3L tetramer DNMT1 (Wu and Zhang 2010). Passive demethylation ap-
is also targeted towards transposable elements by Miwi2 pears to be the mechanism for the global erasure in both
proteins loaded with piRNAs, in order to silence transposable primordial germ cells (PGCs) and in the zygote, where the
elements through CpG methylation (Law and Jacobsen 2010). paternal methylome is first oxidized by ten-eleven transloca-
This piRNA pathway has also been demonstrated in zebra fish tion (TET) enzymes (Smallwood and Kelsey 2012; Wu and
(Houwing et al. 2007; Wei et al. 2012), but no interaction with Zhang 2014). Active demethylation generally follows two
Dnmts or DNA methylation has been reported yet, presum- pathways in mammals. One involves deamination of methyl-
ably due to the nonexistence of the DNMT3L ortholog. The ated cytosines by activation-induced cytosine deaminases
reason why zebra fish contains more de novo Dnmts than (AIDs) or apolipoprotein B mRNA-editing enzyme
mammals is currently not clear; possibly, these enzymes har- (APOBEC), creating a GT mismatch which is removed via
bor tissue or promoter- or gene-specific functions. For exam- base excision repair (BER) using thymine DNA glycolase
ple, tissue-specific expression has been observed for Dnmt4, (TDG) and methyl-binding domain protein 4 (MBD4) (Law
Dnmt6, and Dnmt8 in zebra fish embryos (Takayama et al. and Jacobsen 2010; Wu and Zhang 2014). A second pathway
2014). Also, it has been demonstrated that of all five de novo involves the formation of a hmC intermediate by TET en-
Dnmt homologs, only Dnmt7 morpholinos showed effects on zymes, followed by two possible routes: (1) deamination and
promoter methylation of no tail, a gene essential for notochord BER via the previously mentioned pathway or (2) further
and tail development, whereas no effects on global methyla- oxidation of hmC to cC, which is removed by a yet to be
tion levels were observed, indicating locus-specific recruit- discovered carboxylase or via BER (Pastor et al. 2013; Wu
ment of this Dnmt (Yamakoshi and Shimoda 2003). and Zhang 2014). In zebra fish, both enzyme families are
Interactions of DNMT3B with several histone methyltrans- present. Aid/Apobec deamination has been more thoroughly
ferases (G9a, SUV39h1, enhancer of zeste homologue 2 studied, and evidence of Tet involvement in zebra fish de-
(EHZ2), SET domain bifurcated 1 (SETDB1)) have been methylation mechanisms is emerging.
16266 Environ Sci Pollut Res (2015) 22:16262–16276

Demethylation via deamination by Aid/Apobec the zygotic stage, which to our knowledge has not yet been
performed, could give additional proof that there is no
Until recently, the role of the AID/Apobec-mediated mC reprogramming via hmC. At later embryonic stages, hmC
demethylation pathway in zebra fish was controversial. has been found with immunostaining in the tail section
Although mismatch repair of GT by TDG and BER has been (Jiang et al. 2013) and skeletal muscle, retina, and brain
shown in several studies, human recombinant AID and regions (Almeida et al. 2012), a similar tissue distribution as
APOBEC deaminate cytosines poorly and have an even lower observed in mammals (Globisch et al. 2010; Nestor et al.
activity towards mC (Pastor et al. 2013). However, purified 2012), however, no data is available on the quantitative mea-
zebra fish Aid has a high deamination activity on both C and sures in various tissues of zebra fish. Where present, hmC is
mC, suggesting that demethylation via Aid/Apobec is a plau- accompanied by mRNA increases of all three tet genes
sible pathway in zebra fish (Abdouni et al. 2013). The func- (Almeida et al. 2012; Ge et al. 2014). In zebra fish, a specific
tionality of this cytosine modification mechanism is supported role of Tet2 has been discovered in erythropoiesis, accompa-
by experiments whereby the demethylation of methylated nied by lower promoter methylation of genes involved in
DNA fragments introduced in embryos was monitored (Rai erythropoiesis (Ge et al. 2014). These are the first indications
et al. 2008). Gradually, up to 13 hpf, the DNA fragments were of similar demethylation mechanisms at later developmental
demethylated and remethylated at 28 hpf. At 13 hpf, mRNA stages in mammals and zebra fish.
levels of aid, apobec2a, and apobec2b were increased.
Repression of aid, apobec2a, and apobec2b expression via
MOs showed reduction in demethylation, but only when all Methylation during different developmental stages
three genes were repressed (Rai et al. 2008). Interestingly, of zebra fish
overexpression of Aid/Apobec did not increase demethyla-
tion. Only when hMBD4 was coinjected, demethylation was The studies presented in this review use many techniques for
observed of both inserted methylated DNA fragments and the either fundamental research on DNA methylation or effects on
genomic DNA itself. Catalytically inactive (for thymine DNA methylation following exposure to compounds. A vari-
deglycosylase) hMBD4 coinjected with Aid revealed GT ety of analyses can be used for specific, global, and genome-
mismatches, suggesting active deamination of methylated wide DNA methylation analysis, to assess differences in DNA
CpG sites (Rai et al. 2008). Although AID is able to deaminate methylation during development or following exposures to
both C and mC, the involvement of a methyl domain-binding environmental toxicants. Table 3 presents an overview of some
protein in cooperation with deaminases could provide the of the major techniques applied in the studies described below.
specificity towards mC. The overall guanosine and cytosine (GC) content in the
zebra fish genome is slightly lower compared to mammals
(36.8 versus 42.1, respectively), whereas the o/e is higher
Involvement of TET enzymes in DNA demethylation (0.53 in zebra fish versus 0.17 and 0.20 in mouse and human,
respectively) (Table 2). Methylation levels of CpGs in zebra
Oxidation of mC to hmC by TET enzymes and subsequent fish are generally higher (60–90 % for human/mouse and 70–
removal of the modified base is a second route towards 95 % for zebra fish) (Table 2) (Potok et al. 2013), with the
demethylation. In mice, the hmC intermediate has been found highest levels observed in sperm (91–95 %), whereas oocytes
in the zygote on the paternal genome prior to erasure of the exhibit lower levels (75–80 %) (Jiang et al. 2013; Potok et al.
methylation marks (Ruzov et al. 2011); however, subsequent 2013). Oocytes of mice also have a lower methylation level
demethylation is believed to occur passively (Wu and Zhang than sperm (approx. 40 versus 82 %, respectively), although
2014). Furthermore, hmC has been found in several tissues, the overall methylation levels are generally lower in both germ
including brain, embryonic stem cells, and PGCs (Globisch cell types compared to zebra fish (Kobayashi et al. 2012;
et al. 2010; Nestor et al. 2012). Surprisingly and in contrast to Smith et al. 2012).
mammals, no detectable levels of hmC are present in embryos During the development of the zebra fish embryo, methyl-
up to the tenth somite stage (14 hpf) (Almeida et al. 2012). ation of gene promoters is highly dynamic and varies between
These observations indicate that demethylation of the paternal gametes and developmental stage (Fig. 1). In both zebra fish
genome does not involve hmC. However, low levels of hmC sperm and oocytes, many promoter regions of genes involved
are detectable at the 64-cell stage with a more sensitive hmC in early development and metabolism are hypomethylated and
pull-down method at specific differentially methylated regions remain hypomethylated throughout early development up to
(DMRs) (e.g., hoxc and dnmt3) (Jiang et al. 2013; Potok et al. the sphere stage (Potok et al. 2013). Hypermethylated promot-
2013). However, loci with hmC were rare and it is unlikely er regions in oocytes and sperm are generally found in genes
that this mark is an intermediate in the global erasure of important for later developmental stages, such as neurogenesis.
methylation, as observed in mammals. Analysis of hmC at Interestingly, promoter regions of germ-specific genes (piwi,
Environ Sci Pollut Res (2015) 22:16262–16276 16267

Table 2 Genomic features of human, mice, and zebra fish analysis, since a reduction of methylation by half was ob-
Human Mice Zebra fish served at every cell replication, whereas de novo methylation
of oocyte-specific hypomethylated loci has been observed in
Number of chromosomes 23 20 25 zebra fish with a SNP in the maternal DNA (Jiang et al. 2013).
Total genome (bp) 3.1×109a 2.7×109a 1.4×109a These data suggest that passive demethylation and de novo
Protein coding genes 20,770b 23,139b 26,241b methylation occur simultaneously and that the maternal
CpG sites 2.82×107a 2.13×107a 2.53×107a methylome is reprogrammed to that of the paternal genome.
%CG 42.5a 42.2a 36.8a Though the global erasure of methylation prior to de novo
CGI 28,691a 16,026a 12,683a methylation observed in mammals during early embryogene-
%mdC in CpG context 60–90 %c 60–90 %c 70–95a sis (Smallwood and Kelsey 2012) has not been observed in
%mC of total C ∼1–2 %d ∼3–4 %e ∼8–9 %f zebra fish, there are some features that resemble mammals. In
o/e CpG 0.20a 0.17a 0.53a mice, Smith and coworkers (2012) observed that, despite the
Imprinting Yes Yes No global erasure of DNA, oocyte-specific hypermethylated
(compared to sperm) sites were hypomethylated at E7.5 and
a
Potok et al. (2013) oocyte-specific hypomethylated sites were hypermethylated
b
Ensembl (2013) at E7.5. In other words, the methylome of oocyte-specific
c
Tucker (2001) DMRs is reprogrammed to that of sperm, in both mice and
d
Li and Liu (2011) zebra fish. Furthermore, in both species, the number of
e
Globisch et al. (2010) hypermethylated DMRs in sperm was higher than in oocytes.
f
Rai et al. (2008) Also, the methylome of sperm resembles that of somatic
tissue, and regardless of global erasure of DNA methylation
dnmt6, and vasa) are hypomethylated in sperm and during embryogenesis, the methylome of oocytes is
hypermethylated in oocytes, even though these genes are reprogrammed towards the paternal (or somatic) state, which
known to be maternally transferred (Potok et al. 2013). The would imply that this reprogramming mechanism is rather
developmental genes that are only hypomethylated in sperm conserved between species.
include all the 48 hox genes (Wu et al. 2011; Jiang et al. 2013; As the zebra fish embryo passes through the different
Potok et al. 2013). In oocytes, it appears that in zebra fish, but stages during development, cells differentiate and migrate to
not in mice, the hox gene promoters are hypermethylated specific regions, and organogenesis starts. At this point of
(Potok et al. 2013). Since many genes involved in early devel- development, the epigenetic complexity of the embryo in-
opment were already hypomethylated in sperm, the creases and the embryo cannot be seen as one homogenous
methylome of zebra fish sperm can already be regarded as cell mass, and therefore, specific tissues, rather than whole
programmed for embryonic development. embryos should be compared for methylation status. A num-
During the one- to two-cell stage, a hypomethylated state ber of promoter regions of genes have been shown to be
of the zebra fish genome has been observed in one study, with differentially methylated during myogenesis (Potok et al.
lower methylation levels compared to sperm and oocytes and 2013). For example, promoters of genes encoding for tran-
increasing methylation levels up to 4 hpf (Mhanni and scription factors are hypermethylated at the sphere stage (plu-
McGowan 2004) (Fig. 1). The hypomethylated state at the ripotent state) compared to adult skeletal muscle tissue (ter-
one- to two-cell stage, however, has not been confirmed in minally differentiated), along with genes involved in metabo-
later studies, and no additional information is available on the lism and germ line maintenance (hox, vasa, piwil1, and dazl)
actual methylation status during the formation of the zygote. (Potok et al. 2013). When comparing DMRs (∼3,000) of
Two recent studies by Potok et al. (2013) and Jiang et al. sphere and muscle tissue, ∼2,000 became hypermethylated
(2013) examined DNA methylation at later stages (mixture of in muscle, predominantly in the gene bodies, while ∼1,000
2–16- and 16-cell stage, respectively), using whole-genome became unmethylated (Potok et al. 2013). The function of
shotgun bisulfite sequencing and confirmed the increase in gene body methylation is basically unknown; it occurs mainly
methylation up to the sphere stage. in introns and possibly acts on enhancers (Potok et al. 2013)
More specific analysis of methylation during early devel- and/or is a mechanism for silencing of repeat elements and is
opment has revealed some striking features. For example, it also positively correlated with gene expression (Jones 2012).
has been observed that oocyte-specific hypomethylated re- Additionally, the entire hoxc gene cluster is hypomethylated in
gions are de novo methylated to a level that is similar to the sperm, whereas most of the genes in this cluster are
hypermethylated state of sperm, whereas hypermethylated hypermethylated in muscle tissue (Wu et al. 2011). During
regions in the oocyte are passively demethylated (Potok myogenesis, some muscle-specific genes are hypomethylated
et al. 2013). Also, passive demethylation at oocyte-specific (musk, dystrophin) whereas other genes involved in muscle
hypermethylated loci has been shown by bisulfite sequencing development (myocyte enhancer factors, e.g., pbx1, myod1)
16268

Table 3 Methods used to assess DNA methylation in zebra fish

Method Principle Advantage Disadvantage Study

Global analysis
Methylation-sensitive Methylation-sensitive restriction enzymes Fast, cost effective Not quantitative, no information on Martin et al. (1999), Macleod et al. (1999),
restriction digestion (HPAII/MSPI), combined with Southern specific loci Mhanni and McGowan (2004)
blots
Liquid chromatography- Global analysis of fully digested DNA, Quantitative, fast, combined with mass No information on specific loci Rai et al. (2008), Globisch et al. (2010),
based methods quantitative analysis of single nucleosides spectrometry scan for multiple Ge et al. (2014), Olsvik et al. (2014)
cytosine modifications
Immunostaining Use mCpG- or hmCpG-specific antibodies, Tissue-specific distribution in whole Not quantitative, no information on Li et al. (2009), Ceccaldi et al. (2011),
use with immunoblots or whole embryo body specific loci Almeida et al. (2012)
staining
Genome-wide analysis
Whole-genome bisulfite Whole-genome analysis of Complete methylome analyzed Expensive, no discrimination Jiang et al. (2013), Potok et al. (2013)
sequencing (WGBS) bisulfite-converted DNA between mC and hmC
Reduced representation Representation of high CG content loci with Input amount DNA low, relatively Analysis of only 1 % of the genome, Gu et al. (2011), Smith et al. (2012)
bisulfite sequencing methylation-insensitive restriction cheap covering only 7 % promoters in
(RRBS) digestions (MspI) zebra fish
Methyl DNA Immunoprecipitation of mC followed by Only mC is analyzed, also hmC Bias in cross-reactivity antibodies, Andersen et al. (2012b), Olsvik et al. (2014)
immunoprecipitation microarrays or sequencing immunoprecipitation no single CpG resolution with
(MeDIP) arrays
Specific analysis
Bisulfite sequencing Sanger sequencing, pyrosequencing on Quantitative information on specific PCR bias Strömqvist et al. (2010), Fang et al. (2013),
bisulfite-converted DNA loci, at single-CpG sites, multiplex Hernández et al. (2013), Corrales et al.
sequencing (2014a)
Methylation-sensitive High -resolution melting on Quantitative information on specific No information of single CpG sites Kamstra et al. (2014)
high-resolution melting bisulfite-converted DNA loci, high throughput, correction for
(MS-HRM) PCR bias
Environ Sci Pollut Res (2015) 22:16262–16276
Environ Sci Pollut Res (2015) 22:16262–16276 16269

Fig. 1 Dynamic methylation during early zebra fish development. a Red dynamics a specific loci. b Methylation levels of many developmental
indicates maternal; blue paternal; and green, diploid embryonic genome. genes (e.g., vasa, piwil1, dazl, dnmt3) decrease up to the sphere stage and
Maternal genome is reprogrammed during early development, with pas- increase at later developmental stages; however, c some developmental
sive demethylation starting at zygote and de novo methylation from two- genes are already hypomethylated. d Muscle-specific genes, musk and
cell stage. The paternal genome is unaffected. No data on PGC program- dystrophyn, are hypermethylated in sperm and hypomethylated in oocytes
ming is available. The gray box indicates a sensitive window for inter- (not shown), and methylation increases during development and de-
ference by Dnmt inhibitor exposure. b–d Examples of methylation creases in specific tissues at later stages

were already hypomethylated at early developmental stages Histone modifications associated with DNA methylation
(Potok et al. 2013). The hypomethylated status of these genes during development
suggests a permissive state for transcription of the promoter in
terminal differentiation. Modifications of histones alter transcriptional activity of chro-
When comparing MBT embryos with ZF4 fibroblasts, a matin and may influence DNA methylation. Among the many
cell line derived from 24-hpf zebra fish embryos, representing known histone modifications, H3K4me3 is associated with
a more developed tissue, gene promoters with more than one gene expression, while H3K9me3 and H3K27me3 are asso-
CGI are hypermethylated in ZF4 cells. Genes with multiple ciated with gene repression. H3K9me3 and H3K27me3 are
CGIs often encode transcription factors (such as hox genes), often accompanied by DNA methylation and silencing of
and hypermethylation in ZF4 cells suggests a long-term si- specific loci (Ruthenburg et al. 2007; Lindeman et al. 2011).
lenced state of these genes (Andersen et al. 2012b). The bivalent H3K4me3 and H3K27me3 mark is often found
Pluripotency control genes (pou5f1, sox3, klf4, otx1b, and at developmental genes poised for transcriptional
vasa) are hypermethylated in ZF4 cells when compared to (in)activation during embryonic development and in embry-
MBT embryos, in correspondence with their repressed state in onic stem cells (Ruthenburg et al. 2007; Vastenhouw and
the somatic cell type line (Lindeman et al. 2010b). After MBT, Schier 2012).
many developmental gene promoters are hypomethylated and In zebra fish development, before, during, and after ZGA,
have bivalent histone marks, making them poised for tran- hypomethylated DNA regions contain H3K4me3-marked nu-
scription (Andersen et al. 2012b). It is possible that after cleosomes, though no information was reported about the
differentiation, these developmental genes are subsequently actual transcriptional activity of hypomethylated promoters
permanently silenced by H3K9me3 and H3K27me3, which is with H3K4me3 marks (Andersen et al. 2012b). However,
reinforced and stabilized by de novo promoter methylation Potok et al. (2013) did report an association between
(Jones 2012) (see below). hypomethylated promoters, H3K4me3 marks and
16270 Environ Sci Pollut Res (2015) 22:16262–16276

gene expression. At ZGA and later, an increase in H3K9me3


and H3K27me3 was observed, due to a more epigenetic
complex state of determined and differentiated cells in later
development (Lindeman et al. 2010a). At sphere stage (MBT),
a number of developmental genes were marked with a com-
bination of H3K4me3 and H3K27me3 and low promoter
methylation, correlated with low expression levels
(Andersen et al. 2012a; Potok et al. 2013). The relation
between histone marks and DNA methylation argues that
histone modification analysis should be taken into consider-
ation together with DNA methylation analysis when assessing
toxicant-induced effects on the epigenome.

DNA methylation in environmental toxicology

Interference with enzyme systems

Altered global DNA methylation levels are often associated Fig. 2 De(hydroxy)methylation in 48-hpf embryos following continuous
with compounds that interfere with methyltransferases or its exposure of 5-AC from 4–16-cell stage. a Control versus b 5-AC
(75 μM)-exposed zebra fish, showing short tail phenotype and heart
substrate SAM. Among compounds that have been thorough- edema. c mC levels decline with increasing concentrations of 5-AC. A
ly investigated for their hypomethylating effects and are often similar trend is observed with hmC. Significance was determined with
used as model compounds in studies that assess DNA meth- one-way ANOVA, following a Bonferroni’s multiple comparisons test
ylation are the DNMT inhibitors 5-aza-cytidine (5-AC) or 5- (P<0.05) using GraphPad Prism 5.04. Methylation analysis was carried
out only in embryos showing no phenotypical effects
aza-deoxycytidine (5-AdC). These cytosine analogs have the
potency to inhibit DNMTs as they incorporate in DNA and
covalently bind DNMTs, eventually leading to loss of DNMT arsenite exposure in rat liver cells (Zhao et al. 1997) as well as
activity. As a demonstration of this inhibition, levels of both in livers from mice exposed to sodium arsenite via drinking
methylated and hydroxymethylated DNA are decreased by 5- water for 48 weeks (Chen et al. 2004). The hypomethylating
AC exposure in a concentration-dependent manner (Fig. 2) effect of As in liver could be explained by its methylated
(Kamstra et al. 2014, in preparation). In an earlier study with metabolites, as metabolism involves SAM-dependent arsenic
5-AC exposed to zebra fish, severe phenotypical changes and methyltransferases, thereby depleting the methyl pool for
lower overall methylation levels were also observed (Martin DNMTs (Hamdi et al. 2012). In zebra fish exposed to As, an
et al. 1999), where the timing of exposure to 5-AC appears to aberrant DNA methylation pattern has been observed in trunk
be critical. The most pronounced morphological effects were and tail, with hypomethylation after 24 hpf and hypermethy-
found after exposure to 75 μM 5-AC in embryos exposed lation after 48 hpf compared to controls (Li et al. 2009).
from 1 to 24 hpf. Shorter windows of exposure from 1 to 4 hpf However, exposure levels of As in these experiments were
appeared to be critical and resulted in similar results, whereas very high, and it is not clear if these effects were nonspecific
exposure from 6 to 24 hpf showed almost no effects on due to toxicity and apoptosis which was found in the same
morphology (Martin et al. 1999). This sensitive early stage regions (Li et al. 2009).
window corresponds with the methylation dynamics during Changes in global methylation levels have also been ob-
early development and indicates that this time window must served in zebra fish embryos exposed to the polycyclic aro-
be included in the studies with compounds that are susceptible matic hydrocarbon benzo[a]pyrene (B[a]P) (Fang et al. 2013;
to interfere with Dnmt enzymes (Fig. 1). Corrales et al. 2014a) and the organophosphorus flame retar-
Several environmental pollutants have been found to alter dant tris(1,3-dichloro-2-propyl)phosphate (TDCPP) (McGee
global DNA methylation levels in humans and other model et al. 2012) (Table 4). With B[a]P, a known mutagen through
organisms. For example, changes in global methylation have its formation of DNA adducts, decreased methylation levels
been observed in an in vitro mouse adipocyte differentiation could be due to adduct formation which preferentially takes
model following exposure to the organotin tributyltin (TBT) place on a (hemi)methylated CpG consensus, subsequently
and the plasticizer bisphenol A (BPA) (Bastos Sales et al. leading to GC to TA conversion (Weisenberger and Romano
2013). One of the most studied compounds showing effects 1999). Therefore, these effects, albeit compound specific, are
on global DNA methylation are arsenic (As) compounds. likely due to chemical modifications of DNA by B[a]P and
Genome-wide hypomethylation has been found after sodium not necessarily by inhibition of DNMTs. In the study with
Table 4 Overview of studies examining global DNA methylation and specific gene promoter methylation in zebra fish embryos following exposure to several compounds

Compound Concentration (M) Stage Exposure time Effect Assay Study

Benzo[a]pyrene 1.0E−7 1 hpf 96 h Global hypomethylation ELISA Fang et al. (2013)


Environ Sci Pollut Res (2015) 22:16262–16276

Specific: vasaa
Benzo[a]pyrene 2.0E−7 Mother/embryo 3.3 and 96 h Global hypomethylation ELISA- based bisulfite Corrales et al. (2014a, b)
Specific: c-fos, cyp1b1, dazl, gstp1, mlh1, nqo1, multiplex deep
pten, p53, sox2, sox3a sequencing
Sodium arsenite 2.0E−3 1 hpf 24–48 h Global hypomethylation (24 hpf), global Immunostaining Li et al. (2009)
hypermethylation (48 hpf)
Tris(1,3-dichloro-2-propyl) phosphate 3.0E−6 1 hpf 2h Global hypomethylation Methylation-sensitive McGee et al. (2012)
restriction enzymes
5-Azacytidine 7.5E−5 1 hpf 24 h Global hypomethylation (48 hpf) Methylation-sensitive Martin et al. (1999)
restriction enzymes
5-Azacytidine 10b Adult female 32 days Global hypomethylation (F0), many differentially HPLC-UV MeDIP array Olsvik et al. (2014)
methylated genes in F0 and F1, but few in F2
(one unkown and adra2aa)
Flavone derivates 2.5E−5 1 hpf 24 h Global hypomethylation Immunostaining Ceccaldi et al. (2011)
17α ethinylestradiol 3.4E−10 5–7 monthsa 14 days Vitellogenin I hypomethylation Bisulfite sequencing Strömqvist et al. (2010)
2,3,7,8-Tetrachlorodibenzo-p-dioxin 0.02b Adult female 47 days Few differentially methylated genes in F0, but few MeDIP array Olsvik et al. (2014)
in F1 (F2 not analyzed)
Methyl mercury 10b Adult female 47 days Many differentially methylated genes in F0 and F1, MeDIP array Olsvik et al. (2014)
but one in F2 (rRNA)
a
In liver—functions of genes: vasa and dazl, germ cell development; c-fos, proto-oncogene; cyp1b1, steroid metabolism; gstp1, glutathione metabolism; nqo1, reduction of quinones; mlh1, mismatch
repair; pten, tumor suppressor; p53, tumor suppressor, apoptosis; sox2 and sox3, development, adra2a, adrenoceptor alpha 2A
b
Via feed (milligrams per kilogram)
16271
16272 Environ Sci Pollut Res (2015) 22:16262–16276

TDCPP, methylation was measured at a very early time point caused by compounds directly or as a consequence of their
(2 hpf), and at this early stage, exposure to the chemical could (upstream) toxicological mode of action.
influence cell proliferation, leading to developmental delay There is as of yet little known about promoter-specific
and causing different methylation levels relative to controls. It methylation in zebra fish. In 3.3- and 96-hpf zebra fish ex-
would be important to know if methylation changes caused by posed to B[a]P, differentially methylated promoters were
exposure during early development persist to later stages. found in genes that are involved in early development and
Given the limited number of studies on toxicant effects on metabolism and in proto-oncogenes (Table 4) (Fang et al.
DNMTs, further research is clearly needed. To address this 2013; Corrales et al. 2014a). In this study, however, the
issue, Ceccaldi and colleagues (2011) have developed a changes in promoter methylation were modest and require
screening assay in 96-well plates coated with double- conformation. In adult zebra fish, demethylation of the vitel-
stranded DNA labelled with a fluorophore at one end and a logenin (vtg) gene promoter was observed in livers following
biotin on the other end. When DNMT3A/3L complex is added exposure to 0.34 nM 17α ethinylestradiol (EE2) for 14 days
in combination with SAM, DNA will become methylated. (Table 4) (Strömqvist et al. 2010). These results might indicate
With methylation-sensitive restriction enzymes, these methyl- an effect of estrogenic compounds on the methylation status of
ated pieces of DNA are protected from cleavage and retain the vtg promoter, and since EE2 has shown to increase vtg
their fluorescence after washing. Upon addition of an inhibitor mRNA at even lower concentrations, this promoter demeth-
the DNA remains unmethylated, will be cleaved and the ylation could have a positive effect on the gene expression of
fluorescent signal is lost (Ceccaldi et al. 2011). With this vtg (Strömqvist et al. 2010).
method, over 100 flavone and flavanone derivates and, more Since DNA methylation can be inherited, many studies on
recently, over 1,000 small organic compounds could be effi- mammalian models focus on the transgenerational effects of
ciently screened, some of them are more potent DNMT inhib- environmental stressors on the methylome (Skinner 2013;
itors than 5-AC (Ceccaldi et al. 2011, 2013). Moreover, zebra Vandegehuchte and Janssen 2014). In zebra fish,
fish embryos exposed to two potent DNMT-inhibiting deri- transgenerational studies are emerging (Corrales et al.
vates of flavones identified by this screening assay showed 2014b; Baker et al. 2014); however, limited data is available
similar phenotypical changes as with 5-AC exposure about transgenerational effects of toxicants on DNA methyl-
(Ceccaldi et al. 2011). In future research, similar in vitro ation. A recent study assessed the transgenerational effects of
assays could be developed for enzymes such as zebrafish 5-AC, TCDD, and MeHg in zebra fish (Olsvik et al. 2014).
Apobec and Tet as well as for histone methyltransferases, of Adult female zebra fish were exposed via feed to the com-
which an assay for histone methyltransferase G9a has already pounds (Table 4), and offspring from two generations were
been developed (Ceccaldi et al. 2013). assessed at 3 dpf for methylation changes. Limited effects on
promoter methylation in the second generation were observed
Compound-specific effects on DNA methylation at specific with all compounds, indicating a lack of transgenerational
loci effects of these toxicants in zebra fish (Table 4). However, it
should be noted that only females were exposed, which ex-
As global methylation analyses do not reveal specific gene cludes possible effects on male spermatogenesis. Also, the
targets altered by toxicant exposure, research on specific exposure window did not include DNA methylation
promoter methylation of relevant genes in different species reprogramming during embryonic and PGC development
is increasing (Vandegehuchte and Janssen 2014). Examples of and therefore could be a less sensitive time window for
endocrine-disrupting chemicals (EDCs) that are able to differ- transgenerational effects. Furthermore, in F1 and F2 genera-
entially methylate specific gene promoters in species other tions, whole embryos were assessed for methylation changes
than zebra fish are diethylstilbestrol (DES) (Li et al. 2003), which could dilute tissue-specific effects on the methylome.
bisphenol A (Dolinoy et al. 2007), TCDD (Wu et al. 2004), Clearly, with more improved study designs, more research on
the phytoestrogen genistein (Fukata and Mori 2004), TBT transgenerational effects in the zebra fish is warranted.
(Kirchner et al. 2010), and the organophosphate pesticide
diazinon (Zhang et al. 2012). Furthermore, specific
hypomethylating effects on the Pparγ2 promoter have been
observed following exposures of polybrominated biphenyl Conclusions and outlook
ether-47 (BDE-47) in an in vitro adipocyte differentiation
model (Kamstra et al. 2014). These compounds could alter The genome of zebra fish is heavily methylated, like that of
methylation via endocrine pathways; however, it is not ruled other vertebrates, and it is therefore not surprising that they
out that these compounds could also act via interfering with have maintenance and several de novo DNMTs and methyl
the methylation pathways, as is suggested with As. Therefore, CpG-binding (MBD) proteins (Bogdanović and Veenstra
it is not known whether these changes in methylation are 2009). In addition to mC, hmC has also been found as well
Environ Sci Pollut Res (2015) 22:16262–16276 16273

as TET enzymes responsible for the oxidation of mC into information regarding predicted transgenerational effects be-
hmC. Furthermore, demethylation in zebra fish involves de- tween species. Transgenerational effects in zebra fish remain
amination by Aid and Apobec enzymes, followed by BER. to be studied in greater detail, similar to rodent studies, in
Thus, zebra fish share the basic methylation and demethyla- order to assess whether heritable effects of DNA methylation
tion protein machinery with mammals, implying that zebra exist across species. The strength of the zebra fish is that
fish could be used as a model system or reporter for the genetic tools are available to understand the mechanisms
identification of compounds in the environment that affect underpinning DNA methylation changes. Functional studies,
DNA methylation. Despite the similarities in enzymes and like gene inactivation or overexpression, as well as protein
other proteins involved DNA methylation, there are differ- analysis, can be performed to further elucidate the role of
ences in the ways DNA methylation is applied to control, for proteins involved in methylation pathways and their interac-
example, development in zebra fish and mammals. The most tion with toxicants. Such studies can provide additional infor-
striking difference is that in contrast to most mammals, only mation improving the applicability of zebra fish as a suitable
the maternal genome of the zygote is reprogrammed, whereas model for understanding the role of epigenetics in toxicology.
the paternal genome is unchanged. Consequently, it appears
that no mediated hmC reprogramming takes place in the Acknowledgments JK’s work on global and genome-wide methylation
paternal genome. in zebra fish was performed at NMBU Vetbio, Department BasAm in
Oslo. JK was supported by the Netherlands Organization of Scientific
Recent studies emphasize the complexity of DNA methyl- Research (NWO), project number ASPASIA/864.09.005, and partly by
ation and the use of it to control gene expression, including the Research Council of Norway through its Centre of Excellence funding
tissue- and life stage-specific variation and the interactions scheme, project number 223268/F50, Centre for Environmental Radio-
with environmental factors. Zebra fish are a promising model activity (CERAD) 2013-2022 (JK: Bis-seq, zebra fish exposures and lab
consumables). JL was supported by NWO, project number
system in environmental toxicology to study epigenetic ef- VIDI/864.09.005.
fects, since a number of studies show changes in mC levels
and mC patterns after exposure to toxicants. Regarding the
screening of epigenetic toxicity of EDCs, it has been proposed References
to adapt the newly developed zebra fish embryo toxicity test
(OECD guideline 236) to include measurements of global and
Aanes H, Winata CL, Lin CH et al (2011) Zebrafish mRNA sequencing
gene-specific methylation changes (Greally and Jacobs 2013). deciphers novelties in transcriptome dynamics during maternal to
Some considerations need to be taken into account in zygotic transition. Genome Res 21:1328–1338. doi:10.1101/gr.
further application of zebra fish as a model for DNA methyl- 116012.110
ation in environmental toxicology. First, differentially meth- Abdouni H, King JJ, Suliman M et al (2013) Zebrafish AID is capable of
deaminating methylated deoxycytidines. Nucleic Acids Res 41:
ylated DNA regions following exposure to environmental 5457–5468. doi:10.1093/nar/gkt212
contaminants are hard to predict and therefore genome-wide Almeida RD, Loose M, Sottile V et al (2012) 5-Hydroxymethyl-cytosine
analyses (e.g., MeDIP arrays, WGBS) are recommended as enrichment of non-committed cells is not a universal feature of
the first screening step for effects on DNA methylation after vertebrate development. Epigenetics 7:383–389. doi:10.4161/epi.
19375
exposure. This may reveal specific sequences that can be used Andersen IS, Ostrup O, Lindeman LC et al (2012a) Epigenetic complex-
as epigenetic biomarkers, which later can be analyzed with ity during the zebrafish mid-blastula transition. Biochem Biophys
more specific DNA methylation analyses in high-throughput Res Commun 417:1139–1144. doi:10.1016/j.bbrc.2011.12.077
screens, like MS-HRM or pyrosequencing. This would allow Andersen IS, Reiner AH, Aanes H et al (2012b) Developmental features
of DNA methylation during activation of the embryonic zebrafish
the screening of multiple compounds at different stages of genome. Genome Biol 13:R65. doi:10.1186/gb-2012-13-7-r65
zebra fish development and exposure windows. Second, it is a Baker TR, Peterson RE, Heideman W (2014) Using zebrafish as a model
challenge to assess chemical-mediated methylation effects system for studying the transgenerational effects of dioxin. Toxicol
during the dynamic demethylation and remethylation events Sci 138:403–411. doi:10.1093/toxsci/kfu006
Bastos Sales L, Kamstra JH, Cenijn PH et al (2013) Effects of endocrine
before ZGA onset. This is an extremely important stage to disrupting chemicals on in vitro global DNA methylation and adi-
include in toxicity testing, but care needs to be taken to ensure pocyte differentiation. Toxicol Vitr 27:1634–1643. doi:10.1016/j.
that early exposures do not inhibit cell proliferation of devel- tiv.2013.04.005
oping zebra fish, resulting in differences in methylation dy- Baylin SB, Jones PA (2011) A decade of exploring the cancer epige-
nome—biological and translational implications. Nat Rev Cancer
namics between controlled and exposed groups, rather than 11:726–734. doi:10.1038/nrc3130
chemical-specific effects on methylation. Third, no informa- Bird A (2007) Perceptions of epigenetics. Nature 447:396–398. doi:10.
tion is available on the role of DNA methylation in parent of 1038/nature05913
origin methylation marks and PGC development in zebra fish. Bogdanović O, Veenstra GJC (2009) DNA methylation and methyl-CpG
binding proteins: developmental requirements and function.
Comparative studies should be conducted to map differences Chromosoma 118:549–565. doi:10.1007/s00412-009-0221-9
in methylation dynamics of PGC development between mam- Bollati V, Baccarelli A (2010) Environmental epigenetics. Hered (Edinb)
mals and zebra fish. Such studies could provide pivotal 105:105–112. doi:10.1038/hdy.2010.2
16274 Environ Sci Pollut Res (2015) 22:16262–16276

Ceccaldi A, Rajavelu A, Champion C et al (2011) C5-DNA methyltrans- Hermann A, Gowher H, Jeltsch A (2004) Biochemistry and biology of
ferase inhibitors: from screening to effects on zebrafish embryo mammalian DNA methyltransferases. Cell Mol Life Sci 61:2571–
development. Chembiochem 12:1337–1345. doi:10.1002/cbic. 2587. doi:10.1007/s00018-004-4201-1
201100130 Hernández HG, Tse MY, Pang SC et al (2013) Optimizing methodologies
Ceccaldi A, Rajavelu A, Ragozin S et al (2013) Identification of novel for PCR-based DNA methylation analysis. Biotechniques 55:181–
inhibitors of DNA methylation by screening of a chemical library. 197. doi:10.2144/000114087
ACS Chem Biol 8:543–548. doi:10.1021/cb300565z Houwing S, Kamminga LM, Berezikov E et al (2007) A role for Piwi and
Cermak T, Doyle EL, Christian M et al (2011) Efficient design and piRNAs in germ cell maintenance and transposon silencing in
assembly of custom TALEN and other TAL effector-based con- zebrafish. Cell 129:69–82. doi:10.1016/j.cell.2007.03.026
structs for DNA targeting. Nucleic Acids Res 39:e82. doi:10.1093/ Hwang WY, Fu Y, Reyon D et al (2013) Efficient genome editing in
nar/gkr218 zebrafish using a CRISPR-Cas system. Nat Biotechnol 31:227–229.
Chen H, Li S, Liu J et al (2004) Chronic inorganic arsenic exposure doi:10.1038/nbt.2501
induces hepatic global and individual gene hypomethylation: impli- Jiang L, Zhang J, Wang J-J et al (2013) Sperm, but not oocyte, DNA
cations for arsenic hepatocarcinogenesis. Carcinogenesis 25:1779– methylome is inherited by zebrafish early embryos. Cell 153:773–
1786. doi:10.1093/carcin/bgh161 784. doi:10.1016/j.cell.2013.04.041
Corley-Smith GE, Lim CJ, Brandhorst BP (1996) Production of andro- Jirtle RL, Skinner MK (2007) Environmental epigenomics and disease
genetic zebrafish (Danio rerio). Genetics 142:1265–1276 susceptibility. Nat Rev Genet 8:253–262. doi:10.1038/nrg2045
Corrales J, Fang X, Thornton C et al (2014a) Effects on specific promoter Jones PA (2012) Functions of DNA methylation: islands, start sites, gene
DNA methylation in zebrafish embryos and larvae following bodies and beyond. Nat Rev Genet 13:484–492. doi:10.1038/
benzo[a]pyrene exposure. Comp Biochem Physiol C Toxicol nrg3230
Pharmacol 163:37–46. doi:10.1016/j.cbpc.2014.02.005 Kamstra JH, Hruba E, Blumberg B et al (2014) Transcriptional and
Corrales J, Thornton C, White M, Willett KL (2014b) Multigenerational epigenetic mechanisms underlying enhanced in vitro adipocyte dif-
effects of benzo[a]pyrene exposure on survival and developmental ferentiation by the brominated flame retardant BDE-47. Environ Sci
deformities in zebrafish larvae. Aquat Toxicol 148:16–26. doi:10. Technol. doi:10.1021/es405524b
1016/j.aquatox.2013.12.028 Kimmel CB, Ballard WW, Kimmel SR et al (1995) Stages of embryonic
Deaton AM, Bird A (2011) CpG islands and the regulation of transcrip- development of the zebrafish. Dev Dyn 203:253–310. doi:10.1002/
tion. Genes Dev 25:1010–1022. doi:10.1101/gad.2037511 aja.1002030302
Dolinoy DC, Huang D, Jirtle RL (2007) Maternal nutrient supplementa- Kirchner S, Kieu T, Chow C et al (2010) Prenatal exposure to the
tion counteracts bisphenol A-induced DNA hypomethylation in environmental obesogen tributyltin predisposes multipotent stem
early development. Proc Natl Acad Sci U S A 104:13056–13061. cells to become adipocytes. Mol Endocrinol 24:526–539. doi:10.
doi:10.1073/pnas.0703739104 1210/me.2009-0261
Ensembl (2013) Mouse, zebrafish and human assembly and gene anno- Kobayashi H, Sakurai T, Imai M et al (2012) Contribution of intragenic
tation. http://www.ensembl.org/. Accessed 28 Aug 2013 DNA methylation in mouse gametic DNA methylomes to establish
Fang X, Thornton C, Scheffler BE, Willett KL (2013) Benzo[a]pyrene oocyte-specific heritable marks. PLoS Genet 8:e1002440. doi:10.
decreases global and gene specific DNA methylation during 1371/journal.pgen.1002440
zebrafish development. Environ Toxicol Pharmacol 36:40–50. doi: Landsverk ML, Weiser DC, Hannibal MC, Kimelman D (2010)
10.1016/j.etap.2013.02.014 Alternative splicing of sept9a and sept9b in zebrafish produces
Feng S, Cokus SJ, Zhang X et al (2010) Conservation and divergence of multiple mRNA transcripts expressed throughout development.
methylation patterning in plants and animals. Proc Natl Acad Sci U PLoS One 5:e10712. doi:10.1371/journal.pone.0010712
S A 107:8689–8694. doi:10.1073/pnas.1002720107 Law JA, Jacobsen SE (2010) Establishing, maintaining and modifying
Fukata H, Mori C (2004) Epigenetic alteration by the chemical sub- DNA methylation patterns in plants and animals. Nat Rev Genet 11:
stances, food and environmental factors. Reprod Med Biol 3:115– 204–220. doi:10.1038/nrg2719
121 Li W, Liu M (2011) Distribution of 5-hydroxymethylcytosine in different
Gardiner-Garden M, Frommer M (1987) CpG islands in vertebrate ge- human tissues. J Nucleic Acids 2011:870726. doi:10.4061/2011/
nomes. J Mol Biol 196:261–282 870726
Ge L, Zhang R-P, Wan F et al (2014) TET2 plays an essential role in Li S, Hansman R, Newbold R et al (2003) Neonatal diethylstilbestrol
erythropoiesis by regulating lineage-specific genes via DNA oxida- exposure induces persistent elevation of c-fos expression and hypo-
tive demethylation in a zebrafish model. Mol Cell Biol 34:989– methylation in its exon-4 in mouse uterus. Mol Carcinog 38:78–84.
1002. doi:10.1128/MCB.01061-13 doi:10.1002/mc.10147
Globisch D, Münzel M, Müller M et al (2010) Tissue distribution of 5- Li D, Lu C, Wang J et al (2009) Developmental mechanisms of arsenite
hydroxymethylcytosine and search for active demethylation inter- toxicity in zebrafish (Danio rerio) embryos. Aquat Toxicol 91:229–
mediates. PLoS One 5:e15367. doi:10.1371/journal.pone.0015367 237. doi:10.1016/j.aquatox.2008.11.007
Gokul G, Khosla S (2013) DNA methylation and cancer. In: Kundu TK Lindeman LC, Reiner AH, Mathavan S et al (2010a) Tiling histone H3
(ed) Epigenetics: development and disease. Springer, Dordrecht, pp lysine 4 and 27 methylation in zebrafish using high-density micro-
597–625 arrays. PLoS One 5:e15651. doi:10.1371/journal.pone.0015651
Greally JM, Jacobs MN (2013) In vitro and in vivo testing methods of Lindeman LC, Winata CL, Aanes H et al (2010b) Chromatin states of
epigenomic endpoints for evaluating endocrine disruptors. ALTEX developmentally-regulated genes revealed by DNA and histone
30:445–471 methylation patterns in zebrafish embryos. Int J Dev Biol 54:803–
Gu H, Smith ZD, Bock C et al (2011) Preparation of reduced 813. doi:10.1387/ijdb.103081ll
representation bisulfite sequencing libraries for genome-scale Lindeman LC, Andersen IS, Reiner AH et al (2011) Prepatterning of
DNA methylation profiling. Nat Protoc 6:468–481. doi:10. developmental gene expression by modified histones before zygotic
1038/nprot.2010.190 genome activation. Dev Cell 21:993–1004. doi:10.1016/j.devcel.
Hamdi M, Yoshinaga M, Packianathan C et al (2012) Identification of an 2011.10.008
S-adenosylmethionine (SAM) dependent arsenic methyltransferase Lister R, Pelizzola M, Dowen RH et al (2009) Human DNA methylomes
in Danio rerio. Toxicol Appl Pharmacol 262:185–193. doi:10.1016/ at base resolution show widespread epigenomic differences. Nature
j.taap.2012.04.035 462:315–322. doi:10.1038/nature08514
Environ Sci Pollut Res (2015) 22:16262–16276 16275

Long HK, Sims D, Heger A et al (2013) Epigenetic conservation at gene Schier AF (2007) The maternal-zygotic transition: death and birth of
regulatory elements revealed by non-methylated DNA profiling in RNAs. Science 316:406–407. doi:10.1126/science.1140693
seven vertebrates. Elife 2:e00348. doi:10.7554/eLife.00348 Seritrakul P, Gross JM (2014) Expression of the de novo DNA methyl-
MacKay AB, Mhanni AA, McGowan RA, Krone PH (2007) transferases (dnmt3 - dnmt8) during zebrafish lens development.
Immunological detection of changes in genomic DNA methylation Dev Dyn 243:350–356. doi:10.1002/dvdy.24077
during early zebrafish development. Genome 50:778–785. doi:10. Shimoda N, Yamakoshi K, Miyake A, Takeda H (2005) Identification of a
1139/g07-055 gene required for de novo DNA methylation of the zebrafish no tail
Macleod D, Clark VH, Bird A (1999) Absence of genome-wide changes gene. Dev Dyn 233:1509–1516. doi:10.1002/dvdy.20455
in DNA methylation during development of the zebrafish. Nat Skinner MK (2013) Environmental epigenetics and transgenerational
Genet 23:139–140. doi:10.1038/13767 epigenetic inheritance. In: Jirtle RL, Tyson FL (eds)
Martin CC, Laforest L, Akimenko MA, Ekker M (1999) A role for DNA Environmental epigenomics in health disease. Springer, Berlin, pp
methylation in gastrulation and somite patterning. Dev Biol 206: 245–256
189–205. doi:10.1006/dbio.1998.9105 Smallwood SA, Kelsey G (2012) De novo DNA methylation: a germ cell
Mathavan S, Lee SGP, Mak A et al (2005) Transcriptome analysis of perspective. Trends Genet 28:33–42. doi:10.1016/j.tig.2011.09.004
zebrafish embryogenesis using microarrays. PLoS Genet 1:260– Smith THL, Collins TM, McGowan RA (2011) Expression of the dnmt3
276. doi:10.1371/journal.pgen.0010029 genes in zebrafish development: similarity to Dnmt3a and Dnmt3b.
McGee SP, Cooper EM, Stapleton HM, Volz DC (2012) Early zebrafish Dev Genes Evol 220:347–353. doi:10.1007/s00427-010-0347-z
embryogenesis is susceptible to developmental TDCPP exposure. Smith ZD, Chan MM, Mikkelsen TS et al (2012) A unique regulatory
Environ Health Perspect 120:1585–1591. doi:10.1289/ehp.1205316 phase of DNA methylation in the early mammalian embryo. Nature
Mellén M, Ayata P, Dewell S et al (2012) MeCP2 binds to 5hmC enriched 484:339–344. doi:10.1038/nature10960
within active genes and accessible chromatin in the nervous system. Strähle U, Scholz S, Geisler R et al (2012) Zebrafish embryos as an
Cell 151:1417–1430. doi:10.1016/j.cell.2012.11.022 alternative to animal experiments—a commentary on the definition
Mhanni AA, McGowan RA (2004) Global changes in genomic methyl- of the onset of protected life stages in animal welfare regulations.
ation levels during early development of the zebrafish embryo. Dev Reprod Toxicol 33:128–132. doi:10.1016/j.reprotox.2011.06.121
Genes Evol 214:412–417. doi:10.1007/s00427-004-0418-0 Strömqvist M, Tooke N, Brunström B (2010) DNA methylation levels in
Mhanni AA, Yoder JA, Dubesky C, McGowan RA (2001) Cloning and the 5′ flanking region of the vitellogenin I gene in liver and brain of
sequence analysis of a zebrafish cDNA encoding DNA (cytosine-5)- adult zebrafish (Danio rerio)—sex and tissue differences and effects
methyltransferase-1. Genesis 30:213–219. doi:10.1002/gene.1067 of 17alpha-ethinylestradiol exposure. Aquat Toxicol 98:275–281.
Nestor CE, Ottaviano R, Reddington J et al (2012) Tissue type is a major doi:10.1016/j.aquatox.2010.02.023
modifier of the 5-hydroxymethylcytosine content of human genes. Takai D, Jones PA (2002) Comprehensive analysis of CpG islands in
Genome Res 22:467–477. doi:10.1101/gr.126417.111 human chromosomes 21 and 22. Proc Natl Acad Sci U S A 99:
Olsvik PA, Williams TD, Tung H-S et al (2014) Impacts of TCDD and 3740–3745. doi:10.1073/pnas.052410099
MeHg on DNA methylation in zebrafish (Danio rerio) across two Takayama K, Shimoda N, Takanaga S et al (2014) Expression patterns of
generations. Comp Biochem Physiol C Toxicol Pharmacol 165C: dnmt3aa, dnmt3ab, and dnmt4 during development and fin regen-
17–27. doi:10.1016/j.cbpc.2014.05.004 eration in zebrafish. Gene Expr Patterns 14:105–110. doi:10.1016/j.
Pastor WA, Aravind L, Rao A (2013) TETonic shift: biological roles of gep.2014.01.005
TET proteins in DNA demethylation and transcription. Nat Rev Mol Tittle RK, Sze R, Ng A et al (2011) Uhrf1 and Dnmt1 are required for
Cell Biol 14:341–356. doi:10.1038/nrm3589 development and maintenance of the zebrafish lens. Dev Biol 350:
Pfeifer GP, Kadam S, Jin S-G (2013) 5-Hydroxymethylcytosine and its 50–63. doi:10.1016/j.ydbio.2010.11.009
potential roles in development and cancer. Epigenetics Chromatin 6: Tucker KL (2001) Methylated cytosine and the brain: a new base for
10. doi:10.1186/1756-8935-6-10 neuroscience. Neuron 30:649–652
Pikulkaew S, Benato F, Celeghin A et al (2011) The knockdown of Vandegehuchte MB, Janssen CR (2014) Epigenetics in an ecotoxicolog-
maternal glucocorticoid receptor mRNA alters embryo development ical context. Mutat Res Genet Toxicol Environ Mutagen 764–765:
in zebrafish. Dev Dyn 240:874–889. doi:10.1002/dvdy.22586 36–45. doi:10.1016/j.mrgentox.2013.08.008
Potok ME, Nix DA, Parnell TJ, Cairns BR (2013) Reprogramming the Vastenhouw NL, Schier AF (2012) Bivalent histone modifications in
maternal zebrafish genome after fertilization to match the paternal early embryogenesis. Curr Opin Cell Biol 24:374–386. doi:10.
methylation pattern. Cell 153:759–772. doi:10.1016/j.cell.2013.04.030 1016/j.ceb.2012.03.009
Probst AV, Dunleavy E, Almouzni G (2009) Epigenetic inheritance Wang J, Wu Z, Li D et al (2012) Nutrition, epigenetics, and metabolic
during the cell cycle. Nat Rev Mol Cell Biol 10:192–206. doi:10. syndrome. Antioxid Redox Signal 17:282–301. doi:10.1089/ars.
1038/nrm2640 2011.4381
Rai K, Nadauld LD, Chidester S et al (2006) Zebra fish Dnmt1 and Wang H, Yang H, Shivalila CS et al (2013) One-step generation of mice
Suv39h1 regulate organ-specific terminal differentiation during de- carrying mutations in multiple genes by CRISPR/Cas-mediated
velopment. Mol Cell Biol 26:7077–7085. doi:10.1128/MCB.00312- genome engineering. Cell 153:910–918. doi:10.1016/j.cell.2013.
06 04.025
Rai K, Huggins IJ, James SR et al (2008) DNA demethylation in Wei C, Salichos L, Wittgrove CM et al (2012) Transcriptome-wide
zebrafish involves the coupling of a deaminase, a glycosylase, and analysis of small RNA expression in early zebrafish development.
gadd45. Cell 135:1201–1212. doi:10.1016/j.cell.2008.11.042 RNA 18:915–929. doi:10.1261/rna.029090.111
Rai K, Jafri IF, Chidester S et al (2010) Dnmt3 and G9a cooperate for Weisenberger DJ, Romano LJ (1999) Cytosine methylation in a CpG
tissue-specific development in zebrafish. J Biol Chem 285:4110– sequence leads to enhanced reactivity with benzo[a]pyrene diol
4121. doi:10.1074/jbc.M109.073676 epoxide that correlates with a conformational change. J Biol Chem
Ruthenburg AJ, Li H, Patel DJ, Allis CD (2007) Multivalent engagement 274:23948–23955
of chromatin modifications by linked binding modules. Nat Rev Williams TD, Mirbahai L, Chipman JK (2014) The toxicological applica-
Mol Cell Biol 8:983–994. doi:10.1038/nrm2298 tion of transcriptomics and epigenomics in zebrafish and other tele-
Ruzov A, Tsenkina Y, Serio A et al (2011) Lineage-specific distribution osts. Brief Funct Genomics 13:157–171. doi:10.1093/bfgp/elt053
of high levels of genomic 5-hydroxymethylcytosine in mammalian Wu SC, Zhang Y (2010) Active DNA demethylation: many roads lead to
development. Cell Res 21:1332–1342. doi:10.1038/cr.2011.113 Rome. Nat Rev Mol Cell Biol 11:607–620. doi:10.1038/nrm2950
16276 Environ Sci Pollut Res (2015) 22:16262–16276

Wu H, Zhang Y (2014) Reversing DNA methylation: mechanisms, Yildirim O, Li R, Hung J-H et al (2011) Mbd3/NURD complex
genomics, and biological functions. Cell 156:45–68. doi:10.1016/j. regulates expression of 5-hydroxymethylcytosine marked
cell.2013.12.019 genes in embryonic stem cells. Cell 147:1498–1510. doi:10.
Wu Q, Ohsako S, Ishimura R et al (2004) Exposure of mouse preimplan- 1016/j.cell.2011.11.054
tation embryos to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) al- Yokomine T, Hata K, Tsudzuki M, Sasaki H (2006) Evolution of the
ters the methylation status of imprinted genes H19 and Igf2. Biol vertebrate DNMT3 gene family: a possible link between existence
Reprod 70:1790–1797. doi:10.1095/biolreprod.103.025387 of DNMT3L and genomic imprinting. Cytogenet Genome Res 113:
Wu H, Caffo B, Jaffee HA et al (2010) Redefining CpG islands using 75–80. doi:10.1159/000090817
hidden Markov models. Biostatistics 11:499–514. doi:10.1093/ Zhang X, Wallace AD, Du P et al (2012) Genome-wide study of DNA
biostatistics/kxq005 methylation alterations in response to diazinon exposure in vitro.
Wu S-F, Zhang H, Hammoud SS et al (2011) DNA methylation profiling Environ Toxicol Pharmacol 34:959–968. doi:10.1016/j.etap.2012.
in zebrafish. Methods Cell Biol 104:327–339. doi:10.1016/B978-0- 07.012
12-374814-0.00018-5 Zhao CQ, Young MR, Diwan BA et al (1997) Association of arsenic-
Yamakoshi K, Shimoda N (2003) De novo DNA methylation at the CpG induced malignant transformation with DNA hypomethylation and
island of the zebrafish no tail gene. Genesis 37:195–202. doi:10. aberrant gene expression. Proc Natl Acad Sci U S A 94:10907–
1002/gene.10245 10912

You might also like