ArticlePDF Available

DNMT1, DNMT3A and DNMT3B proteins are differently expressed in mouse oocytes and early embryos

Authors:

Abstract and Figures

DNA methylation is one of the epigenetic mechanisms and plays important roles during oogenesis and early embryo development in mammals. DNA methylation is basically known as adding a methyl group to the fifth carbon atom of cytosine residues within cytosine–phosphate–guanine (CpG) and non-CpG dinucleotide sites. This mechanism is composed of two main processes: de novo methylation and maintenance methylation, both of which are catalyzed by specific DNA methyltransferase (DNMT) enzymes. To date, six different DNMTs have been characterized in mammals defined as DNMT1, DNMT2, DNMT3A, DNMT3B, DNMT3C, and DNMT3L. While DNMT1 primarily functions in maintenance methylation, both DNMT3A and DNMT3B are essentially responsible for de novo methylation. As is known, either maintenance or de novo methylation processes appears during oocyte and early embryo development terms. The aim of the present study is to investigate spatial and temporal expression levels and subcellular localizations of the DNMT1, DNMT3A, and DNMT3B proteins in the mouse germinal vesicle (GV) and metaphase II (MII) oocytes, and early embryos from 1-cell to blastocyst stages. We found that there are remarkable differences in the expressional levels and subcellular localizations of the DNMT1, DNMT3A and DNMT3B proteins in the GV and MII oocytes, and 1-cell, 2-cell, 4-cell, 8-cell, morula, and blastocyst stage embryos. The fluctuations in the expression of DNMT proteins in the analyzed oocytes and early embryos are largely compatible with DNA methylation changes and genomic imprintestablishment appearing during oogenesis and early embryo development. To understand precisemolecular biological meaning of differently expressing DNMTs in the early developmental periods, further studies are required.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
J Mol Hist (2017) 48:417–426
DOI 10.1007/s10735-017-9739-y
ORIGINAL PAPER
DNMT1, DNMT3A andDNMT3B proteins are differently
expressed inmouse oocytes andearly embryos
FatmaUysal1· SaffetOzturk1· GokhanAkkoyunlu1
Received: 26 July 2017 / Accepted: 9 October 2017 / Published online: 13 October 2017
© Springer Science+Business Media B.V. 2017
DNA methylation changes and genomic imprintestablish-
ment appearing during oogenesis and early embryo develop-
ment. To understand precisemolecular biological meaning
of differently expressing DNMTs in the early developmental
periods, further studies are required.
Keywords DNA methyltransferase· DNA methylation·
Oocyte· Early embryo
Introduction
DNA methylation is an epigenetic mechanism and plays crit-
ical roles in transcriptional repression or activation, X-chro-
mosome inactivation, cell differentiation, and tumorogenesis
(Uysal etal. 2015). DNA methylation is specifically cata-
lyzed by various types of DNA methyltransferase (DNMT)
enzymes, and there are two different DNA methylation pro-
cesses: maintenance and de novo. In the maintenance meth-
ylation, previously established DNA methylation patterns
are continued at the end of each round of DNA replication.
By contrast, new DNA methylation marks in the genomic
DNA are created in the de novo methylation process.
TheDNMT enzymes basicallyadd a methyl group to the
fifth carbon atom of cytosine residues commonly in the
cytosine–phosphate–guanine (CpG) and rarely in the non-
CpG islands such as cytosine–phosphate–thymine (CpT),
cytosine–phosphate–adenine (CpA), and cytosine–phos-
phate–cytosine (CpC)by using S-adenosyl methionine
(AdoMet) as a methyl donor (Turek-Plewa and Jagodz-
inski 2005). To date, structurally and functionally six
different DNMTs have been characterized in mammals:
DNMT1, DNMT2, DNMT3A, DNMT3B, DNMT3C,
and DNMT3L. The DNMT1 protein primarily functions
in maintenance methylation by adding methyl groups to
Abstract DNA methylation is one of the epigenetic
mechanisms and plays important roles during oogenesis
and early embryo development in mammals. DNA meth-
ylation is basically known as adding a methyl group to the
fifth carbon atom of cytosine residues within cytosine–
phosphate–guanine (CpG) and non-CpG dinucleotide sites.
This mechanism is composed of two main processes: de
novo methylation and maintenance methylation, both of
which are catalyzed by specific DNA methyltransferase
(DNMT) enzymes. To date, six different DNMTs have been
characterized in mammals defined as DNMT1, DNMT2,
DNMT3A, DNMT3B, DNMT3C, and DNMT3L. While
DNMT1 primarily functions in maintenance methylation,
both DNMT3A and DNMT3B are essentially responsible
for de novo methylation. As is known, either maintenance
or de novo methylation processes appears during oocyte and
early embryo development terms. The aim of the present
study is to investigate spatial and temporal expression lev-
els and subcellular localizations of the DNMT1, DNMT3A,
and DNMT3B proteins in the mouse germinal vesicle (GV)
and metaphase II (MII) oocytes, and early embryos from
1-cell to blastocyst stages. We found that there are remark-
able differences in the expressional levels and subcellular
localizations of the DNMT1, DNMT3A and DNMT3B pro-
teins in the GV and MII oocytes, and 1-cell, 2-cell, 4-cell,
8-cell, morula, and blastocyst stage embryos. The fluctua-
tions in the expression of DNMT proteins in the analyzed
oocytes and early embryos are largely compatible with
* Gokhan Akkoyunlu
gokhan_akkoyunlu@hotmail.com
1 Department ofHistology andEmbryology, Akdeniz
University School ofMedicine, Campus, 07070Antalya,
Turkey
418 J Mol Hist (2017) 48:417–426
1 3
the hemi-methylated DNA strands during DNA replica-
tion (Bestor 2000), and also it partially contributes to de
novo methylation process (Fatemi etal. 2002). And, the
DNMT2 protein can methylate cytosine 38 in the antico-
don loop of aspartic acid transfer RNA instead of methyl-
ating genomic DNA (Goll etal. 2006). On the other hand,
both DNMT3A and DNMT3B essentially implicate in the
de novo methylation process, commonly being appeared in
the unmethylated DNA strands at the CpG islands (Turek-
Plewa and Jagodzinski 2005). DNMT3C is recently identi-
fied de novo methyltransferase which protects male germ
cells from retrotransposon activity and is essential for
mouse fertility (Barau etal. 2016). DNMT3L does not
have any catalytic domain; however, it is capable of induc-
ing DNMT3A and DNMT3B activity, and thereby contrib-
utes to de novo methylation indirectly (Deplus etal. 2002;
Margot etal. 2003).
In mammals, female germ cells are originated from pri-
mordial germ cells (PGCs) arising in the endodermof yolk
sac. When female PGCs reach to the gonadal ridges at the
end of long migration during which they undergo many
mitotic divisions, the PGCs ultimately differentiate into
oogonia. The oogoniaare defined as oocytes after entering
meiotic division, and the oocytes are arrested at prophase I
(PI) stage of first meiotic division in the developing ovary
before birth. The oocytes at the PI stage in the ovarian folli-
cles from primordial to antral follicles are immature oocytes
also described as germinal vesicle (GV) stage. In the ovula-
tory follicles, the GV oocytes fulfill the first meiotic division,
enter into second meiotic division, and become arrested at
the metaphase II (MII) stage known as mature oocytes (Ver-
lhac and Terret 2016). Once the MII oocytes are fertilized
by mature spermatozoa, 1-cell embryos (zygotes) includ-
ing female and male pronuclei are formed. Then, the 1-cell
embryos undergo consecutive mitotic divisions (cleavage)
and develop into 2-cell, 4-cell, 8-cell, morula and blastocyst
stage embryos, respectively. The early embryo development
from 1-cell to blastocysts includes exclusive events such as
cleavage, embryonic genome activation (EGA), compaction,
and cavitation. EGA is shortly defined as synthesis of the
required RNAs from newly formed embryonic genome and
occurs at the 2-cell stage in mice (Bolton etal. 1984). In the
eight-cell stage embryo, the blastomeres enable to create
tight junctions with each other so that compaction is par-
tially commenced. The compaction is thoroughly completed
in the morula stage embryos in which the cellular borders
become fuzzy and cannot be distinguished easily. Following
morula stage, cavitation known as formation of the blasto-
coel cavity filled with fluid take place in the blastocyst stage
embryos. The blastocoel cavity facilitates division of the
embryonic cells into two different layers in the blastocysts:
trophoblast cells (TE) and inner cell mass (ICM) (Fujimori
2010).
During oogenesis and preimplantation embryo devel-
opment, DNA methylation plays critical roles in strictly
regulating stimulation or repression of the development-
related genes and in timely establishing maternal and pater-
nal imprints (Bartolomei and Ferguson-Smith 2011). The
globalDNA methylation in the growing GV oocytes begins
to progressively increase after birth, and reaches to the
highest levels in the MII oocytes at puberty (Saitou etal.
2012; Smallwood and Kelsey 2012) (Fig.4). During early
embryo development, the global DNA methylation gradually
reduces from 1-cell embryos to 16-cell embryos, and it starts
to increase in the blastocyst stage embryos onward (Small-
wood and Kelsey 2012) (Fig.4). Although DNA methylation
profiles during oogenesis and early embryo development are
well-known, the spatial and temporal expression levels and
subcellular localizations of the DNMTs in the oocytes and
early embryos are not characterized in detail. We hypoth-
esized that the DNMT1, DNMT3A, and DNMT3B pro-
teins display different expression patterns and subcellular
localizations during oocyte and early embryo develop-
ment.The aim of thisstudy was to determine the subcellular
localizationsand relative expression levelsof the DNMT1,
DNMT3A, and DNMT3B proteinsin the mouse oocytes and
early embryos.
Materials andmethods
Animals
The experimental protocol was approved by the Animal Care
and Usage Committee of Akdeniz University (Protocol no:
B.30.2.AKD.0.05.07.00/30). The female Balb/C mice at 4–6
weeks and male mice at 8–10 weeks of age were purchased
from Research Animal Laboratory Unit of Akdeniz Univer-
sity Medical Faculty. All mice were hosted with free access
to food and water, and kept in a 12h light/dark cycle.
Collection ofoocytes andearly embryos
The GV and MII oocytes, and 1-cell, 2-cell, 4-cell, 8-cell,
morula and blastocyst stage embryos were obtained from 4
to 6week-old female Balb/C mice superovulated with 5IU
pregnant mare’s serum gonadotropin (PMSG; Intervet, Mil-
ton Keynes, UK) and 5IU human chorionic gonadotropin
(hCG; Sigma-Aldrich, St. Louis, MO, USA). It is impor-
tant to note that each oocyte and early embryo type was
recruited at least three different mice. To obtain GV oocytes,
the female mice were injected intraperitoneally (i.p.) with
0.1mL 5IU of PMSG. Twenty hours after PMSG injection,
the female mice were killed and then ovaries were punctured
with a 23-gauge needle in human tubal fluid (HTF) medium
(Vitrolife, Gotheburg, Sweden), and we collected GV
419J Mol Hist (2017) 48:417–426
1 3
oocytes having a visible germinal vesicle by using a mouth-
controlled pipette under a dissecting microscope (Zeiss,
Oberkochen, Germany) as described previously (Akko-
yunlu etal. 2007). For collection of MII oocytes, the female
mice were injected i.p. with 5IU PMSG, and subsequently
5IU hCG was administered 48h after PMSG injection. We
obtained the cumulus–oocyte complexes (COCs) from ovi-
ducts’ ampulla region of the mice 14h after hCG injection.
The COCs were treated with HTF medium including 1mg/
mL hyaluronidase (Sigma-Aldrich) to remove cumulus cells,
and the MII oocytes in normal morphology and having vis-
ible polar bodies are pooled.
To collect early embryos, female mice at 4–6 weeks old
were injected i.p. with 5IU PMSG. Following 48h PMSG
treatment, 5IU hCG were injected to the PMSG-primed
female mice immediately mated overnight with mature male
mice at a rate of 2 female:1 male. The next morning, pres-
ence of vaginal plug has been controlled, and the female
mice including vaginal plug were used to obtain 1-cell,
2-cell, 4-cell, 8-cell and morula stage embryos from oviducts
and blastocysts from uterus. The early embryos were col-
lected at the following time points after hCG injection: 1-cell
embryos at 20h, 2-cell embryos at 42h, 4-cell embryos at
60h, 8-cell embryo at 68h, morula at 72h, and blastocyst
at 96h. Notably, the cumulus cells surrounding the 1-cell
embryos were removed by using hyaluronidase at a concen-
tration of 1mg/mL as described above.
Immunofluorescence staining
Immunofluorescence staining has been applied to charac-
terize the subcellular localizations and relative expression
profiles of the DNMT1, DNMT3A, and DNMT3B pro-
teins in the GV and MII oocytes, and 1-cell, 2-cell, 4-cell,
8-cell, morula, and blastocyst stage embryos obtained from
superovulated female mice (n 10 oocytes or early embryos
stained per group). All oocytes and early embryos were
fixed in 3% paraformaldehyde (Sigma-Aldrich), and then
permeabilized with 1% tween-20 (Sigma-Aldrich) prepared
in 1 × PBS at room temperature. The fixed oocytes and early
embryos were blocked with blocking solution including 20%
normal goat serum (Vector Laboratory, Burlingame, CA,
USA). Then, we incubated the oocytes and embryos over-
night at 4°C with primary antibodies specific for DNMT1
[Abcam, Cambridge, USA; ab87654, reactive with either
oocyte-specific (DNMT1o) or somatic (DNMT1s) iso-
forms], DNMT3A (Abcam, ab23565) or DNMT3B (Abcam,
ab2851). After washing three times for 10min with 1 × PBS
including 2% bovine serum albumin (BSA) (PBS–BSA;
Sigma-Aldrich), the oocytes and early embryos were incu-
bated with anti-rabbit Alexa 488 secondary antibody (Inv-
itrogen) for 1h at room temperature in the dark. After incu-
bation with secondary antibody, we washed the oocytes and
early embryos three times for 10min with PBS–BSA. After
counterstained with 4,6-diamidino-2-phenylindole (DAPI;
Sigma-Aldrich, D8417) for 2min at room temperature in
the dark, the oocytes and early embryos were washed three
times for 10min with PBS–BSA. All staining steps were
performed using mini well trays (VWR-Thermo Scientific,
Radnor, PA, USA) in a humidified chamber. We mountedthe
oocytes and early embryos onto slides using mounting solu-
tion (Vector Laboratory). All immunostaining experiments
for the oocytes and early embryos were performed at the
same time periods under the same conditions. Fluores-
cence signals originating from oocytes or early embryos
were detected with a motorized fluorescence microscope at
400x magnification (Olympus BX61, Tokyo, Japan). Then,
we have evaluated the micrographs with Image J software
(National Institutes of Health, Bethesda, Maryland, USA),
and the relative expression levels and subcellular locali-
zations of the DNMT proteins have been characterized in
detail.All negative controls involving no primary antibody
were also analyzed, but no signal was detected. It is impor-
tant to note that we carried out all experiments at least three
times.
Statistical analysis
All experiment results were analyzed by one-way analysis
of variance (one-way ANOVA) followed by Dunn’s post hoc
test. We conducted statistical calculations by using SigmaS-
tat for Windows, version 3.5 (Jandel Scientific Corp). For all
tests, P < 0.05 was considered to be statistically significant.
Results
DNMT1 expression intheoocytes andearly embryos
In the current study, subcellular localizations and relative
expression levels of the DNMT1 protein have been ana-
lyzed in the GV and MII oocytes, and 1-cell, 2-cell, 4-cell,
8-cell, morula, and blastocyst stage embryos. DNMT1
protein was abundantly localized in the cytoplasm of the
oocytes and early embryos, but it was at weak levels in
the nuclear regions (Fig.1a). When we have evaluated the
relative DNMT1 expression from GV oocytes to blasto-
cysts by using ImageJ software, it reachedto the highest
levels in both MII oocytes and 1-cell embryos, whereas it
was at the lowest levels in the 8-cell embryos (Fig.1b).
Additionally, MII oocytes, 1-cell embryos and blastocysts
exhibited significantly higher DNMT1 protein expression
than the remaining oocytes and early embryos (P < 0.05;
Fig. 1b). Importantly, DNMT1 expression gradually
decreased from 2-cell embryos to 8-cell embryos, and it
420 J Mol Hist (2017) 48:417–426
1 3
progressively increased from GV oocytes to MII oocytes
and similarly from morula to blastocysts (Fig.1b). As a
result, we revealed here that DNMT1 protein expression
was differently expressed in the oocytes and early embryos
and displayed remarkable fluctuations from GV oocytes
to blastocysts.
DNMT3A expression intheoocytes andearly embryos
When subcellular localizations of the DNMT3A protein in
the GV and MII oocytes, and 1-cell, 2-cell, 4-cell, 8-cell,
morula and blastocyst stage embryos were analyzed, we
found that DNMT3A was strongly localized in the nucleus
of the oocytes and early embryos at all stages (Fig.2a).
Fig. 1 Immunoexpression of the DNMT1 protein in the mouse
oocytes and early embryos. a The immunostaining results of the
DNMT1 protein in the germinal vesicle oocytes (GV), MII oocytes
(MII), 1-cell embryos (1C), 2-cell embryos (2C), 4-cell embryos
(4C), 8-cell embryos (8C), morulae (M) and blastocysts (B) have been
shown. The negative control (NC) included no primary antibody was
used to identify whether there was any non-specific staining. The
DNMT1 protein expression exhibited spatial and temporal differ-
ences from GV oocytes to blastocyst stage embryos. In the micro-
graphs, FITC (green) staining for DNMT1; DAPI (4,6-diamidino-
2-phenylindole; blue) staining for the nuclei; Merge, combination of
the DAPI-stained nucleus with the green fluorescence signal. b The
relative expression levels of the DNMT1 protein in the GV and MII
oocytes, and 1-cell, 2-cell, 4-cell, 8-cell, morula and blastocyst stage
embryos have been analyzed by using ImageJ software. The oocytes
and early embryos expressed the DNMT1 protein at different levels
showing gradual decreases and progressive increases. The statistical
significance among groups has been determined by using one-way
ANOVA test. The letters on the columns denote statistical signifi-
cance, and P < 0.05 is considered as statistically significant. (Color
figure online)
421J Mol Hist (2017) 48:417–426
1 3
However, there was a weak DNMT3A expression in the
cytoplasm of the oocytes and early embryos (Fig.2a).
In addition to subcellular localization analysis, we have
further evaluated the relative DNM3A protein expression
from GV oocytes to blastocyst stage embryos. The blasto-
cyst stage embryos had the highest, but the 8-cell embryos
possessed the lowest DNMT3A expression levels (P < 0.05;
Fig.2b). Importantly, both 1-cell and blastocyst stage
embryos expressed the DNMT3A significantly higher than
the remaining oocytes and early embryos (P < 0.05; Fig.2b).
Remarkably, DNMT3A protein expression progressively
increased from GV oocytes to 1-cell embryos, and it gradu-
ally decreased from 1-cell embryos to 8-cell stage embryos
(Fig.2b). Similarly, a progressive increase from 8-cell to
Fig. 2 Immunoexpression of the DNMT3A protein in the mouse
oocytes and early embryos. a The immunostaining of the DNMT3A
protein in the germinal vesicle oocytes (GV), MII oocytes (MII),
1-cell embryos (1C), 2-cell embryos (2C), 4-cell embryos (4C),
8-cell embryos (8C), morulae (M) and blastocysts (B) have been pre-
sented. The DNMT3A protein was expressed at the higher levels in
the nuclear region than that of the cytoplasm in the oocytes and early
embryos. The negative control (NC) included no primary antibody
has been used to identify whether there is any non-specific stain-
ing. In the micrographs, FITC (green) staining for DNMT3A; DAPI
(4,6-diamidino-2-phenylindole; blue) staining for the nuclei; Merge,
combination of the DAPI-stained nucleus with green fluorescence
signal. b The relative expression levels of the DNMT3A protein in
the GV and MII oocytes, and 1-cell, 2-cell, 4-cell, 8-cell, morulae and
blastocyst stage embryos have been analyzed by using ImageJ soft-
ware. And, we found that the oocytes and early embryos differently
expressed the DNMT3A protein. The statistical significance among
groups was tested by using one-way ANOVA. The letters on the col-
umns depict statistical significance, and P < 0.05 is considered as sta-
tistically significant. (Color figure online)
422 J Mol Hist (2017) 48:417–426
1 3
blastocyst stage embryos has been observed.Taken together,
DNMT3A expression distributes either in the cytoplasm or
nucleus of the oocytes and early embryos at different levels.
Also, it exhibits remarkable fluctuations from GV oocytes
to blastocyst stage embryos.
DNMT3B expression intheoocytes andearly embryos
The subcellular localizations and relative expression levels
of the DNMT3B protein in the GV and MII oocytes, and
1-cell, 2-cell, 4-cell, 8-cell, morula and blastocyst stage
embryos have been examined. The DNMT3B protein was
highly expressed in the nuclear regions of the GV oocytes,
1-cell, 2-cell and morula stage embryos. On the other hand,
it was localized in either nuclear or cytoplasm regions of the
MII oocytes, 4-cell and 8-cell embryos. In the blastocysts,
DNMT3B was more highly expressed in the cytoplasm than
that of in the nucleus (Fig.3a).
When we evaluated the relative expression levels of
DNMT3B from GV oocytes to blastocysts, it reached the
highest levels in the blastocysts, but it was at the lowest
profile in the 8-cell embryos (Fig.3b). Both MII oocytes
and blastocysts had significantly higher DNMT3B protein
expression than that of the remaining oocytes and early
embryos except for the 1-cell embryos (P < 0.05; Fig.3b).
Following predominant increase of the DNMT3B protein
expression from GV oocytes to MII oocytes, it began to
be gradually reduced from MII oocytes to 8-cell embryos.
Then, the DNMT3B expression progressively increased
from 8-cell embryos to blastocyst stage embryos (Fig.3b).
Overall, DNMT3B intensively located to the nuclear
region of the oocytes and early embryos. Besides, the rela-
tive expression of the DNMT3B protein exhibited gradual
decrease from MII oocytes to 8-cell embryos, and progres-
sively increase from 8-cell to blastocyst stage embryos
except for the GV oocytes remained at low level. It is impor-
tant to note that the expression profiles of DNMT3B protein
in the oocytes and early embryos coincide to a large extent
with that of the DNMT3A.
Discussion
In the present study, we characterized for the first time that
DNMT1, DNMT3A, and DNMT3B proteins exhibited
spatial and temporal expressional differences. And, they
were differently localized in the mouse oocytes and early
embryos. The DNMT1 protein was predominantly resided
in the cytoplasm of the oocytes and early embryos, and
showed gradual decrease and progressive increase from GV
oocytes to blastocyst stage embryos. Unlike DNMT1 subcel-
lular localization, DNMT3A was abundantly localized in the
nucleus of the oocytes and early embryos, and it exhibited
expressional fluctuations from GV oocytes to blastocysts.
Similarly, DNMT3B was more intensively expressed in the
nucleus when compared to its cytoplasmic localization in the
oocytes and early embryos. The DNMT3B expression levels
gradually decreased and progressively increased from GV
oocytes to blastocyst stage embryos, largely coinciding with
the expression distributions of the DNMT3A protein at the
same developmental terms.
DNA methylation is one of the epigenetic mechanisms
and plays critical roles in the expressional regulation of
the development-related genes, which are required during
oogenesis and early embryos. During oogenesis, global DNA
methylation begins to progressively increase in the growing
GV oocytes at the time period of from birth to puberty, and
it reaches the highest levels in the MII oocytes in mouse
(Saitou etal. 2012). Following fertilization, the global DNA
methylation levels gradually decreases from 1-cell to 16-cell
or morula stage embryos, and the paternally and maternally
imprinted genes are also demethylated late in embryogenesis
(Yang etal. 2007). In blastocysts, global DNA methylation
reaches the highest levels because new genomic imprints
are established by activating de novo methylation process
as well as maintenance methylation establishments (Saitou
etal. 2012).
In the present study, we demonstrated that the expression
levels of either DNMT1 or DNMT3B proteins were remark-
ably increased from GV oocytes to MII oocytes except for
DNMT3A which remained at the same levels in both oocyte-
types (Fig.4). This predominant increase of either DNMT1
or DNMT3B expression is largely compatible with the
increasing global DNA methylation levels from GV oocytes
to MII oocytes. Both proteins most likely implicate in estab-
lishing maintenance and de novo methylation processes in
both GV and MII oocytes. On the other hand, DNMT1,
DNMT3A, and DNMT3B protein expression were gradu-
ally decreased from 1-cell embryo to morula, and they were
progressively increased from morula to blastocysts (Fig.4).
The expression patterns of the DNMT proteins parallel with
the gradually decreasing DNA methylation levels from 1-cell
to morula stage embryos because there is no need for highly
expressed DNMTs at this time points of early development.
Since new genomic imprints, do novo and maintenance
methylation begin to be reestablished in the blastocyst stage
embryos, consistent with this we found that the expression
levels of the DNMT1, DNMT3A and DNMT3B proteinsare
at the highest levels in the blastocysts.
It has been reported in the previous studies (Mertineit
etal. 1998; Howell etal. 2001; Hirasawa etal. 2008) that
DNMT1 is intensively localized in the cytoplasm of oocytes
and early embryos as is detected in our current study. Rat-
nam etal. (2002) found that Dnmt1mRNA is present in the
postnatal oocytes obtained from days 1, 5, 10, 15, 21 and 35
in mouse (Ratnam etal. 2002). In bovines, DNMT1 is also
423J Mol Hist (2017) 48:417–426
1 3
identified at all GV oocytestages (from GV0 to GV3) and in
the MII oocytes (Lodde etal. 2009). Similar to the findings
from mouse and bovines, DNMT1 is expressed in human GV
and MII oocytes (Huntriss etal. 2004). Although DNMT1
is predominantly localized in the nucleus of GV oocytes,
it is intensively resided in the cytoplasm of MII oocytes in
humans (Petrussa etal. 2014). These results are moderately
comparable with our findings, and the differences former
and present studies are most likely originates from use of dif-
ferent species samples, analyzing transcript level other than
Fig. 3 Immunoexpression of the DNMT3B protein in the mouse
oocytes and early embryos. a The immunostaining of DNMT3B pro-
tein in the germinal vesicle oocytes (GV), MII oocytes (MII), 1-cell
embryos (1C), 2-cell embryos (2C), 4-cell embryos (4C), 8-cell
embryos (8C), morulae (M) and blastocysts (B) have been presented.
The DNMT3B protein expression differs in the oocytes and early
embryos. The negative control (NC) included no primary antibody
has been used to identify whether there is any non-specific stain-
ing. In the micrographs, FITC (green) staining for DNMT3B; DAPI
(4,6-diamidino-2-phenylindole; blue) staining for the nuclei; Merge,
combination of the DAPI-stained nuclei with green fluorescence sig-
nal. b The relative expression levels of the DNMT3B protein in the
GV and MII oocytes, and 1-cell, 2-cell, 4-cell, 8-cell, morula and
blastocyst stage embryoshave been analyzed by using ImageJ soft-
ware.We observed that DNMT3B exhibited gradual decreases and
progressive increases from GV oocytes to blastocysts. The statisti-
cal significance between groups has been determined by using one-
way ANOVA test. The letters on the columns depict statistical sig-
nificance, and P < 0.05 is considered as statistically significant. (Color
figure online)
424 J Mol Hist (2017) 48:417–426
1 3
protein level in the previous investigations and employing
distinct techniques to determine DNMT1 expression.
The Dnmt3a and Dnmt3bmRNAsare also foundat detect-
able levels in mouse, cat and bovine oocytes (Bourc’his etal.
2001; Kaneda etal. 2004; O’Doherty etal. 2012) as well as
in human oocytes (Huntriss etal. 2004). Ko etal. (2005)
revealed that DNMT3A is highly localized in the cytoplasm
of the 8-cell, morula and blastocyst stage embryos, but it is
intensively resided in the nuclear regions of the 1-cell, 2-cell
and 4-cell embryos in mice (Ko etal. 2005). On the other
hand, Hirasawa etal. (2008) demonstrated that DNMT3A
is abundantly resided in the nuclear region of the GV
oocytes, 1-cell, 2-cell, 4-cell, 8-cell, morula stage embryos
and that there is an intensive cytoplasmic expression in the
MII oocytes in mouse (Hirasawa etal. 2008). Intriguingly,
the blastocyst stage embryos do not exhibit any DNMT3A
expression either in the nuclear or cytoplasm regions
(Hirasawa etal. 2008). The DNMT3B expression has not
been observed in the GV oocytes and 1-cell embryos, and
it is localized in the nuclear region of the 2-cell embryos
and onward in mouse (Hirasawa etal. 2008). In contrast,
Ko etal. (2005) reported that DNMT3B is mainly local-
ized in the cytoplasm of the early embryos from 1-cell to
blastocyst stages (Ko etal. 2005). We have revealed in the
present study that both DNMT3A and DNMT3B are inten-
sively detected in the nucleus, and weakly localized in the
cytoplasm of the oocytes and early embryos. Among the
studies, there are prominent differences about intracellular
intensities of the DNMT3A and DNMT3B proteins in the
mouse oocytes and early embryos, this may derive from that
use of distinct technical applications and/or mouse strains
from which oocytes and early embryos are obtained.
In bovines, Dobbs etal. (2013) observed that global
DNA methylation is gradually decreased from 2-cell to
Fig. 4 Global DNA methylation profile and relative expression levels
of DNMT1, DNMT3A and DNMT3B proteins in the mouse oocytes
and early embryos. Red line, the global DNA methylation levels in
the oocytes and early embryos. We have created this line based on
the studies by Saitou etal. (2012) and Smallwood and Kelsey (2012).
Expression level of DNMT1 gene (pink line), DNMT3A (navy blue
line) and DNMT3B (blue line) in the oocytes and early embryos. The
expressional difference has been shown by distinct colour. GV germi-
nal vesicle oocyte, MII metaphase II oocyte, 1C 1-cell embryo, 2C
2-cell embryo, 4C 4-cell embryo, 8C 8-cell embryo, M morula, B
blastocyst. (Color figure online)
425J Mol Hist (2017) 48:417–426
1 3
6–8 cell stage embryos meaning that the lowest DNA
methylation levels has been detected at the 6–8 cell
embryos, and the methylation reaches the highest lev-
els in the blastocysts (Dobbs etal. 2013). In accordance
with reducing global DNA methylation levels after 2-cell
embryos, DNMT3B mRNA expression decreases from
2-cell to 6–8 cell embryo stages, and remarkably increases
in the blastocyst stage embryos (Dobbs etal. 2013). The
expression patterns of theDNMT3B mRNA from this study
and DNMT3B protein expression from our study argues
that DNMT3B likely functions in the genomic imprint
and maintenance methylation establishments during early
development in mouse and bovines.
As is known, the first lineage differentiation occurs
in the 8-cell embryos, and increaseof intercellular adhe-
sion, polarization and compaction take place based on
the increased cell–cell associations among blastomeres
(Albert and Peters 2009). And, DNA methylation is at low
levels in the 8-cell embryos (Dobbs etal. 2013). Consist-
ent with this DNMT1, DNMT3A and DNMT3B protein
expression is remarkably decreased at this stage. Since
many proteins including E-cadherin, catenin, mitogen-
activated protein kinase (MAPK), phosphatidylinositol
3-kinase (PtdIns3K), Wingless (Wnt), and Notch (Zhang
etal. 2007) are highly expressed in the 8-cell embryos,
the decreased global DNA methylation levels at this stage
may be required for facilitating the expression of these
genes in time.
In conclusion, this is the first detailed study analyz-
ing subcellular localizations and relative expression lev-
els of the DNMT1, DNMT3A and DNMT3B proteins
in the mouse oocytes and early embryos. We found that
DNMT1, DNMT3A and DNMT3B exhibit expressional
fluctuations from GV oocytes to blastocyst stage embryos.
These fluctuations are to large extent compatible with
timely establishing global DNA methylation and genomic
imprints in the oocytes and early embryos.We suppose that
more detailed molecular biological studies are required to
clearly understand the potential interactions of the DNMTs
in establishing DNA methylation and in the regulation of
early development-related genes during oogenesis and
early embryo development.
Funding This study was supported by Akdeniz University Sci-
entific Research Projects Coordination Unit (Project Number:
2014.02.0122.013).
Compliance with ethical standards
Conflict of interest The authors declare that there is no conflict of
interest.
References
Akkoyunlu G, Ustunel I, Demir R (2007) The distribution of trans-
glutaminase in the rat oocytes and embryos. Theriogenology
68(6):834–841
Albert M, Peters AH (2009) Genetic and epigenetic control of early
mouse development. Curr Opin Genet Dev 19(2):113–121
Barau J, Teissandier A, Zamudio N, Roy S, Nalesso V, Herault Y,
Guillou F, Bourc’his D (2016) The DNA methyltransferase
DNMT3C protects male germ cells from transposon activity.
Science 354(6314):909–912
Bartolomei MS, Ferguson-Smith AC (2011) Mammalian genomic
imprinting. Cold Spring Harb Perspect Biol 3(7)
Bestor TH (2000) The DNA methyltransferases of mammals. Hum
Mol Genet 9(16):2395–2402
Bolton VN, Oades PJ, Johnson MH (1984) The relationship between
cleavage, DNA replication, and gene expression in the mouse
2-cell embryo. J Embryol Exp Morphol 79:139–163
Bourc’his D, Xu GL, Lin CS, Bollman B, Bestor TH (2001) Dnmt3L
and the establishment of maternal genomic imprints. Science
294(5551):2536–2539
Deplus R, Brenner C, Burgers WA, Putmans P, Kouzarides T, de
Launoit Y, Fuks F (2002) Dnmt3L is a transcriptional repres-
sor that recruits histone deacetylase. Nucleic Acids Res
30(17):3831–3838
Dobbs KB, Rodriguez M, Sudano MJ, Ortega MS, Hansen PJ (2013)
Dynamics of DNA methylation during early development of the
preimplantation bovine embryo. PLoS ONE 8(6):e66230
Fatemi M, Hermann A, Gowher H, Jeltsch A (2002) Dnmt3a and
Dnmt1 functionally cooperate during de novo methylation of
DNA. Eur J Biochem 269(20):4981–4984
Fujimori T (2010) Preimplantation development of mouse: a view
from cellular behavior. Dev Growth Differ 52(3):253–262
Goll MG, Kirpekar F, Maggert KA, Yoder JA, Hsieh CL, Zhang
X, Golic KG, Jacobsen SE, Bestor TH (2006) Methylation of
tRNAAsp by the DNA methyltransferase homolog Dnmt2. Sci-
ence 311(5759):395–398
Hirasawa R, Chiba H, Kaneda M, Tajima S, Li E, Jaenisch R, Sasaki
H (2008) Maternal and zygotic Dnmt1 are necessary and suf-
ficient for the maintenance of DNA methylation imprints during
preimplantation development. Genes Dev 22(12):1607–1616
Howell CY, Bestor TH, Ding F, Latham KE, Mertineit C, Trasler JM,
Chaillet JR (2001) Genomic imprinting disrupted by a maternal
effect mutation in the Dnmt1 gene. Cell 104(6):829–838
Huntriss J, Hinkins M, Oliver B, Harris SE, Beazley JC, Rutherford
AJ, Gosden RG, Lanzendorf SE, Picton HM (2004) Expression
of mRNAs for DNA methyltransferases and methyl-CpG-binding
proteins in the human female germ line, preimplantation embryos,
and embryonic stem cells. Mol Reprod Dev 67(3):323–336
Kaneda M, Okano M, Hata K, Sado T, Tsujimoto N, Li E, Sasaki H
(2004) Essential role for de novo DNA methyltransferase Dnmt3a
in paternal and maternal imprinting. Nature 429(6994):900–903
Ko YG, Nishino K, Hattori N, Arai Y, Tanaka S, Shiota K (2005) Stage-
by-stage change in DNA methylation status of Dnmt1 locus during
mouse early development. J Biol Chem 280(10):9627–9634
Lodde V, Modina SC, Franciosi F, Zuccari E, Tessaro I, Luciano
AM (2009) Localization of DNA methyltransferase-1 during
oocyte differentiation, invitro maturation and early embryonic
development in cow. Eur J Histochem 53(4):199–207
Margot JB, Ehrenhofer-Murray AE, Leonhardt H (2003) Interactions
within the mammalian DNA methyltransferase family. BMC
Mol Biol 4:7
Mertineit C, Yoder JA, Taketo T, Laird DW, Trasler JM, Bestor TH
(1998) Sex-specific exons control DNA methyltransferase in
mammalian germ cells. Development 125(5):889–897
426 J Mol Hist (2017) 48:417–426
1 3
O’Doherty AM, O’Shea LC, Fair T (2012) Bovine DNA methylation
imprints are established in an oocyte size-specific manner, which
are coordinated with the expression of the DNMT3 family pro-
teins. Biol Reprod 86(3):67
Petrussa L, Van de Velde H, De Rycke M (2014) Dynamic regulation
of DNA methyltransferases in human oocytes and preimplanta-
tion embryos after assisted reproductive technologies. Mol Hum
Reprod 20(9):861–874
Ratnam S, Mertineit C, Ding F, Howell CY, Clarke HJ, Bestor TH,
Chaillet JR, Trasler JM (2002) Dynamics of Dnmt1 methyltrans-
ferase expression and intracellular localization during oogenesis
and preimplantation development. Dev Biol 245(2):304–314
Saitou M, Kagiwada S, Kurimoto K (2012) Epigenetic reprogramming
in mouse pre-implantation development and primordial germ
cells. Development 139(1):15–31
Smallwood SA, Kelsey G (2012) De novo DNA methylation: a germ
cell perspective. Trends Genet 28(1):33–42
Turek-Plewa J, Jagodzinski PP (2005) The role of mammalian DNA
methyltransferases in the regulation of gene expression. Cell Mol
Biol Lett 10(4):631–647
Uysal F, Akkoyunlu G, Ozturk S (2015) Dynamic expression of DNA
methyltransferases (DNMTs) in oocytes and early embryos. Bio-
chimie 116:103–113
Verlhac MH, Terret ME (2016) Oocyte Maturation and Development.
F1000Res 5
Yang X, Smith SL, Tian XC, Lewin HA, Renard JP, Wakayama T
(2007) Nuclear reprogramming of cloned embryos and its impli-
cations for therapeutic cloning. Nat Genet 39(3):295–302
Zhang Y, Yang Z, Wu J (2007) Signaling pathways and preimplantation
development of mammalian embryos. FEBS J 274(17):4349–4359
... Finally, we analyzed the expression of molecules involved in epigenetic regulation, which is pivotal in the first stages after fertilization. The DNA methyltransferases (DNMT) are three enzymes that add a methyl group to CpG and non-CpG dinucleotide sites: DNMT1 is essential for the maintenance of methylation and chromatin stability, and DNMT3A and DNMT3B act as de novo methyltransferases, and are important for DNA methylation in the early embryonic stages (Uysal et al., 2017). As reported in Fig. 5 and in Table 3, while DNMT1 was only scarcely affected by the treatments, being modified only by BPS, DNMT3A and DNMT3B were upregulated by BPS alone and downregulated by BPA + PFOA. ...
... These modifications in the expression levels of enzymes responsible for the control of DNA methylation may have important consequences during the early stages of development. In fact, DNMT3A and DNMT3B display remarkable fluctuations from the Germinal Vesicles oocytes to blastocyst stage and keep on modifying in implanted embryos (Uysal et al., 2017), while TETs control the two demethylation waves that characterized the mammalian embryo development: the first follows the fertilization when both maternal and paternal genomes are globally demethylated in zygotes, the second occurs in PGCs, when TETs, especially TET1 and TET2, regulate a crucial set of germline reprogramming-responsive genes, involved in gamete generation and meiosis (Yang et al., 2020). Indeed, mice double knockout for Tet1 and Tet2 display reduced fertility and smaller ovaries in the case of females (Dawlaty et al., 2013). ...
Article
Bisphenols and Perfluoroalkyls are chemical compounds widely used in industry known to be endocrine disruptors (EDs). Once ingested through contaminated aliments, they mimic the activity of endogenous hormones leading to a broad spectrum of diseases. Due to the extensive use of plastic in human life, particular attention should be paid to antenatal exposure to Bisphenols and Perfluoroalkyls since they cross the placental barrier and accumulates in developing embryo. Here we investigated the effects of Bisphenol-A (BPA), Bisphenol-S (BPS), perfluorooctane-sulfonate (PFOS) and perfluorooctanoic-acid (PFOA), alone or combined, on human-induced pluripotent stem cells (hiPSCs) that share several biological features with the stem cells of blastocysts. Our data show that these EDs affect hiPSC inducing a great mitotoxicity and dramatic changes in genes involved in the maintenance of pluripotency, germline specification, and epigenetic regulation. We also evidenced that these chemicals, when combined, may have additive, synergistic but also negative effects. All these data suggest that antenatal exposure to these EDs may affect the integrity of stem cells in the developing embryos, interfering with critical stages of early human development that might be determinant for fertility. The observation that the effects of exposure to a combination of these chemicals are not easily foreseeable further highlights the need for wider awareness of the complexity of the EDs effects on human health and of the social and economic burden attributable to these compounds.
Article
Full-text available
Global methylation levels differ in in vitro- and in vivo-developed embryos. Follicular fluid (FF) contains extracellular vesicles (EVs) containing miRNAs that affect embryonic development. Here, we examined our hypothesis that components in FF affect global DNA methylation and embryonic development. Oocytes and FF were collected from bovine ovaries. Treatment of zygotes with a low concentration of FF induced global DNA demethylation, improved embryonic development, and reduced DNMT1/3A levels. We show that embryos take up EVs containing labeled miRNA secreted from granulosa cells and the treatment of zygotes with EVs derived from FF reduces global DNA methylation in embryos. Furthermore, the methylation levels of in vitro-developed blastocysts were higher than those of in their vivo counterparts. Based on small RNA-sequencing and in silico analysis, we predicted miR-29b, -199a-3p, and -148a to target DNMTs and to induce DNA demethylation, thereby improving embryonic development. Moreover, among FF from 30 cows, FF with a high content of these miRNAs demethylated more DNA in the embryos than FF with a lower miRNA content. Thus, miRNAs in FF play a role in early embryonic development.
Article
Full-text available
DNA methyltransferase 3A (DNMT3A) and its catalytically inactive cofactor DNA methyltransferase 3-Like (DNMT3L) proteins form functional heterotetramers to deposit DNA methylation in mammalian germ cells. While both proteins have an ATRX-DNMT3-DNMT3L (ADD) domain that recognizes histone H3 tail unmethylated at lysine-4 (H3K4me0), the combined and differential roles of the domains in the two proteins have not been fully defined in vivo. Here we investigate DNA methylation landscapes in female and male germ cells derived from mice with loss-of-function amino acid substitutions in the ADD domains of DNMT3A and/or DNMT3L. Mutations in either the DNMT3A-ADD or the DNMT3L-ADD domain moderately decrease global CG methylation levels, but to different degrees, in both germ cells. Furthermore, when the ADD domains of both DNMT3A and DNMT3L lose their functions, the CG methylation levels are much more reduced, especially in oocytes, comparable to the impact of the Dnmt3a/3L knockout. In contrast, aberrant accumulation of non-CG methylation occurs at thousands of genomic regions in the double mutant oocytes and spermatozoa. These results highlight the critical role of the ADD-H3K4me0 binding in proper CG and non-CG methylation in germ cells and the various impacts of the ADD domains of the two proteins.
Article
Amaç: Bu çalışmada, global DNA metilasyonunun Germinal Vezikül (GV) aşamasından Metafaz II (MII) aşamasına kadar olan oosit maturasyonu sürecinde değişim gösterip göstermediğinin ortaya konulması amaçlanmıştır. Yöntem: Bu çalışmada, 4 haftalık Balb/C farelerinin GV ve in vivo MII oosit evreleri arasındaki global DNA metilasyonu farkı immünofloresan yöntemi kullanılarak incelendi. Bu amaçla GV ve MII aşamasındaki oositlerde 5-metil sitozin (5mC) işaretlemesi sonrası Zeiss LSM-880 Airyscan konfokal mikroskopta alınan optik kesitlerinden elde edilen görüntülerden Image-J yazılımı kullanılarak hesaplanan sinyal yoğunlukları değerlendirildi. Bulgular: Global DNA metilasyonu, 5-metil sitozin (5mC) işaretlemesi sonrası değerlendirildiğinde, GV aşamasındaki oositlerde, çekirdek bölgesinde gözlemlenirken, MII aşamasındaki oositlerde metafaz plağına uygun lokasyonda, olduğu izlendi. Global DNA metilasyonunun göreceli sinyal yoğunluğu değerlendirildiğinde; MII aşamasında GV aşamasına göre 3,2 katlık istatistiki olarak anlamlı bir azalma olduğu saptandı. Bu azalışın birinci mayoz sonrası DNA miktarındaki azalmaya bağlı olup olmadığı için yapılan hesaplamalar da bunun sadece DNA miktarındaki azalmadan kaynaklanmadığını gösterdi. Sonuç: Oositlerde, GV aşamasına kıyasla MII evresinde global DNA metilasyon seviyesinin üç kattan daha fazla azalmış olması, fertilizasyon öncesi oositteki DNA metilasyonunun çeşitli mekanizmalarla kontrol edildiğini ve bunun fertilizasyon dinamiğinde önemli olabileceğini göstermiştir.
Article
Background Benzene and its metabolite hydroquinone (HQ) are widely used in daily life, and long‐term exposure to benzene or HQ can induce acute myeloid leukemia (AML). Circular RNAs (circRNAs) are mostly produced by reverse splicing of gene exon mRNA precursors. The modulation of circRNA expression is connected to leukemia progression; however, the molecular mechanism is still unknown. Materials and methods In this study, the cells were divided into four groups: PBS control group (PBS‐TK6), TK6 malignantly transformed cells induced by 10.0 μmol/L HQ (HQ‐TK6), and HQ‐TK6 cells treated with 5 μmol/L 5‐AzaC (DNA methyltransferase inhibitor) for 24 h (HQ + 5‐AzaC). HQ‐TK6 cells were treated with 200 nmol/L TSA (histone deacetylation inhibitor) for 24 h (HQ + TSA). qRT–PCR was used to identify the differential hsa_circ_401351 expression between the four groups. We further determined the hsa_circ_401351 promoter methylation level with methylation‐specific PCR. DNMT1 and DNMT3b were knocked down by CRISPR/Cas9 to elucidate the specific molecular mechanism of hsa_circ_401351 in HQ‐TK6 cells. CCK‐8 and flow cytometry detected cell proliferation and apoptosis, respectively, after hsa_circ_401351 was overexpressed in HQ‐TK6 cells. Results Compared with the PBS‐TK6 group, the expression of hsa_circ_401351 was found to be lower in the HQ‐TK6 group. Nevertheless, treatment with 5‐AzaC or TSA increased hsa_circ_401351 expression, with the upregulation being more pronounced in the TSA group. The expression of hsa_circ_401351 in the DNMT1 knockdown group was dramatically increased by 50% compared to that in the control group, and the DNA methylation level of the hsa_circ_401351 promoter region was decreased. When hsa_circ_401351 was overexpressed, HQ‐TK6 cell proliferation was significantly slowed after 48 h compared with the control group. Flow cytometry showed that cells were mainly arrested in G1 phase, and apoptosis was significantly enhanced. Similarly, qRT–PCR and Western blot data showed significant reductions in Caspase‐3 mRNA and protein production, and Bcl‐2 mRNA levels were also elevated. Conclusions Overall, our research showed that elevated DNMT1 expression in HQ‐TK6 cells increased methylation levels and decreased expression of the hsa_circ_401351 promoter region, limiting its ability to suppress HQ‐TK6 cell growth and enhance apoptosis.
Article
Full-text available
Early diets in honeybees have effects on epigenome with consequences on their phenotype. Depending on the early larval diet, either royal jelly (RJ) or royal worker, 2 different female castes are generated from identical genomes, a long-lived queen with fully developed ovaries and a short-lived functionally sterile worker. To generate these prominent physiological and morphological differences between queen and worker, honeybees utilize epigenetic mechanisms which are controlled by nutritional input. These mechanisms include DNA methylation and histone post-translational modifications, mainly histone acetylation. In honeybee larvae, DNA methylation and histone acetylation may be differentially altered by RJ. This diet has biologically active ingredients with inhibitory effects on the de novo methyltransferase DNMT3A or the histone deacetylase 3 HDAC3 to create and maintain the epigenetic state necessary for developing larvae to generate a queen. DNMT and HDAC enzymes work together to induce the formation of a compacted chromatin structure, repressing transcription. Such dialog could be coordinated by their association with other epigenetic factors including the ubiquitin-like containing plant homeodomain (PHD) and really interesting new gene (RING) finger domains 1 (UHRF1). Through its multiple functional domains, UHRF1 acts as an epigenetic reader of both DNA methylation patterns and histone marks. The present review discusses the epigenetic regulation of honeybee’s chromatin and how the early diets in honeybees can affect the DNA/histone modifying types of machinery that are necessary to stimulate the larvae to turn into either queen or worker. The review also looks at future directions in epigenetics mechanisms of honeybees, mainly the potential role of UHRF1 in these mechanisms.
Article
Full-text available
The present study investigates the impact of two endocrine disruptors, namely Bisphenols (BPs) and Perfluoroalkyls (PFs), on human stem cells. These chemicals leach from plastic, and when ingested through contaminated food and water, they interfere with endogenous hormone signaling, causing various diseases. While the ability of BPs and PFs to cross the placental barrier and accumulate in fetal serum has been documented, the exact consequences for human development require further elucidation. The present research work explored the effects of combined exposure to BPs (BPA or BPS) and PFs (PFOS and PFOA) on human placenta (fetal membrane mesenchymal stromal cells, hFM-MSCs) and amniotic fluid (hAFSCs)-derived stem cells. The effects of the xenobiotics were assessed by analyzing cell proliferation, mitochondrial functionality, and the expression of genes involved in pluripotency and epigenetic regulation, which are crucial for early human development. Our findings demonstrate that antenatal exposure to BPs and/or PFs may alter the biological characteristics of perinatal stem cells and fetal epigenome, with potential implications for health outcomes at birth and in adulthood. Further research is necessary to comprehend the full extent of these effects and their long-term consequences.
Article
Full-text available
Somatic cell nuclear transfer (SCNT) can be used to reprogram differentiated somatic cells to a totipotent state but has poor efficiency in supporting full-term development. H3K9me3 is considered to be an epigenetic barrier to zygotic genomic activation in 2-cell SCNT embryos. However, the mechanism underlying the failure of H3K9me3 reprogramming during SCNT embryo development remains elusive. Here, we perform genome-wide profiling of H3K9me3 in cumulus cell-derived SCNT embryos. We find redundant H3K9me3 marks are closely related to defective minor zygotic genome activation. Moreover, SCNT blastocysts show severely indistinct lineage-specific H3K9me3 deposition. We identify MAX and MCRS1 as potential H3K9me3-related transcription factors and are essential for early embryogenesis. Overexpression of Max and Mcrs1 significantly benefits SCNT embryo development. Notably, MCRS1 partially rescues lineage-specific H3K9me3 allocation, and further improves the efficiency of full-term development. Importantly, our data confirm the conservation of deficient H3K9me3 differentiation in Sertoli cell-derived SCNT embryos, which may be regulated by alternative mechanisms.
Article
Full-text available
Sexual reproduction is essential for many organisms to propagate themselves. It requires the formation of haploid female and male gametes: oocytes and sperms. These specialized cells are generated through meiosis, a particular type of cell division that produces cells with recombined genomes that differ from their parental origin. In this review, we highlight the end process of female meiosis, the divisions per se, and how they can give rise to a functional female gamete preparing itself for the ensuing zygotic development. In particular, we discuss why such an essential process in the propagation of species is so poorly controlled, producing a strong percentage of abnormal female gametes in the end. Eventually, we examine aspects related to the lack of centrosomes in female oocytes, the asymmetry in size of the mammalian oocyte upon division, and in mammals the direct consequences of these long-lived cells in the ovary.
Article
Full-text available
DNA methylation is a key epigenetic modification which is essential for normal embryonic development. Major epigenetic reprogramming takes place during gametogenesis and in the early embryo; the complex DNA methylation patterns are established and maintained by DNA methyltransferases (DNMTs). However the influence of assisted reproductive technologies (ART) on DNA methylation reprogramming enzymes has predominantly been studied in mice and less so in human oocytes and embryos. The expression and localization patterns of the four known DNMTs were analysed in human oocytes and IVF/ICSI embryos by immunocytochemistry and compared between a reference group of good quality fresh embryos and groups of abnormally developing embryos or embryo groups after cryopreservation. In humans, DNMT1o rather than DNMT1s seems to be the key player for maintaining methylation in early embryos. DNMT3b, rather than DNMT3a and DNMT3L, appears to ensure global DNA remethylation in the blastocysts before implantation. DNMT3L, an important regulator of maternal imprint methylation in mouse, was not detected in human oocytes (GV, MI and MII stage). Our study confirms the existence of species differences for mammalian DNA methylation enzymes. In poor quality fresh embryos, the switch towards nuclear DNMT3b expression was delayed and nuclear DNMT1, DNMT1s and DNMT3b expression was less common. Compared to the reference embryos, a smaller number of cryopreserved embryos showed nuclear DNMT1, while a delayed switch to nuclear DNMT3b and an extended DNMT1s temporal expression pattern were also observed. The spatial and temporal expression patterns of DNMTs seem to be disturbed in abnormally developing embryos and in embryos that have been cryopreserved. Further research must be performed in order to understand whether the potentially disturbed embryonic DNMT expression after cryopreservation has any long-term developmental consequences.
Article
Full-text available
There is species divergence in control of DNA methylation during preimplantation development. The exact pattern of methylation in the bovine embryo has not been established nor has its regulation by gender or maternal signals that regulate development such as colony stimulating factor 2 (CSF2). Using immunofluorescent labeling with anti-5-methylcytosine and embryos produced with X-chromosome sorted sperm, it was demonstrated that methylation decreased from the 2-cell stage to the 6-8 cell stage and then increased thereafter up to the blastocyst stage. In a second experiment, embryos of specific genders were produced by fertilization with X- or Y-sorted sperm. The developmental pattern was similar to the first experiment, but there was stage × gender interaction. Methylation was greater for females at the 8-cell stage but greater for males at the blastocyst stage. Treatment with CSF2 had no effect on labeling for DNA methylation in blastocysts. Methylation was lower for inner cell mass cells (i.e., cells that did not label with anti-CDX2) than for trophectoderm (CDX2-positive). The possible role for DNMT3B in developmental changes in methylation was evaluated by determining gene expression and degree of methylation. Steady-state mRNA for DNMT3B decreased from the 2-cell stage to a nadir for D 5 embryos >16 cells and then increased at the blastocyst stage. High resolution melting analysis was used to assess methylation of a CpG rich region in an intronic region of DNMT3B. Methylation percent decreased between the 6-8 cell and the blastocyst stage but there was no difference in methylation between ICM and TE. Results indicate that DNA methylation undergoes dynamic changes during the preimplantation period in a manner that is dependent upon gender and cell lineage. Developmental changes in expression of DNMT3B are indicative of a possible role in changes in methylation. Moreover, DNMT3B itself appears to be under epigenetic control by methylation.
Article
Full-text available
Complementary sets of genes are epigenetically silenced in male and female gametes in a process termed genomic imprinting. TheDnmt3L gene is expressed during gametogenesis at stages where genomic imprints are established. Targeted disruption ofDnmt3L caused azoospermia in homozygous males, and heterozygous progeny of homozygous females died before midgestation. Bisulfite genomic sequencing of DNA from oocytes and embryos showed that removal of Dnmt3L prevented methylation of sequences that are normally maternally methylated. The defect was specific to imprinted regions, and global genome methylation levels were not affected. Lack of maternal methylation imprints in heterozygous embryos derived from homozygous mutant oocytes caused biallelic expression of genes that are normally expressed only from the allele of paternal origin. The key catalytic motifs characteristic of DNA cytosine methyltransferases have been lost from Dnmt3L, and the protein is more likely to act as a regulator of imprint establishment than as a DNA methyltransferase.
Article
Full-text available
A subset of genes, known as imprinted genes, is present in the mammalian genome. Genomic imprinting governs the monoallelic expression of these genes, depending on whether the gene was inherited from the sperm or the egg. This parent-of-origin specific gene expression is generally dependent on the epigenetic modification, DNA methylation, and the DNA methylation status of CpG dinucleotides residing in loci known as differentially methylated regions (DMRs). The enzymatic machinery responsible for the addition of methyl (-CH(3)) groups to the cytosine residue in the CpG dinucleotides are known as DNA methyltransferases (DNMTs). Correct establishment and maintenance of methylation patterns at imprinted genes has been associated with placental function and regulation of embryonic/fetal development. Much work has been carried out on imprinted genes in mouse and human; however, little is known about the methylation dynamics in the bovine oocyte. The primary objective of the present study was to characterize the establishment of methylation at maternally imprinted genes in bovine growing oocytes and to determine if the expression of the bovine DNMTs-DNMT3A, DNMT3B, and DNMT3L-was coordinated with DNA methylation during oocyte development. To this end, a panel of maternally imprinted genes was selected (SNRPN, MEST, IGF2R, PEG10, and PLAGL1) and putative DMRs for MEST, IGF2R, PEG10, and PLAGL1 were identified within the 5' regions for each gene; the SNRPN DMR has been reported previously. Conventional bisulfite sequencing revealed that methylation marks were acquired at all five DMRs investigated in an oocyte size-dependent fashion. This was confirmed for a selection of genes using pyrosequencing analysis. Furthermore, mRNA expression and protein analysis revealed that DNMT3A, DNMT3B, and DNMT3L are also present in the bovine oocyte during its growth phase. This study demonstrates for the first time that an increase in bovine imprinted gene DMR methylation occurs during oocyte growth, as is observed in mouse.
Article
Full-text available
DNA methyltransferase-1 (Dnmt1) is involved in the maintenance of DNA methylation patterns and is crucial for normal mammalian development. The aim of the present study was to assess the localization of Dnmt1 in cow, during the latest phases of oocyte differentiation and during the early stages of segmentation. Dnmt1 expression and localization were assessed in oocytes according to the chromatin configuration, which in turn provides an important epigenetic mechanism for the control of global gene expression and represents a morphological marker of oocyte differentiation. We found that the initial chromatin condensation was accompanied by a slight increase in the level of global DNA methylation, as assessed by 5-methyl-cytosine immunostaining followed by laser scanning confocal microscopy analysis (LSCM). RT-PCR confirmed the presence of Dnmt1 transcripts throughout this phase of oocyte differentiation. Analogously, Dnmt1 immunodetection and LSCM indicated that the protein was always present and localized in the cytoplasm, regardless the chromatin configuration and the level of global DNA methylation. Moreover, our data indicate that while Dnmt1 is retained in the cytoplasm in metaphase II stage oocytes and zygotes, it enters the nuclei of 8-16 cell stage embryos. As suggested in mouse, the functional meaning of the presence of Dnmt1 in the bovine embryo nuclei could be the maintainement of the methylation pattern of imprinted genes. In conclusion, the present work provides useful elements for the study of Dnmt1 function during the late stage of oocyte differentiation, maturation and early embryonic development in mammals.
Article
Combating parasitic DNA by methylation DNA methylation plays an important role in repressing the expression of “parasitic” DNAs, such as transposable elements, which have invaded our genomes. Mammals have three DNA methyltransferase enzymes. Barau et al. discovered a fourth DNA methyltransferase enzyme in mice. The enzyme DNMT3C is a duplication of DNMT3B and is found in male germ cells. There it targets evolutionarily young transposons, of which there is a heavy burden in the mouse genome. DNMT3C methylates and silences the young transposons, preserving male fertility. Science , this issue p. 909
Article
Epigenetic mechanisms play critical roles in oogenesis and early embryo development in mammals. One of these epigenetic mechanisms, DNA methylation is accomplished through the activities of DNA methyltransferases (DNMTs), which are responsible for adding a methyl group to the fifth carbon atom of the cytosine residues within cytosine-phosphate-guanine (CpG) and non-CpG dinuclotide sites. Five DNMT enzymes have been identified in mammals including DNMT1, DNMT2, DNMT3A, DNMT3B, and DNMT3L. They function in two different methylation processes: maintenance and de novo. For maintenance methylation, DNMT1 preferentially transfers methyl groups to the hemi-methylated DNA strands following DNA replication. However, for de novo methylation activities both DNMT3A and DNMT3B function in the methylation of the unmodified cytosine residues. Although DNMT3L indirectly contributes to de novo methylation process, DNMT2 enables the methylation of the cytosine 38in the anticodon loop of aspartic acid transfer RNA and does not methylate DNA. In this review article, we have evaluated and discussed the existing published studies to characterize the spatial and temporal expression patterns of the DNMTs in mouse, bovine and human oocytes and early embryos. We have also reviewed the effects of in vitro culture conditions (serum abundance and glucose concentration), aging, superovulation, vitrification, and somatic cell nuclear transfer technology on the dynamics of DNMTs. Copyright © 2015 Elsevier B.V. and Société FranÇaise de Biochimie et Biologie Moléculaire (SFBBM). All rights reserved.
Article
Epigenetic modifications are crucial for the identity and stability of cells, and, when aberrant, can lead to disease. During mouse development, the genome-wide epigenetic states of pre-implantation embryos and primordial germ cells (PGCs) undergo extensive reprogramming. An improved understanding of the epigenetic reprogramming mechanisms that occur in these cells should provide important new information about the regulation of the epigenetic state of a cell and the mechanisms of induced pluripotency. Here, we discuss recent findings about the potential mechanisms of epigenetic reprogramming, particularly genome-wide DNA demethylation, in pre-implantation mouse embryos and PGCs.