You are on page 1of 12

MARGEN-00423; No of Pages 12

Marine Genomics xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Marine Genomics

Genome Perspectives

Epigenomics in marine fishes


David C.H. Metzger ⁎, Patricia M. Schulte
Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Epigenetic mechanisms are an underappreciated and often ignored component of an organism's response to en-
Received 5 January 2016 vironmental change and may underlie many types of phenotypic plasticity. Recent technological advances in
Received in revised form 20 January 2016 methods for detecting epigenetic marks at a whole-genome scale have launched new opportunities for studying
Accepted 21 January 2016
epigenomics in ecologically relevant non-model systems. The study of ecological epigenomics holds great prom-
Available online xxxx
ise to better understand the linkages between genotype, phenotype, and the environment and to explore mech-
Keywords:
anisms of phenotypic plasticity. The many attributes of marine fish species, including their high diversity, variable
DNA methylation life histories, high fecundity, impressive plasticity, and economic value provide unique opportunities for studying
Epigenetics epigenetic mechanisms in an environmental context. To provide a primer on epigenomic research for fish biolo-
Epigenomics gists, we start by describing fundamental aspects of epigenetics, focusing on the most widely studied and most
Environment well understood of the epigenetic marks: DNA methylation. We then describe the techniques that have been
Fishes used to investigate DNA methylation in marine fishes to date and highlight some new techniques that hold
great promise for future studies. Epigenomic research in marine fishes is in its early stages, so we first briefly dis-
cuss what has been learned about the establishment, maintenance, and function of DNA methylation in fishes
from studies in zebrafish and then summarize the studies demonstrating the pervasive effects of the environ-
ment on the epigenomes of marine fishes. We conclude by highlighting the potential for ongoing research on
the epigenomics of marine fishes to reveal critical aspects of the interaction between organisms and their
environments.
© 2016 Elsevier B.V. All rights reserved.

1. Introduction These longer-lasting forms of plasticity can be grouped together


under the umbrella term of “epigenetics”. Much of the research on epi-
Environmentally-induced phenotypic plasticity is a critical compo- genetic mechanisms has been performed in model systems (Bird, 2002;
nent of organismal responses to a changing environment. Understand- Li and Zhang, 2014), but recent epigenetic studies in ecologically rele-
ing the molecular mechanisms underlying this plasticity is an important vant non-model organisms have begun to demonstrate the promise of
question in basic biology that also has important applications in areas as epigenetic and epigenomic approaches in fields such as ecology, evolu-
diverse as aquaculture and environmental monitoring. Much of the tionary biology and environmental biology (Bonasio, 2015; Bossdorf
work on the mechanisms of environmentally-induced plasticity in fish- et al., 2008; Ledón-Rettig, 2013). Here, we provide a primer for the ap-
es has focused on the reversible phenotypic plasticity that is character- plication of epigenetic and epigenomic research in marine fishes.
istic of acclimation responses, and this type of plasticity has been a
major focus of studies in marine genomics with many studies using 1.1. What is epigenetics?
techniques such as cDNA microarray to examine changes in RNA levels
in response to environmental change (Cossins, 2006; Gracey et al., Conrad Waddington was first credited with using the term “epige-
2004; Kalujnaia et al., 2007; Logan and Buckley, 2015). Less well under- netics” in the 1940s to describe the developmental mechanisms that
stood are the longer-lasting phenotypic changes that occur in response can give rise to alternative phenotypes (Waddington, 1942), but since
to environmental changes that are experienced during development, this time there has been almost continuous refinement of this concept
a phenomenon known as developmental plasticity (Alvarado et al., (Deans and Maggert, 2015). More contemporary definitions are intrin-
2015; Pfennig et al., 2010; Scott and Johnston, 2012). Some of these ac- sically tied to aspects of phenotypic plasticity, or how a single genotype
quired phenotypes can even last for multiple generations, in what has can produce multiple phenotypes in response to changes in the envi-
been termed transgenerational plasticity. ronment. Thus, the broadest definitions of epigenetics include any pro-
cess that results in heritable phenotypic changes such as variation in
⁎ Corresponding author at: Department of Zoology, University of British Columbia, 6270
gene expression. Several attempts have been made, however, to refine
University Blvd., Vancouver, British Columbia V6T 1Z4, Canada. the definition of epigenetics in a way that distinguishes epigenetic
E-mail address: dmetzger@zoology.ubc.ca (D.C.H. Metzger). mechanisms from broader processes that regulate gene expression

http://dx.doi.org/10.1016/j.margen.2016.01.004
1874-7787/© 2016 Elsevier B.V. All rights reserved.

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
2 D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx

and phenotypic plasticity. The most widely accepted contemporary def- genomes are generally methylated to high levels (N 80%). Invertebrates
initions of epigenetics echo the one put forward by Deans and Maggert exhibit a wide range of CpG methylation levels, with the “model” inver-
(2015) who define epigenetics as “The study of phenomena and mech- tebrates Drosophila melanogaster and Caenorhabditis elegans essentially
anisms that cause chromosome-bound, heritable changes to gene lacking DNA methylation and the honeybee having very low CpG meth-
expression that are not dependent on changes to DNA sequence.” ylation levels (~.7%; Lyko et al., 2010). Many other invertebrates have
In this context, the word heritability is defined as involving both intermediate levels of CpG methylation (around 50%) with a “mosaic”
meiotic and mitotic inheritance (Wu and Morris, 2001), and thus ac- pattern of methylation such that the genome consists of interspersed re-
cording to these definitions, epigenetic mechanisms need not be gions of methylated and unmethylated DNA of similar length (Feng
confined to processes that are inherited across generations, but et al., 2010). In contrast, vertebrate genomes consist of long tracts in
must at least be inherited across cell divisions. However, not all which the CpG dinucleotides are methylated to high levels (N 80%)
epigeneticists subscribe to this definition. For example, the field of punctuated by short unmethylated regions. These unmethylated re-
epigenomics (or the study of epigenetics at a whole-genome level) gions typically fall in areas called CpG islands (CGI), which have a GC-
tends to follow a broader definition that includes additional forms rich base composition with a CpG dinucleotide approximately every
of phenotypic plasticity. For example, the NIH epigenomic roadmap 10 base pairs. CpG islands tend to be preferentially located in the prox-
program uses the following definition: “epigenetics refers to both imal promoter regions of genes (Fig. 1; Deaton and Bird, 2011), and this
heritable changes in gene activity and expression (in the progeny accounts for an observed “dip” in the levels of DNA methylation around
of cells or of individuals) and also stable, long-term alterations in the transcription start sites of actively transcribed genes (Jones, 2012;
the transcriptional potential of a cell that are not necessarily herita- Zemach et al., 2010).
ble” (http://www.roadmapepigenomics.org/overview). There is also epigenetic variation among individuals within a species
Because there is still disagreement about how to most appropriately in DNA methylation patterns (e.g. Massicotte et al., 2011; Liebl et al.,
define the field of epigenetics, the mechanisms that constitute an epige- 2013). This individual-level variation in DNA methylation can be main-
netic signal or process are also controversial (Deans and Maggert, 2015; tained throughout the lifespan of an organism and can even be trans-
Schaefer and Nadeau, 2015). Depending on the author, the list of poten- mitted across generations, providing an added source of heritable
tial epigenetic mechanisms may include covalent modification of DNA variation that is independent of genetic variation. Thus, the potential
through methylation of cytosines (Lister et al., 2009), covalent modifica- impact of epigenetic variation has become an important topic in the
tion of histones (Bannister and Kouzarides, 2011) via acetylation, phos- fields of ecology and evolution (Bossdorf et al., 2008; Ledón-Rettig,
phorylation, methylation and other mechanisms, as well as gene 2013), as it has been suggested to be important for evolutionary adapta-
regulation by non-coding RNAs (as microRNAs (miRNA), endogenous tion to altered environments (Flores et al., 2013).
small interfering RNAs (endo-siRNA), and PIWI-interacting RNAs DNA methylation patterns can also vary within a single individ-
(piRNA) (Costa, 2008). In this review we focus on DNA methylation be- ual across life stages or among cell types (Flanagan et al., 2006;
cause it was the first epigenetic mark to be discovered, it has become Massicotte et al., 2011; Richardson, 2003). DNA methylation dur-
one of the most widely studied of the potential epigenetic mechanisms, ing development is thought to play an important role in cellular
and it clearly fulfills the criterion of being a chromosome-bound, herita- differentiation and is involved in maintaining cell-type specific
ble change that is not dependent on a change to DNA sequence (Deans transcriptional activity in the genome (Li, 2002; Monk et al., 1987). Var-
and Maggert, 2015). iation in DNA methylation patterns that are induced by environmental
factors can alter the developmental trajectory of an individual resulting
1.2. DNA methylation as an epigenetic mark in the expression of alternative phenotypes from a single genotype (i.e.
developmental plasticity) (e.g. Wolff et al., 1998; Kucharski et al., 2008;
DNA methylation is a covalent modification in which a methyl group Shao et al., 2014).
(CH3) is added to position 5 of the pyrimidine ring of a cytosine (5mC). Most of what is known about the mechanisms that establish and
In eukaryotic cells, the majority of methylated cytosines occur on maintain DNA methylation patterns in animals has been derived from
cytosine-phosphate-guanine (CpG) dinucleotides resulting in comple- studies in mammals. In mammals, DNA methylation patterns are
mentary methylation of CpG motifs on both strands of DNA. Methylated established by the de novo DNA methylation activity of the DNA
cytosines can also occur on non-CpG sites such as CpHpG and CpHpH methyltransferases, DNMT3A and DNMT3B, which add methyl groups
motifs (where H = A, C or T). However, these epigenetic marks are to previously unmethylated cytosines (Law and Jacobsen, 2010). Main-
fundamentally different from CpG methylation because they are tenance of DNA methylation across cell divisions is achieved by the ad-
strand-specific. These strand-specific marks are rare in animals, al- dition of methyl groups to the newly synthesized strand of DNA by the
though they are prominent in the epigenetic landscape of plants (Law DNA methyltransferase DNMT1 which is recruited to DNA by the SET-
and Jacobsen, 2010). and RING-associated domain contain protein UHRF1 (Bostick et al.,
The addition or removal of methyl groups can regulate the transcrip-
tional activity of neighboring genes by altering the structure and function
of chromatin. High levels of methylation at the promoter of a gene are as-
sociated with repression of transcription (Li and Zhang, 2014). This re-
pression of transcription by promoter methylation is thought to be due
to the formation of inactive heterochromatin (Martinowich et al., 2003;
Razin, 1998). In addition, methylation of enhancer elements can also re-
press transcription by preventing the binding of the transcription factors
that would otherwise induce expression. Alternatively, methylated DNA
can recruit methyl-binding proteins that repress transcription (Klose and
Bird, 2006). DNA methylation of intragenic regions, however, is associ-
ated with active expression of nearby genes (Jjingo et al., 2012;
Zemach et al., 2010) and the regulation of alternative splice variants Fig. 1. Patterns of CpG island methylation in proximal promoter regions. CpG islands are
(Laurent et al., 2010; Lyko et al., 2010; Park et al., 2011). regions with high concentrations of CpG motifs. Shore and shelf regions are located
immediately flanking CpG islands. A High level of methylation in a CpG island near the
There is dramatic variation in the prevalence and patterns of DNA transcription start site of a gene is associated with inactivation of transcription. Bent
methylation among taxa (Feng et al., 2010). For example, in Arabidopsis arrow indicates transcription start site; empty circles indicate unmethylated CpG sites;
only ~22% of CpG sites are methylated, while the CpG sites in vertebrate filled circles indicate methylated CpG sites.

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx 3

2007; Sharif et al., 2007). Two other genes in the mammalian genome are levels to techniques that provide information about methylation status
related to the described DNA methyltransferase genes but do not possess at the single base-pair level. Each of these approaches has advantages
DNA methyltransferase activity. DNMT3L is a methylation cofactor that and disadvantages (Table 1). Choosing the method that is best suited
has been associated with genomic imprinting (Suetake et al., 2004). for a particular study generally comes down to making a complex
Another methyltransferase, DNMT2, predominantly methylates RNA trade-off that weighs the cost, the resolution of the assay, and the extent
molecules, specifically tRNA, as opposed to DNA (Goll et al., 2006). of the genome that is monitored.
While the establishment of DNA methylation marks in the genome
occurs through active processes, de-methylation can occur via both ac- 2.1. Low resolution assessment of global DNA methylation
tive and passive processes. Passive de-methylation occurs when DNA
methylation is not maintained through DNA replication and cell division The most quantitative method that can be used to evaluate whole-
as a result of the lack of DNMT1 activity (Li and Zhang, 2014). Active genome DNA methylation levels at low resolution is reverse-phase
de-methylation requires the recruitment of protein complexes that con- high performance liquid chromatography (RP-HPLC) (Kuo et al.,
vert methylated cytosines to the methylation intermediates that are 1980). In RP-HPLC, DNA is digested down to single nucleotides, and
then removed by base excision repair mechanisms. The intermediate since cytosines and methylated cytosines have different molecular
forms of methylated cytosines include 5-hydroxy-methyl cytosine masses, they can be distinguished and quantified by HPLC or liquid
(5hmC), 5-formyl-cytosine (5fC), and 5-carboxyl-cytosine (5caC) (Shen chromatography coupled with tandem mass spectrometry (LC–MS/
et al., 2014). A family of enzymes known as the ten-eleven translocation MS). Global levels of DNA methylation can also be quantified using
(TET) enzymes catalyzes the stepwise oxidation of 5mC to 5fC and then ELISA-based methods. In these methods, purified genomic DNA is
5caC, which can then be converted to unmethylated cytosines (Shen hybridized to assay wells and an antibody specific to methylated cy-
et al., 2014). These oxidized derivatives were originally thought to be tosines is used to detect methylated DNA. A secondary enzyme-
transient demethylation intermediates, but recent studies suggest that linked antibody is used for colorimetric quantification. These assays
these marks are more persistent in the genome than originally thought, are available as commercial kits, all of which are suitable for use in
and may have functional significance in their own right (Branco et al., non-model organisms such as marine fishes. These methods provide
2011; Kroeze et al., 2015; Véron and Peters, 2011). a global overview of levels of DNA methylation, which can be useful
for detecting large-scale changes in methylation such as those that
2. Methods for assessing DNA methylation occur during development and tumorigenesis. However, these low-
resolution techniques are unable to detect the many important epi-
There are a number of excellent reviews summarizing the major genetic changes that occur at specific loci, which do not affect the
techniques available for assessing levels of DNA methylation (Harrison overall level of methylation of the genome. Responses to environ-
and Parle-McDermott, 2011; Laird, 2010; Plongthongkum et al., 2014; mental change are likely to involve the upregulation of some genes
Shen and Waterland, 2007; Umer and Herceg, 2013). Here we focus and the down regulation of others, which would be predicted to involve
on the techniques that have been used in marine fishes, or that show either demethylation or hypermethylation of specific loci. Low resolu-
considerable promise for application in future studies. We divide these tion methods, which detect the average change in methylation across
techniques based on their level of resolution from techniques that pro- the genome, cannot capture the complexity of changes in DNA methyl-
vide low-resolution information about whole-genome methylation ation at this scale.

Table 1
Advantages and disadvantages of selected methods for assessing DNA methylation.

Method Resolution Advantages Disadvantages

Reverse phase HPLC Low (whole genome) • No sequence information required • Only very large differences can be detected
(RP-HPLC) • Inexpensive • Very low resolution
ELISA Low (whole genome) • No sequence information required • Only very large differences can be detected
• High throughput • Very low resolution
• Inexpensive
MS-AFLP Low–moderate • No sequence information required • Low resolution
• Inexpensive • Provides +/− information (qualitative)
• Some information about location of meth- • Screens anonymous loci (candidates can be sequenced)
ylated sites can be obtained (if candidate • Reproducibility can be poor
loci sequenced)
Bisulfite sequencing of a candidate Single base pair • All CpG sites in targeted region can be an- • Low coverage of the genome
locus alyzed • Low throughput
• Generates information about location of • Identifying appropriate candidate regions is challenging
methylated sites • Sequence information is required for primer design
• Inexpensive
EpiRADseq Single base pair • High resolution • Expensive
• Quantitative • Requires genomic sequence information
• Genome-wide (~2 million CpG sites ex- • Only examines methylation at HpaII sites
amined per sample; ~6% of total sites)a • Fully methylated sites are not captured
Reduced representation bisulfite Single base pair • High resolution • Expensive
sequencing (RRBS) • Quantitative • Requires genomic sequence information
• Genome-wide (~3 million CpG examined • Only examines sites within ~100 bp of an enzyme rec-
per sample; ~10% of total sites)a ognition site
MeDIP-Seq/MDBSeq 100–300 bp • Fairly high resolution • Expensive
(Single base pair when • Quantitative • Requires genomic sequence information
combined with bisulfite • Genome-wide (~6 million CpG examined • Quantification complex
sequencing) per sample; ~20% of total sites)a
Whole-genome bisulfite Single base pair • High resolution • Extremely expensive
sequencing (WGBS) • Quantitative • Requires sophisticated bioinformatics
• Genome-wide (all CpG sites examined) • Requires assembled genome sequence
a
Based on a 3 Gb genome size.

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
4 D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx

2.2. Methods with intermediate resolution of multiple clones (Frommer et al., 1992), methylation sensitive dot
blot assays (MS-DBA)(Clément and Benhattar, 2005) and pyrosequenc-
The most widely-used method for detecting DNA methylation pat- ing (Dupont et al., 2004).
terns at intermediate resolution is methylation sensitive amplified poly- The primary advantage of taking a single-locus approach is the rela-
morphism analysis (Reyna-López et al., 1997). This method (which goes tively low cost and the fact that this method is fairly accessible for any
by various abbreviations such as MSAP, MS-AFLP etc.) is a modification laboratory equipped for molecular biology. The primary disadvantage
of amplified fragment length polymorphism (AFLP) analysis. MS-AFLP of a single-locus approach is the difficulty of identifying appropriate
relies on parallel digestion of genomic DNA by methylation-sensitive genomic regions of interest. In addition, sufficient sequence informa-
and non-methylation sensitive isoschizomeric restriction enzymes tion must be available so that appropriate primers can be designed.
such as HpaII and MspI. The resulting DNA fragments are then ligated Note that designing PCR primers for use on bisulfite-converted
to adaptors and amplified using fluorescently labeled primers. The DNA can be challenging, because bisulfite conversion alters the se-
resulting PCR products can then be run on a gel or analyzed on a quence of unmethylated cytosines, while leaving methylated cytosines
capillary sequencer. By comparing the banding pattern between DNA untouched. Therefore it is important to design the PCR primers away
digested with MspI and HpaII it is possible to determine the methylation from any regions that have the potential to be methylated. This is partic-
state of a particular locus. These differentially methylated bands can ularly challenging in species with unsequenced genomes.
then by isolated and sequenced to identify differentially methylated The most comprehensive method for assessing patterns of DNA
genes. In addition, the overall banding patterns can provide information methylation at single base-pair resolution is bisulfite conversion of ge-
about variation in methylation patterns among individuals or popula- nomic DNA followed by whole-genome shotgun sequencing (whole-
tions (e.g. Liebl et al., 2013). genome bisulfite sequencing WGBS; Laird, 2010). However, the cost of
The main advantage of MS-AFLP is that it can be applied in species comprehensive sequencing remains prohibitive for organisms with
where genomic information is limited, and it requires no specialized large genomes, such as most marine fishes, and thus WGBS is usually
equipment other than that found in many labs equipped for molecular only applied to questions that can be answered using small numbers
biology. It is also cost-effective, so large numbers of samples can be an- of biological replicates. For example, current recommendations are
alyzed. Since only loci of interest need be sequenced, MS-AFLP has the that 10× genome coverage and at least two biological replicates should
advantage that sequencing resources are not wasted on regions of the be used at a minimum for bisulfite sequencing (Ziller et al., 2014).
genome that do not differ in methylation levels among samples. The Studies looking to address epigenomic questions in an ecological or evo-
most important limitation of MS-AFLP is that the banding patterns pro- lutionary context (including many questions in the epigenomics of ma-
duced can be quite difficult to interpret, because inconsistent restriction rine fishes) typically require larger sample sizes, which makes WGBS
digestion or PCR can easily be confused with changes in methylation prohibitively expensive. As the costs of genome sequencing decrease,
levels. As a result, this method has been challenging to replicate across however, WGBS will become more accessible, and this technique is like-
labs (Fulneček and Kovařík, 2014). An additional concern for studies ly to have much wider application. An additional barrier to applying this
in ecological or evolutionary epigenomics is that genetic variation method stems from the challenges of the associated bioinformatics,
among individuals can be confused with epigenetic variation because which can be difficult even in model organisms (Adusumalli et al.,
differences in banding pattern can be produced by sequence variation 2015; Bock, 2012) and are a particular barrier in species that lack a se-
as well as by epigenomic variation. Finally, MS-AFLP treats methylation quenced genome, such as many marine fishes. However, there has
as a +/− state and cannot distinguish between (for example) a site that been some recent development of bioinformatics methods that should
is 50% methylated and a site that is 100% methylated within a tissue. allow the application of these techniques even in the absence of a se-
These differences in relative levels of methylation could have important quenced genome (Bewick et al., 2015).
phenotypic consequences, and this type of variation cannot be detected A variety of methods have been devised to mitigate the high cost of
by MS-AFLP. As a result, MS-AFLP is gradually being replaced with other WGBS. In general, these methods reduce costs by focusing sequencing
methods (Schrey et al., 2013). effort on the methylated portions of the genome. This enrichment is
performed either using affinity-based or restriction-enzyme based
2.3. Methods with single base-pair resolution approaches. In the methods that rely on affinity-based enrichment,
DNA is first immunoprecipitated with an antibody that recognizes
Most methods for measuring DNA methylation with single base-pair methylated regions of the genome and the immunoprecipitated DNA
resolution rely on bisulfite conversion of genomic DNA. When DNA is is then sequenced. For example, in MeDIPseq (methylated DNA immu-
treated with sodium bisulfite or metabisulfite, the unmethylated cyto- noprecipitation and sequencing; Borgel et al., 2010) genomic DNA is
sine bases (C) in the DNA are chemically converted into the base uracil first sheared and then immunoprecipitated with an antibody that rec-
(U), whereas methylated cytosines (5mC) are protected from this bisul- ognizes methylated cytosines. This technique is particularly suitable
fite conversion. During subsequent PCR amplification, a thymine for use in non-model organisms as the antibody should recognize
(T) base is incorporated at any position that has been converted to a methylated cytosines in all genomes, and has been used successfully
U, which changes the original sequence from a C to a T in unmethylated in zebrafish (Lee et al., 2015). An alternative technique called MDBseq
positions, whereas methylated positions remain as C's (Fig. 2). By com- (methyl CpG binding domain immunoprecipitation and sequencing;
paring the bisulfite converted and untreated genomic DNA sequences it Aberg et al., 2012) uses an antibody that recognizes the Methyl-CpG-
is possible to detect the location of methylated cytosines. Individual binding domain protein 2, which is a nuclear protein that binds spe-
cells within a tissue can vary in whether a particular position is methyl- cifically to methylated DNA. Because the MDBseq method relies on
ated or not, and the initial DNA isolation step that is the starting point of antibody recognition of a specific protein, inter-specific variation of
bisulfite conversion is usually a sample that contains multiple cells. the protein motif could make it difficult to adapt the technique to
Thus, results from these assays provide information on the percentage non-model species. The primary disadvantage of these techniques
of cells that have a methylated cytosine at a particular position. is that in the absence of a bisulfite conversion step, their resolution
Bisulfite treatment can be used to investigate methylation levels at a is limited to the size of the immunoprecipitated DNA fragments (usual-
particular candidate locus. Bisulfite-converted genomic DNA can be am- ly 100-300 bp).
plified using primers directed to a particular candidate region of inter- There are several restriction-enzyme based approaches that enrich
est, followed by detection of the altered bases. The most commonly for areas likely to be targets of DNA methylation, including reduced
used methods for detecting bisulfite-converted sites are real-time quan- representation bisulfite sequencing (RRBS; Gu et al., 2010; Jeddeloh
titative PCR (Eads et al., 2000), cloning followed by Sanger sequencing et al., 2008) and EpiRADseq (Schield et al., 2015). These methods

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx 5

Fig. 2. Bisulfite sequencing. Treatment of genomic DNA with sodium bisulfite converts unmethylated cytosines (C) into uracil (U), while methylated cytosines are protected. During PCR
and sequencing uracil is converted to thymine (T). Sequence comparison of untreated and bisulfite converted DNA sequences allows the detection of methylated sites.

allow sequencing efforts to be focused on methylated regions, which re- 5hMeDIP-seq utilizes antibodies to target 5hmC. Other techniques
duces costs relative to WGBS, but still provides single-base pair resolu- such as oxidative bisulfite sequencing (oxBS-Seq) (Booth et al., 2012),
tion detection of methylation status at a subset of locations within the which targets 5hmC, and reduced bisulfite sequencing (Booth et al.,
genome. 2014), which targets 5fC, rely on modifications of bisulfite sequencing.
In RRBS, genomic DNA is first digested using a methylation- To date these techniques have not been applied in any fish species
insensitive restriction enzyme such as MspI that specifically target CpG other than zebrafish.
motifs. These fragments are then bisulfite treated and sequenced. This
allows sequencing efforts to be concentrated primarily on regions of 3. DNA methylation in zebrafish
the genome that are likely to be methylated (Gu et al., 2010; Jeddeloh
et al., 2008). Although the RRBS technique helps to mitigate the cost Much of what is currently understood about mechanisms and pat-
of sequencing per individual, the downstream analysis of RRBS data in terns of DNA methylation in fish comes from studies conducted in
the absence of a reference genome can be complicated. However, zebrafish, and there are several excellent reviews that focus on this spe-
there are currently a variety of tools are available to help perform cies (Goll and Halpern, 2011; Kamstra et al., 2015). Here, we briefly re-
these analyses in species without a reference genome (e.g. Chen et al., view what is known about DNA methylation in zebrafish. In the next
2010; Stockwell et al., 2014). section we summarize the relatively few studies that have been per-
EpiRADseq (Schield et al., 2015) has recently been developed as an formed in marine and diadromous fishes.
alternative to RRBS that can be applied in the absence of genome
sequence information. EpiRADseq is a modification of a technique 3.1. Mechanisms of DNA methylation in zebrafish
known as ddRADseq (Peterson et al., 2012), and does not rely on bisul-
fite conversion of genomic DNA, but still provides single base pair reso- As in mammals, the major proteins involved in DNA methylation in
lution of methylated sites. In EpiRADseq genomic DNA is first doubled- fishes are the DNA methyltransferases (DNMTs). The zebrafish genome
digested with two restriction enzymes that recognize different se- contains 8 DNA methyltransferase orthologs: one dnmt1 ortholog, one
quences, one of which is methylation insensitive (such as PstI) and dnmt2 ortholog, two dnmt3a orthologs, and four dnmt3b orthologs, al-
one of which is methylation-sensitive (such as HpaII). Because the though they lack an ortholog of the mammalian dnmt3L isoform. The
methylation-sensitive restriction enzyme will only cut un-methylated teleosts are known to have undergone a whole-genome duplication rel-
DNA, sites that are methylated will be under-represented in the ative to mammals (Jaillon et al., 2004), thus zebrafish might be expected
resulting sequences. Therefore, methylation state can be inferred by to have two copies of each mammalian Dnmt family member. This sug-
comparing the frequencies at which loci are sampled. As a result, this gests that the duplicates of dnmt1 and dnmt2 were lost during the
technique only provides information about methylation state at the en- rediploidization following the whole genome-duplication event, and
zyme cut-sites, and thus provides information at a reduced genome- that dnmt3b has undergone an additional round or rounds of duplica-
coverage relative to RRBS. EpiRADseq can be applied in the absence of tion in zebrafish following the teleost whole-genome duplication.
a genome sequence, although the downstream bioinformatics is some- The complex evolutionary history of the dnmt3 gene family has re-
what more challenging in this case. Proof of principle of the utility of this sulted in challenges in the naming conventions for these genes, and
method, even in the absence of genomic information, has been demon- there are several proposed names for the zebrafish dnmt3 genes, as
strated using a water flea, Daphnia ambigua (Schield et al., 2015), but to they were originally named without reference to phylogenetic relation-
date the method has not yet been applied in other organisms. ships (Shimoda et al., 2005). Here, we follow the new phylogenetic
The techniques outlined above are all designed to detect methylated naming convention suggested by Campos et al. (2012). dnmt3a1 and
cytosines (5mC). A variety of other approaches are available for the dnmt3a2 in zebrafish (also referred to as dnmt6 and dnmt8) are most
detection of demethylation intermediates such as 5hmC and 5fC. similar to the mammalian dnmt3a gene. dnmt3a1 and dnmt3a2 in

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
6 D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx

zebrafish share a generally similar developmental expression pattern DNA is hyper-methylated (95%) relative to oocyte DNA (75%). Follow-
with the mammalian dnmt3a, further supporting the phylogenetic rela- ing fertilization, 16-cell embryo DNA methylation levels are intermedi-
tionships among these genes (Smith et al., 2011). The two zebrafish iso- ate to those of sperm and oocytes, but whether this is the result of
forms differ in length at the 5′ end by 150 amino acids, whereas in erasure is unclear (Jiang et al., 2013; Potok et al., 2013). The lack of clar-
mouse the single dnmt3a gene is expressed as two isoforms, one short ity regarding the level of erasure of methylation may be due, at least in
and one long (Goll and Halpern, 2011). part, to the asynchronous development of zebrafish oocytes (Kimmel
The zebrafish dnmt3b1, dnmt3b2, dnmt3b3, and dnmt3b4 (also re- et al., 1995) and the associated difficulty of obtaining enough develop-
ferred to as dnmt4 dnmt7 dnmt3, and dnmt5 respectively) are most sim- mentally synchronized embryos to obtain the quantity of genomic
ilar to the mammalian dnmt3b gene, and share a similar developmental DNA necessary for this type of analysis. Perhaps using a fish species in
expression pattern, suggesting that they are related isoforms (Smith which more synchronous fertilization can be achieved would allow for
et al., 2011). The fact that zebrafish have four dnmt3b isoforms suggests a more detailed analysis of whole genome DNA methylation levels ear-
duplications in addition to the teleost whole-genome duplication in this lier in development.
gene for zebrafish. The current model for the evolution of the zebrafish After the 16 cell stage, whole-embryo DNA methylation levels
dnmt3b genes (Campos et al., 2012) is that the teleost whole-genome increase at each cell division, approaching the methylation levels
duplication produced dnmt3b2 and another dnmt3b paralogue that observed in sperm DNA by the time the embryo reaches gastrulation.
then went through two consecutive duplications that resulted in It has been proposed that patterns of DNA methylation in developing
dnmt3b1, dnmt3b3 and dnmt3b4. Interestingly, the Threespine Stickle- embryos are reset to the patterns observed in sperm. However, parthe-
back genome contains five dnmt3 genes (two dnmt3a homologs and nogenic fertilization of zebrafish oocytes results in the same whole-
three dnmt3b homologs). This suggests that at least one of the duplica- genome DNA methylation patterns as traditional fertilization (Potok
tion events that resulted in the four zebrafish dmnt3b isoforms is et al., 2013), suggesting that components within the oocyte provide all
lineage-specific. of the information required to establish methylation patterns in the off-
The functions of the various dnmt genes in zebrafish remain poorly spring. It is possible that the demethylated state of the oocyte is indica-
understood. Knockdown of dnmt1 has been shown to result in changes tive of a more totipotent state of the oocyte compared to the more
in methylation patterns and developmental abnormalities (Anderson differentiated state of the embryo, with increased whole genome meth-
et al., 2009; Rhee et al., 2012). Expression of dnmt3 genes has been ylation levels. Alternatively, piRNAs could survive the UV irradiation
shown to vary during development and can be affected by developmental treatment of sperm DNA prior to gynogenic fertilization acting as the
temperature, with a unique pattern for each isoform (Campos et al., primary regulators of DNA methylation reprogramming to a paternal
2012). The lack of a dnmt3L gene in zebrafish is consistent with the re- pattern (Potok et al., 2013). Whether the changes in methylation levels
duced importance of imprinting in fishes. For example, in contrast to and patterns during development are conserved across fishes or are
mammals, homozygous diploid clones are viable in fishes demonstrating unique to zebrafish remains relatively unexplored.
that imprinting must be fundamentally different in fishes compared to
mammals (Streisinger et al., 1981). DNMT3L is also absent from amphib- 4. Studies of DNA methylation in marine fishes
ian and bird genomes, suggesting that this gene may have arisen in mam-
mals along with the evolution of X-inactivation (Yokomine et al., 2006). Although DNA methylation has been studied for several decades in
In addition to the presence of DNA methyltransferases, the zebrafish zebrafish (e.g. Macleod et al., 1999), much less is known about DNA
genome also contains many of the other genes known to be involved methylation in marine fishes. Database searches for “DNA methylation”
DNA methylation in mammals such as the CXXC finger protein 1 (cfp1), and “fish” yielded less than 30 studies in any marine or diadromous fish
methyl binding proteins mecp2 and mbd2, the Piwi-related proteins species (Table S1). The earliest studies that include data on marine fish-
ziwi and zili, and the dnmt1 cofactor uhrf1 which is required for the main- es focussed on the identification of DNA methylation in fish genomes
tenance of DNA methylation patterns (Goll and Halpern, 2011). and/or broad-scale taxonomic comparisons (Bird and Taggart, 1980;
Kato, 1996; Vanyushin et al., 1970, 1973). In addition, an early compar-
3.2. Patterns of DNA methylation in zebrafish ative study using medaka (Oryzias latipes) and the Atlantic Killifish
(Fundulus heteroclitus) examined levels of methylation of an introduced
As is the case for our understanding of the mechanisms of DNA transgene (Winn et al., 1995), and demonstrated that this transgenic
methylation in fish, current studies of patterns of DNA methylation DNA was methylated. More recent studies have focussed on changes
are dominated by work in zebrafish. Overall levels of DNA methylation in DNA methylation in response to environmental factors or during
appear to be similar among vertebrates, with 80% of CpG dinucleotides development.
in the larval zebrafish genome being methylated compared to about 74%
in the mouse (Feng et al., 2010). Similarly, as is the case for other verte- 4.1. Environmental contaminants
brates, zebrafish genomes have prominent unmethylated CpG islands
located near the transcription start sites of actively transcribed genes. Some of the earliest studies of changes in DNA methylation associat-
Differential methylation of distal enhancer elements has also been ed with environmental factors in marine fishes examined the effect of
shown to play a biologically important role in zebrafish (Lee et al., environmental toxins (Aniagu et al., 2008; Timme-Laragy et al., 2005;
2015). The de-methylation intermediate 5hmC has also been observed Wang et al., 2009), and toxicology has continued to be an important
in zebrafish, with the highest levels being observed in brain tissue focus of DNA methylation studies in fishes (Pierron et al., 2014). These
(Kamstra et al., 2015). Non-CpG methylation, such as CpHpG and studies span a range of organisms, pollutants, and methods. The first
CpHpH, has also been detected in zebrafish but at low levels (2.13% in toxicological study of DNA methylation in a marine fish used single-
embryos) and these sites are evenly distributed throughout the ge- locus bisulfite sequencing to examine DNA methylation in the cyp1a
nome. By comparison, non-CpG methylation in mouse embryos is promoter of the Atlantic Killifish (F. heteroclitus) from creosote contam-
0.59% (Feng et al., 2010). inated and reference sites, and found no detectable DNA methylation in
In mammals, methylation patterns are thought to be globally erased the cyp1a promoter region (Timme-Laragy et al., 2005). This study illus-
and reset soon after fertilization. Whether or not this occurs in zebrafish trates one of the risks of using candidate approaches to studying DNA
is not clear. The earliest studies of this phenomenon in zebrafish yielded methylation. These authors focussed on the proximal promoter region
contradictory results (Macleod et al., 1999; Mhanni and McGowan, of an important candidate gene in the detoxification pathway for organ-
2004), whereas the most recent work suggests that intermediate levels ic contaminants. The promoter of this gene was known to be CpG-rich,
of erasure occur (Jiang et al., 2013; Potok et al., 2013). Zebrafish sperm and thus contained many potential methylation sites. However, cyp1a

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx 7

is expressed at detectable levels under control conditions in killifish tis- phenotypes. These results also illustrate the important point that al-
sues, and is induced by contaminant exposure (Meyer et al., 2003), though proximal promoter regions tend to be hypomethylated relative
which suggests that it is an actively transcribed gene under a variety to other regions of the genome, changes in methylation status in the
of environmental conditions. CpG rich regions (CpG islands) located promoter at specific loci can be important.
immediately upstream of the transcription start sites of actively tran-
scribed genes typically have low levels of methylation, and may not un- 4.3. Development and growth
dergo dynamic changes in DNA methylation, which may not make them
ideal candidate regions to explore without some a priori indication of It has been well documented in mammalian systems that whole-
differential methylation. genome DNA methylation patterns undergo dramatic changes during
Several studies have demonstrated that whole-genome DNA development and cellular differentiation. As briefly summarized
methylation levels change in response to toxicant exposure in ways above, much of what is known about changes in methylation patterns
that can be detected using low-resolution approaches (Aniagu et al., during development is derived from work in zebrafish (e.g. Jiang et al.,
2008; Mirbahai et al., 2011; Pierron et al., 2014; Wang et al., 2009). In 2013; Potok et al., 2013). However, two studies have examined changes
Threespine Stickleback (Gasterosteus aculeatus) there were significant in DNA methylation with development in marine fishes. For example,
changes in whole-genome DNA methylation levels in gonad tissue fol- early maturation in Atlantic Salmon (Salmo salar) is associated with
lowing environmental exposure to estradiol (E2) as detected by HPLC, changes in DNA methylation patterns as detected using MS-AFLP
but no changes in response to hexa-bromo-cyclododecane (HBCD) (Morán and Pérez-Figueroa, 2011). Observed differences DNA methyla-
exposure in liver (Aniagu et al., 2008). HPLC-based methods were tion associated with early maturation were consistent across two popu-
also used to examine the effects of contaminant exposure in a rock- lations in testis tissue, whereas methylation differences in the brain
fish (Sebastiscus marmoratus). In this case, there were significant associated with early maturation were observed in only one of two pop-
concentration-dependent changes in whole-genome DNA methyla- ulations studied. In contrast, no differences in DNA methylation pat-
tion levels in liver tissue following environmental exposure to tribu- terns associated with early maturation were observed in liver tissue.
tyltin and triphenyltin in Wang et al. (2009). Results from these The role of DNA methylation in metamorphosis has been explored in
studies illustrate both the potential and the limitations of low- Sea Lamprey (Petromyzon marinus). Larval lampreys (ammocoetes) are
resolution DNA methylation assays in that important DNA methyla- filter feeders that burrow into streambeds. During the larval stage, lam-
tion changes could be occurring at targeted locations that would not preys go through a series of metamorphoses until the final transforma-
be detected at the whole-genome level, making negative results diffi- tion into a tissue-feeding adult that migrates to the marine environment
cult to interpret. (Manzon et al., 2015). There are substantial differences in whole-
Studies using low-resolution whole-genome assays followed by genome methylation levels in the muscle tissue of larval and adult lam-
intermediate-resolution methods and targeted or whole-genome se- preys as detected by MS-AFLP (Covelo-Soto et al., 2015a, 2015b). These
quencing to detect differentially methylated loci have yielded additional authors then sequenced eleven of the differentially methylated bands,
insights into the effects of environmental toxicants on the epigenome. and were able to identify six of these bands, including several hox
For example, exposure of European Eel (Anguilla anguilla) to environ- genes, which are known to be involved in metamorphosis.
mental cadmium resulted in a dose-dependent increase in whole- Changes in DNA methylation may also play a role in the regulation of
genome methylation levels using a low-resolution ELISA method growth during development. A study conducted by Si et al. (2015) in
(Pierron et al., 2014). These authors then identified multiple genes Half-Smooth Tongue Sole (Cynoglossus semilaevis) used bisulfite treat-
that were differentially methylated using MS-AFLP. Similarly, tumori- ment followed by sequencing of a specific locus to examine the effect
genesis has been shown to have an epigenomic component in the Com- of genotype on methylation level and gene expression of pituitary ade-
mon Dab (Limanda limanda) (Mirbahai et al., 2011). These authors first nylate cyclase activating polypeptide (pacap). In this study, the authors
used an HPLC-based approach to detect a reduction in whole-genome examine three pacap genotypes that are known to differ in growth and
methylation levels in tumor tissues. They then used MeDIP-seq to spe- show that DNA methylation levels and gene expression vary significant-
cifically identify differentially methylated genomic regions at single ly between the three pacap alleles suggesting that there may be an epi-
base-pair resolution, which were then correlated with changes in gene genetic component to genotype-mediated differences in growth rates.
expression. Many of the differentially methylated and expressed genes Changes in DNA methylation may also play a role in establishing var-
were in pathways known to be associated with tumorigenesis in other iation in life-history traits. For example, a recent study (Baerwald et al.,
organisms. 2015) used RRBS to examine variation in methylation patterns associat-
ed with variation in the timing of smoltification and the propensity to
4.2. Temperature migrate in Rainbow Trout (Oncorhynchus mykiss). This study took ad-
vantage of the existence of double-haploid clonal lines of resident and
Temperature has profound effects on the biology of fishes, and there anadromous Rainbow Trout to produce double-haploid F2 siblings
have been a few studies investigating its effects on the epigenome of with varying genomic combinations of parental migratory and resident
marine fishes (Campos et al., 2013; Shao et al., 2014; Varriale and DNA. These F2s were used to identify fifty-seven differentially methylat-
Bernardi, 2006). A particularly intriguing study (Varriale and Bernardi, ed regions that correlated with smoltification, including multiple genes
2006) found that whole-genome methylation levels (as assessed using in pathways associated with circadian rhythms. Over half of the differ-
RP-HPLC) were inversely correlated with temperature, with the ge- entially methylated regions were in or near CpG islands associated
nomes of fishes from cooler habitats exhibiting higher levels of with proximal promoters, suggesting that they could be functionally
whole-genome DNA methylation compared to fish from warmer important and affect gene expression. These data demonstrate the
habitats. Several subsequent studies have also identified tempera- power of RRBS for the detection of differential methylation patterns as-
ture as an important factor that may modify methylation levels in sociated with ecologically important traits in marine fishes.
marine fishes (Campos et al., 2013; Shao et al., 2014). For example,
temperature has been shown to affect methylation levels in the pro- 4.4. Sex determination
moter of the myogenin gene in Senegalese Sole (Solea senegalensis), as
detected using candidate-gene bisulfite sequencing (Campos et al., Fishes provide an excellent model for the study of sex-determination
2013). These changes were correlated with changes in myogenin mechanisms, as they exhibit a wide range of sex-determination strategies,
expression and muscle cellularity, clearly demonstrating the linkages including both genetic sex-determination and environmental sex deter-
between changes in DNA methylation and important organismal mination (Devlin and Nagahama, 2002). While the mechanisms of sex

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
8 D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx

determination are beyond the scope of this review, the studies conducted previously been shown to differ between morphotypes. Because these
to date suggest an important role of epigenetics in sex-determination in analyses were performed on wild-caught individuals, it is not possible
several species. to determine whether the observed differences in methylation were a
The first study to associate changes in DNA methylation patterns cause or consequence of the morphological differences between the
with sex determination in a marine fish was conducted in the ecotypes or whether they may have been caused by environment-
European Sea Bass (Dicentrarchus labrax). In this study Navarro- dependent effects.
Martín et al. (2011) used a targeted bisulfite sequencing approach
to show that the methylation patterns of CpG locations in the pro- 4.6. Aquaculture
moter of the cyp19a gene are hypermethylated in male gonad tissue
compared to female gonad tissue. cyp19a is an aromatase gene that Artificial triploidization is commonly used in aquaculture settings to
converts androgen into estrogen. Hypermethylation in male gonad alter the growth and fertility of fishes (Piferrer et al., 2009). Using MS-
tissue is therefore consistent with reduced expression of cyp19a AFLP, Covelo-Soto et al. (2015a) measured whole genome DNA methyl-
and a reduction in the synthesis of estrogen in male gonad tissue. Inter- ation levels in the brain, gill, heart, liver, kidney and muscle tissue of
estingly, exposure of European Sea Bass larvae to elevated temperatures diploid and triploid Brown Trout, (Salmo trutta L.) to test whether
resulted in increased methylation of the cyp19a promoter and a mascu- changes to whole-genome DNA methylation levels are important for
linization of female gonad tissue. the maintenance of polyploidy in fish. Although they did not find signif-
The role of epigenetics in sex determination has also been investigat- icant differences between any of the diploid and triploid tissues at the
ed in the Half-Smooth Tongue Sole (C. semilaevis), which undergoes whole genome level there were intriguing patterns at a small number
environmentally induced changes in sex ratios following exposure to of individual loci. Thus, it is possible that compensatory epigenetic
elevated temperatures during development. This species has a ZZ/ZW mechanisms may still play a role in the proper genomic function of
sex determination system in which it has been previously demonstrated polyploidy in fish but that these changes could not be reliably detected
that male-specific expression of the Z-linked dmrt1 gene is associated at the level of resolution provided by MS-AFLP.
with male gonad development (Chen et al., 2014). Shao et al. (2014) Exposure to differing environments during early life has been
used WGBS to determine whole-genome DNA methylation patterns in shown to cause persistent changes in DNA methylation patterns in
male, female, and pseudo-male gonads. They found that genes associated mammals (Bird, 2002). These observations have motivated interest
with sex-determination pathways are differentially methylated between in examining whether the effects of hatchery rearing on perfor-
male and female gonad tissue. Specifically, the authors discovered that mance could be due to epigenetic effects (Taylor et al., 2010). No differ-
the dmrt1 gene has high levels of DNA methylation in female gonads, ences in MS-AFLP banding patterns were detected between returning
whereas ZZ males and ZW pseudo-males were hypomethylated at the wild and hatchery-reared adult steelhead Trout (O. mykiss). However,
dmrt1 locus suggesting that demethylation of dmrt1 is necessary for MS-AFLP is unable to resolve quantitative differences in the percent
male gonad development. Interestingly, although ZW pseudo-males can DNA methylation at any given site. Thus, it is impossible to rule out
be produced by exposing ZW females to high environmental tempera- the possibility differences in DNA methylation between wild and
tures, pseudo-males can also give rise to ZW pseudo-male offspring in hatchery-reared fish from these data.
the absence of a high-temperature environmental cue (Chen et al., The role of changes in DNA methylation has also been examined in
2014). This suggests that there may be a transgenerational epigenetic hatchery-reared Brown Trout (S. trutta L.) (Morán et al., 2013). Using
mechanism of sex determination in this species. MS-AFLP, these authors observed substantial differences in DNA
A study by Wen et al. (2014) measured DNA methylation and gene methylation patterns in the gill between fish held in fresh water and
expression levels of both cyp19a and dmrt1 in Japanese Flounder fish acclimated to seawater for 10–12 days. These data clearly show
(Paralichthys olivaceus). They found that high levels of dmrt1 expression that environmental salinity can have a profound effect on DNA methyl-
in male gonad tissue are correlated with low levels of DNA methylation ation in fish. In addition, these authors examined the role of the salt con-
in the dmrt1 promoter, whereas low levels of expression are correlated tent of the diet in altering DNA methylation levels, as high-salt diets
with high levels of DNA methylation. In contrast, the promoter of have been shown to be important in the transformation of the gill to
cyp19a was hypermethylated in testes compared to ovaries, which saltwater phenotype (Perry et al., 2006). There were transient changes
corresponded to lower levels of cyp19a mRNA in testis tissue compared in DNA methylation in the gills of fish fed a high-salt diet that made
to ovaries. Subsequent studies in Japanese Flounder have observed sim- their methylation patterns somewhat more similar to fish held in sea-
ilar patterns in which DNA methylation patterns of cyp19a1a and foxl2 water than those of fish in freshwater that were fed the control diet
are inversely correlated with mRNA expression levels during ovarian (in at least on one of the two principle component axes). These data
development (Si et al., 2016). Taken together, these studies demon- suggest that diet-induced epigenetic modifications may be important
strate that the methylation of the dmrt1 and cyp19a genes is important for the transformation of the gill during salinity acclimation.
for sex determination in a variety of fish species. Geay et al. (2012) used a candidate-locus approach to examine
the effects of diets differing in their levels of highly unsaturated
4.5. Morphological variation fatty acids (HUFA) on methylation levels in the promoter of the
Delta(6) desaturase (fads2) gene in European Sea Bass (D. labrax). The
There has been considerable interest in the potential role of epige- promoter of the fads2 gene was methylated at very low levels, and
netic mechanisms in causing intra-specific morphological variation there was no significant change in DNA methylation with diet. This il-
(Bonasio, 2015). There is a single study examining this question in a lustrates the general principle that the DNA methylation status of the
species with marine populations: the Threespine Stickleback (Smith promoters of highly expressed genes tends to be maintained at consti-
et al., 2015).The Threespine Stickleback species complex contains ma- tutively low levels. However, the expression of the fads2 gene was ob-
rine, anadromous, and freshwater ecotypes that differ in many morpho- served to increase in Sea Bass fed a low HUFA diet, illustrating that
logical and physiological characteristics, including variation in the changes in gene expression can occur in the absence of changes in
extent of bony lateral plates (Bell and Foster, 1994). Smith et al. methylation at the proximal promoter of a gene.
(2015) used RRBS to characterize whole-genome DNA methylation pat-
terns in muscle tissue from low plated and high plated stickleback 5. The future of epigenomics research in marine fishes
morphotypes from a single population. Using this method, a variety of
differentially methylated regions were identified, some of which were The studies presented here clearly demonstrate the potential for
associated with genes involved in biological processes that have epigenomic research in marine fishes. The continued development of

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx 9

methods that can be applied to non-model systems is making address- appear to differ in the extent to which methylation patterns are erased
ing these epigenomic questions increasingly tractable. As sequencing early in development, with current studies suggesting that fish retain a
technologies continue to develop and the cost of whole genome sequenc- higher proportion of methylated sites (Jiang et al., 2013; Potok et al.,
ing decreases, we expect that high-resolution epigenomic studies in ma- 2013). In mammals, the high degree of erasure of methylated sites has
rine fishes will become increasingly common. Below we provide some been used to argue against the role of DNA methylation in transmitting
concluding remarks regarding the utility of fish as model systems for transgenerational epigenetic information. This difference highlights the
epigenomic research, methodological considerations regarding applying potential importance of pursuing studies of transgenerational epigenet-
epigenomic techniques in marine fishes, and some particularly promising ic inheritance of DNA methylation patterns in fish. Fish genomes also
areas of research where epigenomic methods could be applied. contain additional DNA methyltransferase genes not present in the ge-
nomes of other organisms. Whether these genes facilitate a unique
5.1. Fish as model systems for epigenomics functional role of DNA methylation, and the evolution of this gene fam-
ily in fish remains to be studied.
Fish have many useful characteristics that make them attractive
model systems for addressing questions in evolutionary biology, embry- 5.3. Methodological considerations
ology, and physiology (Cossins and Crawford, 2005; Powers, 1989).
Many of these characteristics also make fish particularly suitable Throughout this review we have highlighted the particular strengths
model systems for epigenomic research (Kamstra et al., 2015). For and weaknesses associated with the various techniques that can be used
example, one of the key questions in epigenomics is to understand the to measure DNA methylation in marine fishes. Low-resolution tech-
transgenerational effects of environmental factors on organismal phe- niques such as ELISA-based detection of whole-genome methylation
notypes, and whether and how methylation patterns can be transmitted levels are easy to apply and can be useful for the identification of large-
across generations. In mammals, an environmentally-induced pheno- scale changes in methylation levels. However, most questions require
type must be maintained at least to the F3 generation to convincingly higher-resolution approaches. Although MS-AFLP provides only inter-
demonstrate a true epigenetic effect. This is because exposure of the mediate resolution, this technique can be fairly easily applied in any lab-
gestating female (F0) simultaneously exposes the developing embryo oratory that is equipped for molecular biology, and access to a reference
(F1) and the developing germ line of the embryo (F2). In contrast, in genome and sophisticated bioinformatics expertise is not required. As a
fish only the mother (FO) and her eggs (F1) are exposed. Thus, in fish result, MS-AFLP can be an appropriate choice for preliminary studies or
it is possible to conclusively demonstrate an epigenetic effect by the for laboratories that are beginning to address epigenomic questions.
F2 generation. This, in combination with the rapid generation time of MS-AFLP can be particularly useful for screening to identify candidate
many species of fish makes them a compelling model for demonstrating differentially methylated loci for further examination.
transgenerational effects. Another key question in epigenomics is the The current gold-standard for characterizing DNA methylation levels
role of early developmental environment on the phenotype of the at high resolution is the application of bisulfite sequencing, which can
offspring and the mechanisms underlying developmental plasticity. Be- be applied to single candidate loci, subsets of the whole genome (via
cause many fishes develop externally, it is relatively straightforward to RRBS), or as WGBS. Bisulfite sequencing of candidate loci is cost-
manipulate the environment during development, especially in com- effective and straightforward, but is critically dependent on the appropri-
parison to the challenges of these approaches in mammals. In addition, ate choice of candidate gene. This can be difficult in the absence of prior
many species have large clutch sizes, which allows for full-sib compar- information suggesting that a genomic region is likely to be differentially
isons by using a split-clutch design in which multiple members of the methylated. Application of MS-AFLP to identify promising candidates
same family are exposed to different treatments. Similarly, a split clutch followed by bisulfite sequencing may provide a good balance of the
design can be used to generate multiple half-sib families, which could strengths of these two techniques. In contrast, WGBS provides complete
help to disentangle paternal and maternal effects. information about the methylation status of every cytosine in the ge-
In addition, many of the unique biological features of fish make them nome, but this technique remains prohibitively expensive for many
an interesting model of the investigation of the role of epigenomics in a studies. RRBS provides a good alternative that captures a large subset
variety of fundamental biological processes. For example, fish demon- of the methylated regions of the genome at a more reasonable cost.
strate a wide range of sex-determining mechanisms, including both ge- Similarly, the recently developed non-bisulfite-dependent method of
netic and environmental sex-determination, but the underlying EpiRADseq represents another approach to increase the tractability of
mechanisms remain poorly understood (Devlin and Nagahama, 2002). high-resolution epigenomic techniques. The development of these
Recent studies suggest that there may be an epigenomic component to reduced-representation sequencing techniques opens exciting new
sex-determination in some species (Navarro-Martín et al., 2011; Shao possibilities for studies of epigenomics in marine fishes.
et al., 2014), highlighting the potential for research in this area. Similarly, One key challenge for epigenomic studies of DNA methylation is that
polyploidization is common in fish, and induction of triploidy is routinely these studies examine correlations between changes in methylation
used in aquaculture. Thus fish provide a vertebrate model for the study of patterns and phenotypic changes, which does not directly establish cau-
the role of epigenomics in dosage compensation in polyploids (Pala et al., sation. To demonstrate that changes in DNA methylation levels are
2008). causing the observed changes in phenotype requires direct functional
assays such as mutagenesis of the putatively important methylated
5.2. Unique features of epigenomics in fishes sites. New technologies such as the CRISPR/Cas9 endonuclease system
(Hsu et al., 2014) should allow these causal links to be established.
Although fish and mammals share many common features of their This technique has been proposed to allow targeted genetic manipula-
methylation patterns, which are distinct from the patterns in other tion of essentially any species and has already been successfully applied
major taxonomic groups such as the invertebrates, fungi and plants in a marine fish species (Aluru et al., 2015).
(Zemach et al., 2010), there are also fundamental differences in methyla-
tion patterns and mechanisms between fish and mammals (Goll and 5.4. Research on epigenomics in aquaculture
Halpern, 2011). For example, genomic imprinting is thought to be absent
in fish, and they lack the dnmt3L gene that is involved in mammalian im- Studies of diet (Geay et al., 2012; Morán et al., 2013), the effects of
printing. However, recent data suggest that sex-specific methylation may hatchery rearing (Taylor et al., 2010), triploidization (Covelo-Soto et al.,
occur in fish (Jiang et al., 2013; Potok et al., 2013; Shao et al., 2014), but 2015a, 2015b), and early maturation (Morán and Pérez-Figueroa, 2011)
the mechanisms involved remain unknown. Similarly, fish and mammals demonstrate the enormous potential for the application of epigenomics

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
10 D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx

in aquaculture. Many other aspects of aquaculture practice may have References


epigenomic effects including disease exposure, stress, density, water
quality, none of which have yet been investigated. Determining whether Aberg, K.A., McClay, J.L., Nerella, S., Xie, L.Y., Clark, S.L., Hudson, A.D., Bukszár, J., Adkins, D.,
Consortium, S.S., Hultman, C.M., Sullivan, P.F., Magnusson, P.K., Van Den Oord, E.J.,
exposure to such conditions has persistent effects the phenotype of 2012. MBD-seq as a cost-effective approach for methylome-wide association studies:
cultured fishes could have important implications for improving the demonstration in 1500 case–control samples. Epigenomics 4, 605–621. http://dx.doi.
yield or efficiency of aquaculture operations. org/10.2217/epi.12.59.
Adusumalli, S., Mohd Omar, M.F., Soong, R., Benoukraf, T., 2015. Methodological aspects of
whole-genome bisulfite sequencing analysis. Brief. Bioinform. 16, 369–379. http://dx.
5.5. Research in environmental biology and evolution doi.org/10.1093/bib/bbu016.
Aluru, N., Karchner, S.I., Franks, D.G., Nacci, D., Champlin, D., Hahn, M.E., 2015. Targeted
mutagenesis of aryl hydrocarbon receptor 2a and 2b genes in Atlantic killifish
Epigenomic changes have the potential to play an important role in (Fundulus heteroclitus). Aquat. Toxicol. 158, 192–201. http://dx.doi.org/10.1016/j.
the response of marine fishes to environmental change, and may influ- aquatox.2014.11.016.
ence adaptation to new environments (Franks and Hoffmann, 2012). Alvarado, S., Rajakumar, R., Abouheif, E., Szyf, M., 2015. Epigenetic variation in the Egfr
gene generates quantitative variation in a complex trait in ants. Nat. Commun. 6,
These questions are of increasing importance in the context of global 6513. http://dx.doi.org/10.1038/ncomms7513.
environmental change and other human impacts on the marine envi- Anderson, R.M., Bosch, J.A., Goll, M.G., Hesselson, D., Dong, P.D.S., Shin, D., Chi, N.C., Shin,
ronment. For example, as discussed above, multiple studies have con- C.H., Schlegel, A., Halpern, M., Stainier, D.Y.R., 2009. Loss of Dnmt1 catalytic activity
reveals multiple roles for DNA methylation during pancreas development and regen-
vincingly shown that the DNA methylation patterns of marine fishes
eration. Dev. Biol. 334, 213–223. http://dx.doi.org/10.1016/j.ydbio.2009.07.017.
change following exposure to environmental toxicants, highlighting Aniagu, S.O., Williams, T.D., Allen, Y., Katsiadaki, I., Chipman, J.K., 2008. Global genomic
an underappreciated impact of environmental toxicants on organismal methylation levels in the liver and gonads of the three-spine stickleback
biology. However, many of the studies to date have utilized low- (Gasterosteus aculeatus) after exposure to hexabromocyclododecane and 17-β
oestradiol. Environ. Int. 34, 310–317. http://dx.doi.org/10.1016/j.envint.2007.03.009.
resolution methods, and thus the specific biological processes and path- Baerwald, M.R., Meek, M.H., Stephens, M.R., Nagarajan, R.P., Goodbla, A.M., Tomalty, K.M.,
ways that are being affected remain unknown. Continued investigation Thorgaard, G.H., May, B., Nichols, K.M., 2015. Migration-related phenotypic diver-
into the environmental effects on DNA methylation using higher resolu- gence is associated with epigenetic modifications in rainbow trout. Mol. Ecol.
http://dx.doi.org/10.1111/mec.13231.
tion methods and assessment of the persistent consequences of these Bannister, A.J., Kouzarides, T., 2011. Regulation of chromatin by histone modifications. Cell
changes is urgently needed to fully appreciate and predict the effects Res. 21, 381–395. http://dx.doi.org/10.1038/cr.2011.22.
of toxicant exposure and other environmental changes at the individual, Bell, M.A., Foster, S.A., 1994. The Evolutionary Biology of the Threespine Stickleback. Ox-
ford University Press, New York.
population, and ecological levels. A long-term goal of research efforts in Bewick, A.J., Hofmeister, B.T., Lee, K., Zhang, X., Hall, D.W., Schmitz, R.J., 2015. FASTmC: a
this area would be to establish DNA methylation-based assays as an suite of predictive models for non-reference-based estimations of DNA methylation.
indicator of the effects of anthropogenic activities, and incorporate G3; Genes Genomes Genet. 49, 2–15. http://dx.doi.org/10.1534/g3.115.025668.
Bird, A., 2002. DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21.
this information into the risk-assessment process (Ray et al., 2014). http://dx.doi.org/10.1101/gad.947102.
Multiple studies have shown that epigenetic mechanisms may be Bird, A.P., Taggart, M.H., 1980. Variable patterns of total DNA and rDNA methylation in an-
important in the colonization of new environments and the process of imals. Nucleic Acids Res. 8, 1485–1497. http://dx.doi.org/10.1093/nar/8.7.1485.
Bock, C., 2012. Analysing and interpreting DNA methylation data. Nat. Rev. Genet. 13,
adaptation to environmental change (e.g. Schrey et al., 2012). For exam-
705–719. http://dx.doi.org/10.1038/nrg3273.
ple, variation in DNA methylation patterns could be an important source Bonasio, R., 2015. The expanding epigenetic landscape of non-model organisms. J. Exp.
of inter-individual and inter-population variation on which natural Biol. 218, 114–122. http://dx.doi.org/10.1242/jeb.110809.
selection could act (Massicotte et al., 2011). At the present time these Booth, M.J., Branco, M.R., Ficz, G., Oxley, D., Krueger, F., Reik, W., Balasubramanian, S., 2012.
Quantitative sequencing of 5-methylcytosine and 5-hydroxymethylcytosine at single-
questions have not been addressed in any marine fish species, but base resolution. Science 336, 934–937. http://dx.doi.org/10.1126/science.1220671.
there is substantial scope for experimentation, and addressing these Booth, M.J., Marsico, G., Bachman, M., Beraldi, D., Balasubramanian, S., 2014. Quantitative
questions may be particularly important as we attempt to predict the sequencing of 5-formylcytosine in DNA at single-base resolution. Nat. Chem. 6,
435–440. http://dx.doi.org/10.1038/nchem.1893.
responses of commercially important marine fishes to climate change Borgel, J., Guibert, S., Li, Y., Chiba, H., Schübeler, D., Sasaki, H., Forné, T., Weber, M., 2010.
(Franks and Hoffmann, 2012). Targets and dynamics of promoter DNA methylation during early mouse develop-
ment. Nat. Genet. 42, 1093–1100. http://dx.doi.org/10.1038/ng.708.
Bossdorf, O., Richards, C.L., Pigliucci, M., 2008. Epigenetics for ecologists. Ecol. Lett. 11,
6. Conclusions 106–115. http://dx.doi.org/10.1111/j.1461-0248.2007.01130.x.
Bostick, M., Kim, J.K., Estève, P.-O., Clark, A., Pradhan, S., Jacobsen, S.E., 2007. UHRF1 plays
The studies outlined above clearly demonstrate the pervasive effects a role in maintaining DNA methylation in mammalian cells. Science 317, 1760–1764.
http://dx.doi.org/10.1126/science.1147939.
of the environment on the epigenomes of marine fishes, and indicate Branco, M.R., Ficz, G., Reik, W., 2011. Uncovering the role of 5-hydroxymethylcytosine in
the potential for ongoing epigenomic research to provide an increased the epigenome. Nat. Rev. Genet. 13, 7–13. http://dx.doi.org/10.1038/nrg3080.
understanding of the responses of fishes to environmental change. Campos, C., Valente, L.M.P., Conceição, L.E.C., Engrola, S., Fernandes, J.M.O., 2013. Temper-
ature affects methylation of the myogenin putative promoter, its expression and
The integration of the various “omics” approaches, including tran-
muscle cellularity in Senegalese sole larvae. Epigenetics 8. http://dx.doi.org/10.
scriptomics, proteomics, genomics, and epigenomics holds particular 4161/epi.24178.
promise in linking genotype, phenotype and environment and dissect- Campos, C., Valente, L.M.P., Fernandes, J.M.O., 2012. Molecular evolution of zebrafish
ing the mechanisms underlying genotype by environment interactions. dnmt3 genes and thermal plasticity of their expression during embryonic develop-
ment. Gene 500, 93–100. http://dx.doi.org/10.1016/j.gene.2012.03.041.
Studies of these phenomena in marine fishes will provide new insights Chen, P.-Y., Cokus, S.J., Pellegrini, M., 2010. BS Seeker: precise mapping for bisulfite se-
that will increase our understanding of epigenomic phenomena in nat- quencing. BMC Bioinf. 11, 203. http://dx.doi.org/10.1186/1471-2105-11-203.
ural environments. Chen, S., Zhang, G., Shao, C., Huang, Q., Liu, G., Zhang, P., Song, W., An, N., Chalopin, D.,
Volff, J.-N., Hong, Y., Li, Q., Sha, Z., Zhou, H., Xie, M., Yu, Q., Liu, Y., Xiang, H., Wang,
Supplementary data to this article can be found online at http://dx. N., Wu, K., Yang, C., Zhou, Q., Liao, X., Yang, L., Hu, Q., Zhang, J., Meng, L., Jin, L., Tian,
doi.org/10.1016/j.margen.2016.01.004. Y., Lian, J., Yang, J., Miao, G., Liu, S., Liang, Z., Yan, F., Li, Y., Sun, B., Zhang, H., Zhang,
J., Zhu, Y., Du, M., Zhao, Y., Schartl, M., Tang, Q., Wang, J., 2014. Whole-genome se-
quence of a flatfish provides insights into ZW sex chromosome evolution and adapta-
Acknowledgments tion to a benthic lifestyle. Nat. Genet. 46, 253–260. http://dx.doi.org/10.1038/ng.2890.
Clément, G., Benhattar, J., 2005. A methylation sensitive dot blot assay (MS-DBA) for the
This work was supported through a Natural Sciences and Engineer- quantitative analysis of DNA methylation in clinical samples. J. Clin. Pathol. 58,
155–158. http://dx.doi.org/10.1136/jcp.2004.021147.
ing Research Council of Canada (NSERC) Discovery Grant to PMS and a Cossins, A., 2006. Post-genomic approaches to understanding the mechanisms of environ-
University of British Columbia PhD Fellowship to DCHM. Thanks to the mentally induced phenotypic plasticity. J. Exp. Biol. 209, 2328–2336. http://dx.doi.
participants in the symposium “Genome-Powered Perspectives in org/10.1242/jeb.02256.
Cossins, A.R., Crawford, D.L., 2005. Fish as models for environmental genomics. Nat. Rev.
Integrative Physiology and Evolutionary Biology” for their insightful
Genet. 6, 324–333. http://dx.doi.org/10.1038/nrg1590.
discussions and to the Society for Experimental Biology for supporting Costa, F.F., 2008. Non-coding RNAs, epigenetics and complexity. Gene 410, 9–17. http://
this session. dx.doi.org/10.1016/j.gene.2007.12.008.

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx 11

Covelo-Soto, L., Leunda, P.M., Pérez-Figueroa, A., Morán, P., 2015a. Genome-wide methyl- Kalujnaia, S., McWilliam, I.S., Zaguinaiko, V.A., Feilen, A.L., Nicholson, J., Hazon, N., Cutler,
ation study of diploid and triploid brown trout (Salmo trutta L.). Anim. Genet. 46, C.P., Balment, R.J., Cossins, A.R., Hughes, M., Cramb, G., 2007. Salinity adaptation and
280–288. http://dx.doi.org/10.1111/age.12287. gene profiling analysis in the European eel (Anguilla anguilla) using microarray tech-
Covelo-Soto, L., Saura, M., Morán, P., 2015b. Does DNA methylation regulate metamor- nology. Gen. Comp. Endocrinol. 152, 274–280. http://dx.doi.org/10.1016/j.ygcen.
phosis? the case of the sea lamprey (Petromyzon marinus) as an example. Comp. 2006.12.025.
Biochem. Physiol., Part B Biochem. Mol. Biol. 185, 42–46. http://dx.doi.org/10.1016/ Kamstra, J.H., Aleström, P., Kooter, J.M., Legler, J., 2015. Zebrafish as a model to study the
j.cbpb.2015.03.007. role of DNA methylation in environmental toxicology. Environ. Sci. Pollut. Res. 22,
Deans, C., Maggert, K.A., 2015. What do you mean, “epigenetic”? Genetics 199, 887–896. 16262–16276. http://dx.doi.org/10.1007/s11356-014-3466-7.
http://dx.doi.org/10.1534/genetics.114.173492. Kato, M., 1996. Methylation status of Sillago japonica satellite DNA examined by bisulfite
Deaton, A.M., Bird, A., 2011. CpG islands and the regulation of transcription. Genes Dev. modification. Mol. Biol. Rep. 23, 123–127. http://dx.doi.org/10.1007/BF00424438.
25, 1010–1022. http://dx.doi.org/10.1101/gad.2037511. Kimmel, C.B., Ballard, W.W., Kimmel, S.R., Ullmann, B., Schilling, T.F., 1995. Stages of em-
Devlin, R.H., Nagahama, Y., 2002. Sex determination and sex differentiation in fish: an bryonic development of the zebrafish. Dev. Dyn. 203, 253–310. http://dx.doi.org/10.
overview of genetic, physiological, and environmental influences. Aquaculture 208, 1002/aja.1002030302.
191–364. http://dx.doi.org/10.1016/S0044-8486(02)00057-1. Klose, R.J., Bird, A.P., 2006. Genomic DNA methylation: the mark and its mediators. Trends
Dupont, J.-M., Tost, J., Jammes, H., Gut, I.G., 2004. De novo quantitative bisulfite sequenc- Biochem. Sci. 31, 89–97. http://dx.doi.org/10.1016/j.tibs.2005.12.008.
ing using the pyrosequencing technology. Anal. Biochem. 333, 119–127. http://dx. Kroeze, L.I., van der Reijden, B.A., Jansen, J.H., 2015. 5-Hydroxymethylcytosine: an epige-
doi.org/10.1016/j.ab.2004.05.007. netic mark frequently deregulated in cancer. Biochim. Biophys. Acta, Rev. Cancer
Eads, C.A., Danenberg, K.D., Kawakami, K., Saltz, L.B., Blake, C., Shibata, D., Danenberg, P.V., 1855, 144–154. http://dx.doi.org/10.1016/j.bbcan.2015.01.001.
Laird, P.W., 2000. MethyLight: a high-throughput assay to measure DNA methylation. Kucharski, R., Maleszka, J., Foret, S., Maleszka, R., 2008. Nutritional control of reproductive
Nucleic Acids Res. 28, E32. http://dx.doi.org/10.1093/nar/28.8.e32. status in honeybees via DNA methylation. Science 319, 1827–1830. http://dx.doi.org/
Feng, S., Cokus, S.J., Zhang, X., Chen, P.-Y., Bostick, M., Goll, M.G., Hetzel, J., Jain, J., Strauss, 10.1126/science.1153069.
S.H., Halpern, M.E., Ukomadu, C., Sadler, K.C., Pradhan, S., Pellegrini, M., Jacobsen, S.E., Kuo, K.C., McCune, R.A., Gehrke, C.W., Midgett, R., Ehrlich, M., 1980. Quantitative
2010. Conservation and divergence of methylation patterning in plants and animals. reversed-phase high performance liquid chromatographic determination of major
Proc. Natl. Acad. Sci. U. S. A. 107, 8689–8694. http://dx.doi.org/10.1073/pnas. and modified deoxyribonucleosides in DNA. Nucleic Acids Res. 8, 4763–4776.
1002720107. http://dx.doi.org/10.1093/nar/8.20.4763.
Flanagan, J.M., Popendikyte, V., Pozdniakovaite, N., Sobolev, M., Assadzadeh, A., Laird, P.W., 2010. Principles and challenges of genomewide DNA methylation analysis.
Schumacher, A., Zangeneh, M., Lau, L., Virtanen, C., Wang, S.-C., Petronis, A., 2006. Nat. Rev. Genet. 11, 191–203. http://dx.doi.org/10.1038/nrg2732.
Intra- and interindividual epigenetic variation in human germ cells. Am. J. Hum. Laurent, L., Wong, E., Li, G., Hodges, E., Smith, A.D., Kendall, J., Irizarry, R.A., Ladd-acosta, C.,
Genet. 79, 67–84. http://dx.doi.org/10.1086/504729. Carvalho, B., Edwards, J.R., Donnell, A.H.O., Rollins, R.A., Huynh, T., Tsirigos, A., Ong,
Flores, K.B., Wolschin, F., Amdam, G.V., 2013. The role of methylation of DNA in environ- C.T., Low, H.M., Wing, K., Sung, K., 2010. Dynamic changes in the human methylome
mental adaptation. Integr. Comp. Biol. 53, 359–372. http://dx.doi.org/10.1093/icb/ during differentiation Dynamic changes in the human methylome during differenti-
ict019. ation. Genome Res. 20, 320–331. http://dx.doi.org/10.1101/gr.101907.109.
Franks, S.J., Hoffmann, A.A., 2012. Genetics of climate change adaptation. Annu. Rev. Law, J.A., Jacobsen, S.E., 2010. Establishing, maintaining and modifying DNA methylation
Genet. 46, 185–208. http://dx.doi.org/10.1146/annurev-genet-110711-155511. patterns in plants and animals. Nat. Rev. Genet. 11, 204–220. http://dx.doi.org/10.
Frommer, M., McDonald, L.E., Millar, D.S., Collis, C.M., Watt, F., Grigg, G.W., Molloy, P.L., 1038/nrg2719.
Paul, C.L., 1992. A genomic sequencing protocol that yields a positive display of 5- Ledón-Rettig, C.C., 2013. Ecological epigenetics: an introduction to the symposium. Integr.
methylcytosine residues in individual DNA strands. Proc. Natl. Acad. Sci. U. S. A. 89, Comp. Biol. 53, 307–318. http://dx.doi.org/10.1093/icb/ict053.
1827–1831. http://dx.doi.org/10.1073/pnas.89.5.1827. Lee, H.J., Lowdon, R.F., Maricque, B., Zhang, B., Stevens, M., Li, D., Johnson, S.L., Wang, T.,
Fulneček, J., Kovařík, A., 2014. How to interpret methylation sensitive amplified polymor- 2015. Developmental enhancers revealed by extensive DNA methylome maps of
phism (MSAP) profiles? BMC Genet. 15, 2. http://dx.doi.org/10.1186/1471-2156-15-2. zebrafish early embryos. Nat. Commun. 6, 6315. http://dx.doi.org/10.1038/
Geay, F., Zambonino-Infante, J., Reinhardt, R., Kuhl, H., Santigosa, E., Cahu, C., Mazurais, D., ncomms7315.
2012. Characteristics of fads2 gene expression and putative promoter in European sea Li, E., 2002. Chromatin modification and epigenetic reprogramming in mammalian devel-
bass (Dicentrarchus labrax): comparison with salmonid species and analysis of CpG opment. Nat. Rev. Genet. 3, 662–673. http://dx.doi.org/10.1038/nrg887.
methylation. Mar. Genomics 5, 7–13. http://dx.doi.org/10.1016/j.margen.2011.08. Li, E., Zhang, Y., 2014. DNA methylation in mammals. Cold Spring Harb. Perspect. Biol. 6,
003. 1–21. http://dx.doi.org/10.1101/cshperspect.a019133.
Goll, M.G., Halpern, M.E., 2011. DNA methylation in zebrafish. Prog. Mol. Biol. Transl. Sci. Liebl, A.L., Schrey, A.W., Richards, C.L., Martin, L.B., 2013. Patterns of DNA methylation
101, 193–218. http://dx.doi.org/10.1016/B978-0-12-387685-0.00005-6. throughout a range expansion of an introduced songbird. Integr. Comp. Biol. 53,
Goll, M.G., Kirpekar, F., Maggert, K.A., Yoder, J.A., Hsieh, C.-L., Zhang, X., Golic, K.G., 351–358. http://dx.doi.org/10.1093/icb/ict007.
Jacobsen, S.E., Bestor, T.H., 2006. Methylation of tRNAAsp by the DNA methyltransfer- Lister, R., Pelizzola, M., Dowen, R.H., Hawkins, R.D., Hon, G., Tonti-Filippini, J., Nery, J.R.,
ase homolog Dnmt2. Science 311, 395–398. http://dx.doi.org/10.1126/science. Lee, L., Ye, Z., Ngo, Q.-M., Edsall, L., Antosiewicz-Bourget, J., Stewart, R., Ruotti, V.,
1120976. Millar, A.H., Thomson, J.A., Ren, B., Ecker, J.R., 2009. Human DNA methylomes at
Gracey, A.Y., Fraser, E.J., Li, W., Fang, Y., Taylor, R.R., Rogers, J., Brass, A., Cossins, A.R., 2004. base resolution show widespread epigenomic differences. Nature 462, 315–322.
Coping with cold: An integrative, multitissue analysis of the transcriptome of a poiki- http://dx.doi.org/10.1038/nature08514.
lothermic vertebrate. Proc. Natl. Acad. Sci. U. S. A. 101, 16970–16975. http://dx.doi. Logan, C.A., Buckley, B.A., 2015. Transcriptomic responses to environmental temperature
org/10.1073/pnas.0403627101. in eurythermal and stenothermal fishes. J. Exp. Biol. 218, 1915–1924. http://dx.doi.
Gu, H., Bock, C., Mikkelsen, T.S., Jäger, N., Smith, Z.D., Tomazou, E., Gnirke, A., Lander, E.S., org/10.1242/jeb.114397.
Meissner, A., 2010. Genome-scale DNA methylation mapping of clinical samples at Lyko, F., Foret, S., Kucharski, R., Wolf, S., Falckenhayn, C., Maleszka, R., 2010. The honey
single-nucleotide resolution. Nat. Methods 7, 133–136. http://dx.doi.org/10.1038/ bee epigenomes: differential methylation of brain DNA in queens and workers.
nmeth.1414. PLoS Biol. 8, e1000506. http://dx.doi.org/10.1371/journal.pbio.1000506.
Harrison, A., Parle-McDermott, A., 2011. DNA methylation: a timeline of methods and ap- Macleod, D., Clark, V.H., Bird, A., 1999. Absence of genome-wide changes in DNA methyl-
plications. Front. Genet. 2, 74. http://dx.doi.org/10.3389/fgene.2011.00074. ation during development of the zebrafish. Nat. Genet. 23, 139–140. http://dx.doi.
Hsu, P.D., Lander, E.S., Zhang, F., 2014. Development and applications of CRISPR-Cas9 for org/10.1038/13767.
genome engineering. Cell 157, 1262–1278. http://dx.doi.org/10.1016/j.cell.2014.05. Manzon, R.G., Youson, J.H., Holmes, J.A., 2015. Lampreys: Biology, Conservation and Con-
010. trol. Springer, Netherlands, Dordrecht http://dx.doi.org/10.1007/978-94-017-9306-3.
Jaillon, O., Aury, J.-M., Brunet, F., Petit, J.-L., Stange-Thomann, N., Mauceli, E., Bouneau, L., Martinowich, K., Hattori, D., Wu, H., Fouse, S., He, F., Hu, Y., Fan, G., Sun, Y.E., 2003. DNA
Fischer, C., Ozouf-Costaz, C., Bernot, A., Nicaud, S., Jaffe, D., Fisher, S., Lutfalla, G., methylation-related chromatin remodeling in activity-dependent BDNF gene regula-
Dossat, C., Segurens, B., Dasilva, C., Salanoubat, M., Levy, M., Boudet, N., Castellano, tion. Science 302, 890–893. http://dx.doi.org/10.1126/science.1090842.
S., Anthouard, V., Jubin, C., Castelli, V., Katinka, M., Vacherie, B., Biémont, C., Skalli, Massicotte, R., Whitelaw, E., Angers, B., 2011. DNA methylation: a source of random var-
Z., Cattolico, L., Poulain, J., De Berardinis, V., Cruaud, C., Duprat, S., Brottier, P., iation in natural populations. Epigenetics 6, 421–427. http://dx.doi.org/10.4161/epi.6.
Coutanceau, J.-P., Gouzy, J., Parra, G., Lardier, G., Chapple, C., McKernan, K.J., McEwan, 4.14532.
P., Bosak, S., Kellis, M., Volff, J.-N., Guigó, R., Zody, M.C., Mesirov, J., Lindblad-Toh, K., Meyer, J.N., Wassenberg, D.M., Karchner, S.I., Hahn, M.E., Di Giulio, R.T., 2003. Expression
Birren, B., Nusbaum, C., Kahn, D., Robinson-Rechavi, M., Laudet, V., Schachter, V., and inducibility of aryl hydrocarbon receptor pathway genes in wild-caught killifish
Quétier, F., Saurin, W., Scarpelli, C., Wincker, P., Lander, E.S., Weissenbach, J., Roest (Fundulus heteroclitus) with different contaminant-exposure histories. Environ.
Crollius, H., 2004. Genome duplication in the teleost fish Tetraodon nigroviridis reveals Toxicol. Chem. 22, 2337–2343. http://dx.doi.org/10.1897/02-495.
the early vertebrate proto-karyotype. Nature 431, 946–957. http://dx.doi.org/10. Mhanni, A.A., McGowan, R.A., 2004. Global changes in genomic methylation levels during
1038/nature03025. early development of the zebrafish embryo. Dev. Genes Evol. 214, 412–417. http://dx.
Jeddeloh, J.A., Greally, J.M., Rando, O.J., 2008. Reduced-representation methylation map- doi.org/10.1007/s00427-004-0418-0.
ping. Genome Biol. 9, 231. http://dx.doi.org/10.1186/gb-2008-9-8-231. Mirbahai, L., Yin, G., Bignell, J.P., Li, N., Williams, T.D., Chipman, J.K., 2011. DNA methyla-
Jiang, L., Zhang, J., Wang, J.-J., Wang, L., Zhang, L., Li, G., Yang, X., Ma, X., Sun, X., Cai, J., tion in liver tumorigenesis in fish from the environment. Epigenetics 6, 1319–1333.
Zhang, J., Huang, X., Yu, M., Wang, X., Liu, F., Wu, C.-I., He, C., Zhang, B., Ci, W., Liu, http://dx.doi.org/10.4161/epi.6.11.17890.
J., 2013. Sperm, but not oocyte, DNA methylome is inherited by zebrafish early em- Monk, M., Boubelik, M., Lehnert, S., 1987. Temporal and regional changes in DNA methyl-
bryos. Cell 153, 773–784. http://dx.doi.org/10.1016/j.cell.2013.04.041. ation in the embryonic, extraembryonic and germ cell lineages during mouse embryo
Jjingo, D., Conley, A.B., Yi, S.V., Lunyak, V.V., King, I., 2012. On the presence and role of development. Development 99, 371–382.
human gene-body DNA methylation. Oncotarget 3, 462–474. Morán, P., Pérez-Figueroa, A., 2011. Methylation changes associated with early matura-
Jones, P.A., 2012. Functions of DNA methylation: islands, start sites, gene bodies and be- tion stages in the Atlantic salmon. BMC Genet. 12, 86. http://dx.doi.org/10.1186/
yond. Nat. Rev. Genet. 13, 484–492. http://dx.doi.org/10.1038/nrg3230. 1471-2156-12-86.

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004
12 D.C.H. Metzger, P.M. Schulte / Marine Genomics xxx (2016) xxx–xxx

Morán, P., Marco-Rius, F., Megías, M., Covelo-Soto, L., Pérez-Figueroa, A., 2013. Environ- Shen, L., Song, C.-X., He, C., Zhang, Y., 2014. Mechanism and function of oxidative reversal
mental induced methylation changes associated with seawater adaptation in of DNA and RNA methylation. Annu. Rev. Biochem. 83, 585–614. http://dx.doi.org/10.
brown trout. Aquaculture 392-395, 77–83. http://dx.doi.org/10.1016/j.aquaculture. 1146/annurev-biochem-060713-035513.
2013.02.006. Shimoda, N., Yamakoshi, K., Miyake, A., Takeda, H., 2005. Identification of a gene required
Navarro-Martín, L., Viñas, J., Ribas, L., Díaz, N., Gutiérrez, A., Di Croce, L., Piferrer, F., 2011. for de novo DNA methylation of the zebrafish no tail gene. Dev. Dyn. 233, 1509–1516.
DNA methylation of the gonadal aromatase (cyp19a) promoter is involved in http://dx.doi.org/10.1002/dvdy.20455.
temperature-dependent sex ratio shifts in the European sea bass. PLoS Genet. 7. Si, Y., Ding, Y., He, F., Wen, H., Li, J., Zhao, J., Huang, Z., 2016. DNA methylation level of
http://dx.doi.org/10.1371/journal.pgen.1002447. cyp19a1a and Foxl2 gene related to their expression patterns and reproduction traits
Pala, I., Coelho, M.M., Schartl, M., 2008. Dosage compensation by gene-copy silencing in a during ovary development stages of Japanese flounder (Paralichthys olivaceus). Gene
triploid hybrid fish. Curr. Biol. 18, 1344–1348. http://dx.doi.org/10.1016/j.cub.2008. 575, 321–330. http://dx.doi.org/10.1016/j.gene.2015.09.006.
07.096. Si, Y., He, F., Wen, H., Li, J., Zhao, J., Ren, Y., Zhao, M., Ji, L., Huang, Z., Zhang, M., Chen, S.,
Park, J., Peng, Z., Zeng, J., Elango, N., Park, T., Wheeler, D., Werren, J.H., Yi, S.V., 2011. Com- 2015. Genetic polymorphisms and DNA methylation in exon 1 CpG-rich regions of
parative analyses of DNA methylation and sequence evolution using Nasonia ge- PACAP gene and its effect on mRNA expression and growth traits in half smooth
nomes. Mol. Biol. Evol. 28, 3345–3354. http://dx.doi.org/10.1093/molbev/msr168. tongue sole (Cynoglossus semilaevis). Fish Physiol. Biochem. http://dx.doi.org/10.
Perry, S.F., Rivero-Lopez, L., McNeill, B., Wilson, J., 2006. Fooling a freshwater fish: how di- 1007/s10695-015-0147-5.
etary salt transforms the rainbow trout gill into a seawater gill phenotype. J. Exp. Biol. Smith, T.H.L., Collins, T.M., McGowan, R.A., 2011. Expression of the dnmt3 genes in
209, 4591–4596. http://dx.doi.org/10.1242/jeb.02558. zebrafish development: similarity to Dnmt3a and Dnmt3b. Dev. Genes Evol. 220,
Peterson, B.K., Weber, J.N., Kay, E.H., Fisher, H.S., Hoekstra, H.E., 2012. Double digest 347–353. http://dx.doi.org/10.1007/s00427-010-0347-z.
RADseq: an inexpensive method for de novo SNP discovery and genotyping in Smith, G., Smith, C., Kenny, J.G., Chaudhuri, R.R., Ritchie, M.G., 2015. Genome-wide DNA
model and non-model species. PLoS One 7, e37135. http://dx.doi.org/10.1371/ methylation patterns in wild samples of two morphotypes of threespine Stickleback
journal.pone.0037135. (Gasterosteus aculeatus). Mol. Biol. Evol. 32, 888–895. http://dx.doi.org/10.1093/
Pfennig, D.W., Wund, M.A., Snell-Rood, E.C., Cruickshank, T., Schlichting, C.D., Moczek, molbev/msu344.
A.P., 2010. Phenotypic plasticity's impacts on diversification and speciation. Trends Stockwell, P.A., Chatterjee, A., Rodger, E.J., Morison, I.M., 2014. DMAP: differential methyl-
Ecol. Evol. 25, 459–467. http://dx.doi.org/10.1016/j.tree.2010.05.006. ation analysis package for RRBS and WGBS data. Bioinformatics 30, 1814–1822.
Pierron, F., Baillon, L., Sow, M., Gotreau, S., Gonzalez, P., 2014. Effect of low-dose cadmium http://dx.doi.org/10.1093/bioinformatics/btu126.
exposure on DNA methylation in the endangered European eel. Environ. Sci. Technol. Streisinger, G., Walker, C., Dower, N., Knauber, D., Singer, F., 1981. Production of clones of
48, 797–803. http://dx.doi.org/10.1021/es4048347. homozygous diploid zebra fish (Brachydanio rerio). Nature 291, 293–296. http://dx.
Piferrer, F., Beaumont, A., Falguière, J.-C., Flajšhans, M., Haffray, P., Colombo, L., 2009. Poly- doi.org/10.1038/291293a0.
ploid fish and shellfish: production, biology and applications to aquaculture for per- Suetake, I., Shinozaki, F., Miyagawa, J., Takeshima, H., Tajima, S., 2004. DNMT3L stimulates
formance improvement and genetic containment. Aquaculture 293, 125–156. the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction.
http://dx.doi.org/10.1016/j.aquaculture.2009.04.036. J. Biol. Chem. 279, 27816–27823. http://dx.doi.org/10.1074/jbc.M400181200.
Plongthongkum, N., Diep, D.H., Zhang, K., 2014. Advances in the profiling of DNA modifi- Taylor, E., Blouin, M.S., Thuillier, V., Cooper, B., Amarasinghe, V., Cluzel, L., Araki, H.,
cations: cytosine methylation and beyond. Nat. Rev. Genet. 15, 647–661. http://dx. Grunau, C., 2010. No evidence for large differences in genomic methylation between
doi.org/10.1038/nrg3772. wild and hatchery steelhead (Oncorhynchus mykiss). Can. J. Fish. Aquat. Sci. 67,
Potok, M.E., Nix, D.A., Parnell, T.J., Cairns, B.R., 2013. Reprogramming the maternal 217–224. http://dx.doi.org/10.1139/F09-174.
zebrafish genome after fertilization to match the paternal methylation pattern. Cell Timme-Laragy, A.R., Meyer, J.N., Waterland, R.A., Di Giulio, R.T., 2005. Analysis of CpG
153, 759–772. http://dx.doi.org/10.1016/j.cell.2013.04.030. methylation in the killifish CYP1A promoter. Comp. Biochem. Physiol. C Toxicol.
Powers, D., 1989. Fish as model systems. Science 246, 352–358. http://dx.doi.org/10.1126/ Pharmacol. 141, 406–411. http://dx.doi.org/10.1016/j.cbpc.2005.09.009.
science.2678474. Umer, M., Herceg, Z., 2013. Deciphering the epigenetic code: an overview of DNA meth-
Ray, P.D., Yosim, A., Fry, R.C., 2014. Incorporating epigenetic data into the risk assessment ylation analysis methods. Antioxid. Redox Signal. 18, 1972–1986. http://dx.doi.org/
process for the toxic metals arsenic, cadmium, chromium, lead, and mercury: strate- 10.1089/ars.2012.4923.
gies and challenges. Front. Genet. 5, 1–26. http://dx.doi.org/10.3389/fgene.2014. Vanyushin, B.F., Mazin, A.L., Vasilyev, V.K., Belozersky, A.N., 1973. The content of 5-
00201. methylcytosine in animal DNA: the species and tissue specificity. Biochim. Biophys.
Razin, A., 1998. CpG methylation, chromatin structure and gene silencing-a three-way Acta 299, 397–403. http://dx.doi.org/10.1016/0005-2787(73)90264-5.
connection. EMBO J. 17, 4905–4908. http://dx.doi.org/10.1093/emboj/17.17.4905. Vanyushin, B.F., Tkacheva, S.G., Belozersky, A.N., 1970. Rare bases in animal DNA. Nature
Reyna-López, G.E., Simpson, J., Ruiz-Herrera, J., 1997. Differences in DNA methylation pat- 225, 948–949. http://dx.doi.org/10.1038/225948a0.
terns are detectable during the dimorphic transition of fungi by amplification of re- Varriale, A., Bernardi, G., 2006. DNA methylation and body temperature in fishes. Gene
striction polymorphisms. Mol. Gen. Genet. 253, 703–710. http://dx.doi.org/10.1007/ 385, 111–121. http://dx.doi.org/10.1016/j.gene.2006.05.031.
s004380050374. Véron, N., Peters, A.H.F.M., 2011. Epigenetics: Tet proteins in the limelight. Nature 473,
Rhee, K.-D., Yu, J., Zhao, C.Y., Fan, G., Yang, X.-J., 2012. Dnmt1-dependent DNA methyla- 293–294. http://dx.doi.org/10.1038/473293a.
tion is essential for photoreceptor terminal differentiation and retinal neuron surviv- Waddington, C.H., 1942. The epigenotype. Endeavour 18–20. http://dx.doi.org/10.1093/
al. Cell Death Dis. 3, e427. http://dx.doi.org/10.1038/cddis.2012.165. ije/dyr184.
Richardson, B., 2003. Impact of aging on DNA methylation. Ageing Res. Rev. 2, 245–261. Wang, Y., Wang, C., Zhang, J., Chen, Y., Zuo, Z., 2009. DNA hypomethylation induced by tri-
http://dx.doi.org/10.1016/S1568-1637(03)00010-2. butyltin, triphenyltin, and a mixture of these in Sebastiscus marmoratus liver. Aquat.
Schaefer, S., Nadeau, J.H., 2015. The genetics of epigenetic inheritance: modes, molecules, Toxicol. 95, 93–98. http://dx.doi.org/10.1016/j.aquatox.2009.06.008.
and mechanisms. Q. Rev. Biol. 90, 381–415. Wen, A.Y., You, F., Sun, P., Li, J., Xu, D.D., Wu, Z.H., Ma, D.Y., Zhang, P.J., 2014. CpG methyl-
Schield, D.R., Walsh, M.R., Card, D.C., Andrew, A.L., Adams, R.H., Castoe, T.A., 2015. ation of dmrt1 and cyp19a promoters in relation to their sexual dimorphic expression
EpiRADseq: scalable analysis of genomewide patterns of methylation using next- in the Japanese flounder Paralichthys olivaceus. J. Fish Biol. 84, 193–205. http://dx.doi.
generation sequencing. Methods Ecol. Evol. http://dx.doi.org/10.1111/2041-210X. org/10.1111/jfb.12277.
12435. Winn, R.N., Van Beneden, R.J., Burkhart, J.G., 1995. Transfer, methylation and spontaneous
Schrey, A.W., Alvarez, M., Foust, C.M., Kilvitis, H.J., Lee, J.D., Liebl, A.L., Martin, L.B., mutation frequency of ΦX174am3cs70 sequences in medaka (Oryzias latipes) and
Richards, C.L., Robertson, M., 2013. Ecological epigenetics: beyond MS-AFLP. Integr. mummichog (Fundulus heteroclitus): implications for gene transfer and environmen-
Comp. Biol. 53, 340–350. http://dx.doi.org/10.1093/icb/ict012. tal mutagenesis in aquatic species. Mar. Environ. Res. 40, 247–265. http://dx.doi.org/
Schrey, A.W., Richards, C.L., Meller, V., Sollars, V., Ruden, D.M., 2012. The role of epige- 10.1016/0141-1136(94)00144-E.
netics in evolution: the extended synthesis. Genet. Res. Int. 2012, 286164. http://dx. Wolff, G.L., Kodell, R.L., Moore, S.R., Cooney, C.A., 1998. Maternal epigenetics and methyl
doi.org/10.1155/2012/286164. supplements affect agouti gene expression in Avy/a mice. FASEB J. 12, 949–957.
Scott, G.R., Johnston, I.A., 2012. Temperature during embryonic development has persis- Wu, C.-t, Morris, J.R., 2001. Genes, genetics, and epigenetics: a correspondence. Science
tent effects on thermal acclimation capacity in zebrafish. Proc. Natl. Acad. Sci. 109, 293, 1103–1105. http://dx.doi.org/10.1126/science.293.5532.1103.
14247–14252. http://dx.doi.org/10.1073/pnas.1205012109. Yokomine, T., Hata, K., Tsudzuki, M., Sasaki, H., 2006. Evolution of the vertebrate DNMT3
Shao, C., Li, Q., Chen, S., Zhang, P., Lian, J., Hu, Q., Sun, B., Jin, L., Liu, S., Wang, Z., Zhao, H., gene family: a possible link between existence of DNMT3L and genomic imprinting.
Jin, Z., Liang, Z., Li, Y., Zheng, Q., Zhang, Y., Wang, J., Zhang, G., 2014. Epigenetic mod- Cytogenet. Genome Res. 113, 75–80. http://dx.doi.org/10.1159/000090817.
ification and inheritance in sexual reversal of fish. Genome Res. 24, 604–615. http:// Zemach, A., McDaniel, I.E., Silva, P., Zilberman, D., 2010. Genome-wide evolutionary anal-
dx.doi.org/10.1101/gr.162172.113. ysis of eukaryotic DNA methylation. Science 328, 916–919. http://dx.doi.org/10.1126/
Sharif, J., Muto, M., Takebayashi, S., Suetake, I., Iwamatsu, A., Endo, T.A., Shinga, J., science.1186366.
Mizutani-Koseki, Y., Toyoda, T., Okamura, K., Tajima, S., Mitsuya, K., Okano, M., Ziller, M.J., Hansen, K.D., Meissner, A., Aryee, M.J., 2014. Coverage recommendations for
Koseki, H., 2007. The SRA protein Np95 mediates epigenetic inheritance by recruiting methylation analysis by whole-genome bisulfite sequencing. Nat. Methods 12, 2–5.
Dnmt1 to methylated DNA. Nature 450, 908–912. http://dx.doi.org/10.1038/ http://dx.doi.org/10.1038/nmeth.3152.
nature06397.
Shen, L., Waterland, R.A., 2007. Methods of DNA methylation analysis. Curr. Opin. Clin.
Nutr. Metab. Care 10, 576–581. http://dx.doi.org/10.1097/MCO.0b013e3282bf6f43.

Please cite this article as: Metzger, D.C.H., Schulte, P.M., Epigenomics in marine fishes, Mar. Genomics (2016), http://dx.doi.org/10.1016/
j.margen.2016.01.004

You might also like