ArticlePDF AvailableLiterature Review

Wang H, Dey SK. Roadmap to embryo implantation: clues from mouse models. Nat Rev Genet 7: 185-199

Authors:
  • School of Medicine, Xiamen University, Xiamen China

Abstract

Implantation involves an intricate discourse between the embryo and uterus and is a gateway to further embryonic development. Synchronizing embryonic development until the blastocyst stage with the uterine differentiation that takes place to produce the receptive state is crucial to successful implantation, and therefore to pregnancy outcome. Although implantation involves the interplay of numerous signalling molecules, the hierarchical instructions that coordinate the embryo-uterine dialogue are not well understood. This review highlights our knowledge about the molecular development of preimplantation and implantation and the future challenges of the field. A better understanding of periimplantation biology could alleviate female infertility and help to develop novel contraceptives.
© 2006 Nature Publishing Group
Departments of Pediatrics,
Cell & Developmental
Biology, and Pharmacology,
Division of Reproductive and
Developmental Biology,
Vanderbilt University Medical
Center, Nashville, Tennessee
37232, USA.
Correspondence to S.K.D.
e-mail: sk.dey@vanderbilt.edu
doi:10.1038/nrg1808
Blastocyst
An embryonic stage in
mammals that is derived from
a morula and is comprised of a
fluid-filled cavity (blastocoel)
and two cell types, the inner cell
mass and the trophectoderm.
Roadmap to embryo implantation:
clues from mouse models
Haibin Wang and Sudhansu K. Dey
Abstract | Implantation involves an intricate discourse between the embryo and uterus and
is a gateway to further embryonic development. Synchronizing embryonic development
until the blastocyst stage with the uterine differentiation that takes place to produce the
receptive state is crucial to successful implantation, and therefore to pregnancy outcome.
Although implantation involves the interplay of numerous signalling molecules, the
hierarchical instructions that coordinate the embryo–uterine dialogue are not well
understood. This review highlights our knowledge about the molecular development of
preimplantation and implantation and the future challenges of the field. A better
understanding of periimplantation biology could alleviate female infertility and help to
develop novel contraceptives.
The implantation of the blastocyst into the maternal
uterus is a crucial step in mammalian reproduction
and, like many developmental processes, it involves
an intricate succession of genetic and cellular interac-
tions, all of which must be executed within an optimal
time frame. In mammals, the beginning of new life is
seeded at fertilization. The fertilized egg undergoes
many cell divisions to form a blastocyst (
BOX 1, part a).
These developmental events are synchronized with the
proliferation and differentiation of specific uterine cell
types, primarily under the direction of ovarian oestro-
gen and progesterone (P
4
). These hormones make the
uterus conducive (‘receptive’) to accept a blastocyst for
implantation
1–4
(BOX 2).
A reciprocal interaction between the blastocyst and
receptive uterus is essential for implantation. Early preg-
nancy loss in humans, which often occurs due to defects
that occur before, during or immediately after implan-
tation, is a worldwide social and economic concern.
Although the human population is growing rapidly and
will probably reach nine billion by 2050, 15% of couples
worldwide are childless because of infertility. Many under-
lying causes of human infertility have been overcome by
in vitro fertilization and embryo-transfer techniques;
implantation rates, however, remain disappointingly
low, probably owing to embryos being transferred into
a nonreceptive uterus. There is, therefore, a continuing
need to unravel the complexities of preimplantation
embryonic development and implantation to address
two contrasting global issues: to improve infertility,
and to develop novel contraceptives.
Here, we focus on the molecular and genetic mecha-
nisms of implantation that have been gleaned primarily
from mouse models, and which could be relevant to
humans. We describe the signalling networks that direct
preimplantation embryonic development, confer blas-
tocyst competency and uterine receptivity to implanta-
tion, instigate the blastocyst–uterine dialogue at various
phases of implantation, and finally, participate in orient-
ing the embryo–uterine axis during the postimplanta-
tion period. The clinical implications of these findings
are also illustrated.
Revisiting this field is timely because of the emergence
of technological advances that allow us, for example, to
profile global gene and protein expression in the embryo
and uterus, and to predict how molecules interact during
implantation. Many gene-knockout mouse models have
also provided a wealth of information that needs to be
carefully interpreted in addressing human fertility.
Although the cellular events that define the various
stages of implantation have been described
1,5
, the molecu-
lar genetic pathways that are crucial to this process, and
how they interact, are not clearly understood. Because
this complex process varies across species (
BOX 1, part b)
1
,
the formulation of a unified model for the molecular basis
of implantation in mammals seems unrealistic at present.
However, inroads can be made by addressing a few crucial
questions to determine: which signalling pathways are
crucial; which are complementary or antagonistic; and
how these pathways are coordinated. Another challenge
is to identify the signalling pathways that have a limited
role during normal pregnancy, but that become important
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
185
© 2006 Nature Publishing Group
Oviduct
E0
E1.0
E1.5
E2.0
E2.5
E3.0
2 cells
4 cells
8 to 16-cell
compacted
morula
Early
blastocyst
Late
blastocyst
Implantation
Uterus
Oocyte
Ovulation
Fertilized
egg
(zygote)
Fertilization
E0.5
Zona pellucida
En
En
T
S
D
ZP
En
ICM
T
S
LE
LE
LE
LE
T
En
ICM
T
E4.0
Trophectoderm
Epiblast
Primitive endoderm
Blastocoel
Inner cell mass
Ovary
8-cell
uncompacted
embryo
a
b
RabbitGuinea pig PrimateMouse and rat
under conditions of stress. Answers to these questions
might help to improve fertility and fertility-associated
issues in
women.
Preimplantation embryonic development
The development of the mammalian preimplantation
embryo encompasses the period from fertilization to
implantation. This period is marked by three princi-
pal transitions, all of which involve dynamic genetic
programming: fertilization and the first cell division;
continued cell division; the establishment of cell polarity
and
compaction to form a morula; and lineage differentiation
to form a blastocyst (followed by implantation).
At the blastocyst stage, embryos mature and escape
from their outer shells
(zona pellucidae) and then gain
implantation competency. The mature blastocyst
is composed of three cell types: the outer epithelial
trophectoderm (Tr), the primitive endoderm (PE) and
the pluripotent
inner cell mass (ICM) (BOX 1, part a). The
ICM generates future cell lineages of the embryo proper,
Box 1 | Preimplantation and implantation events
a
| Preimplantation embryo development and implantation in mice. Following fertilization in the oviduct, the embryo
undergoes several rounds of mitotic cell division, ultimately forming a ball of cells called a morula. At the late morula
stage, the embryo enters the uterine lumen and transforms into a blastocyst that contains a cavity (called blastocoel) with
two distinct cell populations, the inner cell mass (ICM) and the trophectoderm (the progenitor of trophoblast cells).
Before implantation, the blastocyst escapes from its outer shell (the zona pellucida) and differentiates to produce
additional cell types — the epiblast and the primitive endoderm. At this stage, the trophectoderm attaches to the uterine
lining to initiate the process of implantation. E, embryonic day.
b | Implantation strategies in different species. The main
purpose of implantation is to ensure that trophoblast cells firmly anchor into the endometrial stroma. Ultrastructural
studies have revealed that there are different modes of implantation in mammals : the trophectoderm-derived
trophoblast (T) cells can breach the uterine luminal epithelium, coalesce with it or trespass between the uterine cells to
home in on the underlying stroma. In mice and rats, the attachment of the blastocyst (represented by En, the embryonic
endoderm) to the luminal epithelium (LE) imparts epithelial apoptosis locally at the site of attachment, facilitating the
penetration of trophoblast cells through the LE layer into the stroma (S). In guinea pigs, the
syncytial trophoblast makes
focal protrusions through the zona pellucida (ZP) and intrudes between epithelial cells, ultimately embedding the
embryo in the uterine stroma.
In rabbits, clusters of trophoblast cells (trophoblastic knobs) fuse with the LE (T–LE fusion)
to form symplasma. In primates, the syncytial trophoblast is formed near the ICM, which intrudes between uterine
epithelial cells and penetrates the basal lamina. D, decidual cells. Part
a is adapted with permission from REF. 88 © (2001)
Terese Winslow. Part
b is adapted with permission from REF. 1 © (2000) Elsevier Science.
Compaction
An embryonic state in which
the cells of the morula are
flattened and cell outlines
are not clearly distinguishable.
Morula
A cluster of blastomeres that
results from the early cleavages
of a zygote.
Zona pellucida
An outer shell composed of
glycoproteins that encircles
oocytes or preimplantation
embryos.
Trophectoderm
The outer layer of the
blastocyst that is the progenitor
of future trophoblast cell types.
Inner cell mass
Cells that are present inside
the blastocyst. These cells are
pluripotent and give rise to the
embryo proper (that is, the
cells that are not destined to
become the placenta).
Syncytial trophoblast
The syncytial multinucleated
outer layer of the trophoblast.
REVIEWS
186
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
Proestrus
–14 –7 0 7 10 14
LH
LH
Pre-receptive
Pre-receptive
Proliferative phase Secretory phase
Receptive Refractory
P
4
E
2
P
4
E
2
Mouse
Human
Day of pregnancy or pseudopregnancy
Day of cycle
Receptive Refractory
123456
mRNA differential display
A technique for detecting
genes that are expressed only
under specific conditions; it
involves isolating mRNA from
two or more cell populations
and comparing their transcript-
expression levels.
Pseudopregnancy
A condition similar to
pregnancy, without the
presence of a fertilized egg,
which is produced by sterile
mating or hormone treatment.
while the Tr makes the first physical and physiological
connection with the luminal epithelium (LE) of the
maternal uterus for implantation.
Fertilization and first cell division: maternal to zygotic
expression. A unique feature of preimplantation embry-
onic development is the presence of maternally stored
RNAs and proteins in mature, unfertilized eggs. In mice,
fertilization triggers the degradation of oocyte-stored
transcripts, which is 90% complete by the 2-cell stage
6
.
Transcription from the newly formed zygotic genome,
known as zygotic genome activation (ZGA), establishes
the gene-expression patterns that are required for con-
tinued development. A comprehensive molecular charac-
terization of the developmental reprogramming from
maternal to zygotic gene expression has been hindered by
the scarcity of embryonic tissues and lack of appropriate
molecular approaches. Conventional methods, such as
reverse-trancriptase PCR, western blotting and immuno-
histochemistry have been used to examine the expression
patterns of a limited number of genes, but not the dynamic
changes that occur during early development. To assess
more robust gene-expression patterns, high-resolution 2D
protein gels
7, 8
, mRNA differential display
9
and the analysis
of expressed sequence tags (ESTs) that were derived from
libraries of several preimplantation stages
10
were carried
out. However, these studies provided information on
signature transcripts and/or proteins, but not global gene
expression or proteome profiles.
Recently, global gene-expression profiles that were
derived from microarray experiments have gener-
ated a comprehensive data set that covers nearly
all mouse genes during preimplantation develop-
ment
11–13
. One remarkable finding is the existence
of programmed waves of upregulated and downregu-
lated gene expression, which parallels the stages of
embryonic development. According to Hamatani et al.
11
,
maternal-to-zygotic gene activation shows two
principal transient waves of de novo transcription.
The first wave corresponds to the ‘major ZGA, which
peaks between the 2- and 4-cell stages and leads to
the most marked genetic reprogramming. The second
wave, mid-preimplantation gene activation (MGA),
peaks at the 8-cell stage and precedes the morula-to-
blastocyst formation
(FIG. 1a)
11
; indeed, MGA involves
the expression of intercellular adhesion molecules dur-
ing blastomere polarity and compaction. Irrespective of
the underlying mechanisms, the identification of zygotic
transcription cascades that occur at each successive
phase (minor ZGA>major ZGA>MGA;
FIG. 1a) is the
first step towards analysing the complex gene regulatory
network that governs embryonic development.
Studies of transgenic mice show that many genes have
vital functions in preimplantation embryonic devel-
opment, and that their functions, as inferred by gene
targeting, are consistent with their gene-expression pro-
files (see
Supplementary information S1 (table); FIG. 1a).
For example, the maternal-effect gene Mater is detected
Box 2 | The window of uterine receptivity in mice and humans
In placental mammals, the uterus differentiates into an altered state when
implantation-competent blastocysts are ready to initiate implantation. This
state is called uterine receptivity for implantation and lasts for a limited
time. At this stage, the uterine environment is conducive to blastocyst
growth, attachment and the subsequent events of implantation. The major
hormones that specify uterine receptivity are the ovarian steroids
progesterone (P
4
) and oestrogen (E
2
). In mice, the oestrous cycle is short
(~4 days) and often irregular. Therefore, it is difficult to determine the
receptive phase during the cycle. Blastocyst transfers in pseudopregnant
recipients were used to determine various phases of uterine sensitivity to
implantation. In contrast, the menstrual cycle in women is long and the
hormonal changes are more predictable, which allows the state of uterine
receptivity to be determined. A surge of leutinizing hormone (LH), which is
secreted from the pituitary, is essential to ovulation and in programming
the secretion of oestrogen and progesterone by the ovary.
Uterine sensitivity to implantation is classified into prereceptive,
receptive and nonreceptive (refractory) phases. During the prereceptive
phase, the uterus is unable to initiate implantation, but the uterine
environment is less hostile to blastocyst survival. In contrast, during the
refractory phase, the uterine environment is unfavourable to blastocyst
survival. In mice (top diagram), the uterus is receptive on day 4 of
pregnancy or
pseudopregnancy, whereas it is prereceptive on days 1–3,
and by the afternoon of day 5 it becomes nonreceptive (refractory) to
implantation.
In humans (bottom diagram), the uterus is classified histologically and
functionally into proliferative (follicular) and secretory (luteal) phases
during the average 28–30-day menstrual cycle. During the secretory phase,
the uterus is considered prereceptive for the first ~7 days following
ovulation (day 0). The uterus then becomes receptive during the mid-
secretory phase, which spans 7–10 days after ovulation; the nonreceptive
(refractory) phase comprises the rest of the secretory phase.
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
187
© 2006 Nature Publishing Group
Tr
Unfertilised 1-cell 2-cell 4-cell 8-cell Morula Blastocyst
Degradation of
maternal mRNAs
Embryonic–abembryonic axis
Inner cell mass
(pluripotent)
Precursor
(totipotent)
Nanog
Gata6
Oct4
Cdx2
a
Trophectoderm
(multipotent)
c
ICM
Tr
Primitive endoderm
(multipotent)
Epiblast
(pluripotent)
Minor
ZGA
Major
ZGA
MGA
Morula
compaction
Blastocyst
cavitation
Embryonic region
including ICM cells
Extraembryonic cells
interacting with uteri
during implantation
Boundary
Blastocyst Late blastocystLate morula
b
Cdx2
Oct4/Nanog
Oct4
Gata6
only in mature, unfertilized eggs, and its deletion
restricts development beyond the 2-cell stage
14
. Other
studies have identified additional preimplantation
maternal and zygotic genes (see
Supplementary infor-
mation S1
(table); FIG. 1a). Collectively, these studies
have advanced our knowledge of preimplantation
embryonic development, but a comprehensive under-
standing of embryonic development, especially in
humans, is far from complete.
Cell polarity.
Preimplantation development involves the
transition of totipotent, fertilized eggs to blastocysts that
contain both pluripotent ICM cells and the Tr, which
is the progenitor of the trophoblast. One fundamental
question, discussed in this section, is how the cellular
polarity of the embryonic–abembryonic (Em–Ab) axis
is established with the formation of a blastocyst
(FIG. 1b).
The traditional opinion is that embryonic develop-
ment is symmetrical before implantation because each
blastomere in 8- or 16-cell mouse embryos can produce
an offspring. However, recent studies have revealed
asymmetries in the potential of cells, even at very early
stages
(BOX 3).
Lineage differentiation. Irrespective of the debate about
polarity, the molecular crosstalk that segregates and
differentiates the ICM and Tr lineages in blastocysts
is essential to implantation because it is the Tr that
initiates this process together with the LE. Microarray
and deletion studies have identified several genes that
are crucial for these two cell-lineage segregations (see
Supplementary information S1 (table)), which include
those that encode many transcription factors, such as
OCT4, SOX2, NANOG, CDX2
and Eomesodermin
(EOMES)
15, 16
(FIG. 1b,c).
ICM formation depends on OCT4, which is encoded
by Pou5f1, as Pou5f1-mutant blastocysts contain only Tr.
Pou5f1 is restricted to the ICM at the blastocyst
stage
17
. SOX2, a high-mobility group (HMG)-box
transcription factor, shows a similar expression pro-
file to Pou5f1, and together they prevent trophoblast
specification
18
. However, the inability of OCT4 alone
to maintain
embryonic-stem-cell (ES cell) pluripotency
in the absence of leukaemia inhibitory factor (LIF)
19
indicates that other pluripotency-promoting factors,
such as NANOG (a homeobox (Hox) transcription
factor), are involved. The consensus is that, while both
NANOG and OCT4 are required for ICM specifica-
tion, they suppress the formation of extraembryonic
lineages; OCT4 represses trophoblast and NANOG
parietal–visceral endoderm formation
20,21
. A recent
genome-scale location analysis in human ES cells has
revealed a novel mechanism for establishing pluripo-
tency. It showed that OCT4, SOX2 and NANOG co-
occupy a substantial portion of their target genes and
collaborate to form a circuitry of autoregulatory and
feed-forward loops
22
.
It has been proposed that the Tr develops by default
in the absence of OCT4. However, CDX2, a caudal-type
homeodomain protein, is crucial for segregating ICM
and Tr lineages at the blastocyst stage by ensuring the
Figure 1 | Genes governing the development of the preimplantation mouse
embryo. a | Gene expression during preimplantation embryo development. The
diagram depicts the waves of gene expression that occur in preimplantation embryos,
based on microarray studies, from the degradation of maternal genes, to the three
successive waves of overlapping zygotic gene expression (minor and major ZGA
(zygotic genome activation), and MGA (mid-preimplantation gene activation)) —
following the MGA, genes that are specific to morula compaction and blastocyst
cavitation are also expressed in a wave-like fashion. In addition, many genes that are
expressed in embryos and stem cells are crucial to preimplantation development
(see
Supplementary information S1 (table)). b,c | Genes that are crucial for cell-fate
and lineage segregations during the morula-to-blastocyst transition, as determined by
expression and mutation studies in mice. Panel (b) represents the gene-expression
patterns during blastocyst formation. Whereas Oct4 (dark pink) is expressed throughout
the embryo before the late morula stage, the expression of Nanog (light pink) is
specifically induced in the inside cells of late morulae. Cdx2 (blue) is expressed in the
outer layer of cells in late morulae and is required for the repression of Oct4 and Nanog
in the trophectoderm (Tr) of the blastocyst. Oct4 is crucial for inner-cell-mass (ICM)
formation. Gata6 (green) is expressed in the primitive endoderm of the late blastocyst,
where Oct4 and Nanog are repressed. Panel (c) shows the genetic model of lineage
decision. Oct4 represses Cdx2 expression, which in turn represses Oct4 expression to
allow segregation of the ICM and Tr lineages of the blastocyst. An antagonism between
Nanog and Gata6 segregates epiblast and primitive endoderm within the ICM. Panel
(a) is adapted with permission from
REF. 11 © (2004) Elsevier Science. Panel (b) is
adapted with permission from
REF. 113 © (2005) Blackwell Publishing Ltd.
REVIEWS
188
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
Cavitation
The creation of a hollow space
that appears within the early-
cleaving embryos to form a
blastocyst.
Embryonic stem cells
(ES cells). Stem cells have the
dual capacity to self-replicate
and differentiate into several
specialized derivatives. ES cells
are pluripotent cells that are
derived from pre-implantation-
stage (usually blastocyst)
mammalian embryos. Mouse
ES cells can be propagated
and manipulated in vitro, yet
still retain their pluripotency.
Polar body
The structure that is extruded
from the oocyte during
meiosis, which contains one
haploid set of chromosomes.
Window of implantation
A limited time period when
the uterine environment is
conducive to supporting
blastocyst growth, attachment
and the subsequent events of
implantation.
Delayed implantation
A state of suspended animation
of the blastocyst, characterized
by halted growth and
postponement of implantation.
In mice, ovariectomy on day 4
morning of pregnancy, before
ovarian oestrogen secretion,
initiates blastocyst dormancy,
which can last for many days if
treated with P
4
; an oestrogen
injection rapidly activates
blastocysts and initiates their
implantation.
Blastocyst activation
The event that leads to the
competency of the blastocyst
to implant.
repression of OCT4 and NANOG in the Tr (FIG. 1b,c).
This was shown by the finding that CDX2 deficiency
results in a failure to downregulate
Oct4 and Nanog in
the outer cells of the blastocyst, which results in the
loss of the epithelial integrity of these cells and their
ultimate demise
23
. Another gene that is involved in Tr
development is that encoding the T-box transcription
factor EOMES, which, like
Cdx2, is expressed in the Tr
24
.
However, embryos that lack
Eomes develop to blastocysts
and correctly express Cdx2 and Oct4 in Tr and ICM cells,
respectively
23
. It is suggested that Cdx2 is the earliest
inducer of the Tr lineage in late morulae, whereas Eomes
is required for Tr proliferation and differentiation at the
blastocyst stage.
Determinants of blastocyst competency
For successful implantation to occur in the receptive
uterus, the blastocyst must also attain implantation
competency. The first evidence that the state of activity
of the blastocyst determines the
‘window’ of implantation
in the receptive uterus was derived from reciprocal
blastocyst-transfer experiments in a
delayed-implantation
mouse model
25,26
. This model is a powerful tool for
defining the molecular signalling components that direct
blastocyst activation or dormancy. Nearly 100 mammals in
seven orders undergo delayed implantation
27,28
, but the
underlying mechanism remains largely unknown. Using
this model, a global gene-expression study showed that
these two different physiological states of the blastocyst
are molecularly distinguishable
29
. The main functional
categories of altered genes include cell-cycle, cell-
signalling and energy-metabolic pathways. This study
also showed an upregulated expression of Hegf1 (which
encodes heparin-binding EGF-like growth factor (
HB-
EGF
)) in activated blastocysts, a finding that is comple-
mentary to earlier reports of upregulated expression of
its receptors
ErbB1 and ErbB4 in similar blastocysts
29–31
.
Other signalling molecules also participate in blas-
tocyst dormancy and activation. There is evidence that
catecholoestrogens that are produced from primary
oestrogens in the uterus activate blastocysts
32
. Another
lipid-signalling molecule that targets blastocysts is
an endocannabinoid anandamide, which activates
G-protein-coupled cannabinoid receptors CB1 and CB2.
Expression of
Cb1 in the Tr, and uterine synthesis of anan-
damide, indicate that endocannabinoid signalling is crucial
to implantation in mice
33–35
. Levels of uterine anandamide
and blastocyst CB1 are coordinately downregulated with
the attainment of uterine receptivity and blastocyst acti-
vation, respectively, in contrast to their elevated levels in
the nonreceptive uterus and dormant blastocysts
33,36,37
.
Indeed, implantation is postponed in wild-type mice that
are maintained on sustained levels of exogenously admin-
istered cannabinoid ligands, an effect that depends on the
expression of CB1 receptors on the embryo
37
. Anandamide
regulates blastocyst function by differentially modulating
mitogen-activated protein kinase (MAPK) signalling and
Ca
2+
-channel activity via CB1 (REF. 36). This is consist-
ent with findings that MAPK and phosphatidylinositol
3-kinase/Ca
2+
-signalling cascades are crucial to blastocyst
development and activation
38–41
.
Most gene-expression studies have so far pointed
towards changes in Tr gene expression during blastocyst
dormancy or activation. It remains to be seen whether
gene expression in the ICM also changes with the state
of activity of the blastocyst. A greater insight into the
molecular basis of blastocyst competency for implanta-
tion might help to improve pregnancy rates in human
IVF programs.
Determinants of uterine receptivity
Molecular and genetic evidence indicates that locally
produced signalling molecules, including cytokines,
growth factors, homeobox transcription factors, lipid
mediators and morphogens, together with ovarian
hormones, serve as autocrine, paracrine and juxta-
crine factors to specify uterine receptivity
2
(TABLES 1,2).
In this section, evidence is presented for a novel signal-
ling network that involves cytokines, homeotic proteins
and morphogens in implantation.
Box 3 | Cell polarity in the early embryo: an ongoing debate
Specific labelling of blastomeres or zona pellucidae indicates that the plane of the first cleavage specifies embryonic
polarity. That is, the plane of cleavage is orthogonal to the future embryonic–abembryonic axis of the blastocyst, with one
blastomere predominantly contributing to the embryonic pole (polar trophectoderm (Tr) and deeper inner-cell-mass (ICM)
cells) and the other to the abembryonic pole (mural Tr and more superficial ICM)
89–93
. This indicates a developmental
asymmetry at the 2-cell stage, which is supported by lineage-tracing experiments
94–96
, and by observations that 2-cell
embryos divide asynchronously, with daughter cells making a differential contribution to the future ICM or Tr
97,98
. However,
the observation that both blastomeres in 2-cell embryos make a similar contribution to all cell types during the
postimplantation development of embryos that harbour a Cre-reporter indicates that there is no absolute programming of
lineage segregation for either the ICM or Tr at the 2-cell stage
90
. This is consistent with studies that used fluorescent-tracer
labelling and time-lapse imaging
99–101
. Collective analysis provides evidence that polarity with lineage differentiation is
first clearly discernible with the onset of blastocyst formation
101
.
Another question in this ongoing debate is whether the first cleavage occurs randomly or is prepatterned
102,103
. It was
proposed that the sperm-entry site, along with the second
polar body from the meiotic division, marks the first cleavage
plane
92,104,105
. However, the results of the labelling of internalized sperm components
106
, and the fact that the polar body is
not stationary but can move to the cleft between two blastomeres after cleavage
107
, challenge this view. Time-lapse
recordings show that the first cleavage plane is not predetermined, but is defined by the apposition of two pronuclei at the
centre of the zygote
107
. The use of videomicroscopy to visualize the mitotic spindle using GFP-labelled tubulin also shows
that the cleavage plane is randomly oriented in 2-cell embryos, arguing against prepatterning before embryonic
compaction
108
. Therefore, the question of the origin of cell polarity and lineage differentiation is still under debate.
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
189
© 2006 Nature Publishing Group
Oestrogen and progesterone. The principal hormones
that direct uterine receptivity are ovarian P
4
and oes-
trogen
2
. P
4
is essential for implantation and pregnancy
maintenance in all mammals studied, whereas the
requirement for ovarian oestrogen is species-specific
2
.
For example, ovarian P
4
and oestrogen are essential to
implantation in mice and rats, but ovarian oestrogen is
dispensable in pigs, guinea pigs, rabbits and hamsters.
However, oestrogen that is produced by embryos is
considered important for implantation in these last
four species
2
; whether ovarian or embryonic oestrogen
participates in human implantation remains unknown.
The uterine effects of oestrogen and P
4
are primarily
executed by nuclear oestrogen (ER) and progesterone
Table 1 | Genes implicated in human uterine receptivity for implantation by data from independent microarray analysis
Comparison of five independent studies
Gene Molecule encoded (Putative function) LH+(8–10) vs
LH–(4–6)
60
LH+(7–9) vs
LH+(2–4)
59
LH+7 vs
LH+2
62
LH+(6–8) vs
LH–(3–5)
58
LH+8 vs
LH+3
61
Upregulated
ANXA4 Annexin-4 (SP) + + +
APOD Apolipoprotein D (LP) + + + +
BNIP2 BCL2/adenovirus E1B 19kDa interacting
protein-2 (cell-death protein)
+++
CLDN4 Claudin-4/CEP-R (R) + + +
C1R Complement component-1r (Imm) + + +
DAF* Decay accelerating factor for complement (Imm) + + + +
DF Complement factor D/Adipsin (Imm) + + +
DKK1 Dickkopf-1 (WNT antagonist) + + + +
GADD45A Growth arrest and DNA-damage-inducible
protein (DNA excision repair, cell-cycle regulator)
++++
GBP3 Guanylate-binding protein-2 (GTP-BP) + + +
ID4 Inhibitor DNA binding-4 (transcription
coregulator)
+++
IL15 Interleukin-15 (cytokine) + + + +
MAP3K5 Mitogen activated protein kinase kinase kinase 5
(MAPK signalling)
++++
MT1 Metallothionein-1 family proteins (MBP) + + + + +
MAOA Monoamine oxidase A (catechol-NT
metabolizing enzyme)
++++
PAEP Progestagen-associated endometrial protein
(SecP)
+++
SERPING1 Ser (or Cys), clade G (C1 inhibitor), member 1
(proteolysis inhibitor)
+++
SPP1
Secreted phosphoprotein-1 (StrP) + + + + +
TGFB
TGFβ-super-family proteins
++ +
Downregulated
CCNB Cyclin B proteins (cell-cycle regulator) + + +
FRPHE Frizzled-related protein frpHE (WNT antagonist) + + +
GATA2 GATA-binding protein-2 + + +
MSX1 Hox Msh-like protein-1 + + +
MSX2 Hox Msh-like protein-2 + + +
OLFM1 Olfactomedin-related ER-localized protein-1
(SecP)
++++
Assuming a cycle length of 28–30 days, the surge in the levels of luteinizing hormone (LH) at mid-cycle heralds the onset of ovulation (BOX 2). The comparative
levels of global gene expression in the human uterus
presented here show that only few genes reveal similar changes (up or downregulation) across five
experiments. This poor consistency is perhaps due to changes in the timing of the assay, experimental designs, methods for data analysis, and/or geographical
location where subjects reside or the geographical location of the origin of subjects selected. The experiments compare gene-expression profiles between post-LH (+)
surge versus pre-LH surge (–) or early versus late post-LH surge. The receptive period in humans spans the period from days 7–10 after the LH surge (LH+(7–10)).
Genetic studies in mice might prove fruitful to assess whether these genes are critical to uterine receptivity in humans, since such studies are not possible in
humans except for the identification of mutations of these genes in human populations. *DAF, also known as CD55, Cromer blood-group system.
SPP1, also known
as osteopontin. BP, binding protein; GF, growth factor; Hox, homeobox; Imm, immunomodulator; LP, lipoprotein; MBP, metal-binding protein; NT, neurotransmitter;
R, receptor; SecP, secretory protein; SP, signalling protein; StrP, structural protein; TF, transcription factor.
REVIEWS
190
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
(PR) receptors. The recent discovery of ER (ERα and
ERβ) and PR (PRA and PRB) isoforms and studies of
the effect of their selective deletion provide evidence
for their isoform-specific functions in uterine biology
and implantation.
Er
α
–/–
uteri are hypoplastic and unable
to support implantation
42
, whereas Er
β
–/–
uteri retain
biological functions that allow normal implantation
2
.
Interestingly, P
4
is sufficient for decidualization in Er
α
–/–
mice in response to artificial stimuli, which indicates
that ERα might be essential for blastocyst
attachment,
but dispensable for subsequent decidualization
43,44
. The
uterus expresses
PRA and PRB. While mice that lack both
PRA and PRB show many defects
in ovarian and uter-
ine functions, which leads to female infertility
45
, these
responses are normal in mice that are missing only PRB,
which indicates that essential P
4
-regulated functions are
primarily mediated by PRA
46
.
Cytokines. Among the cytokines, LIF, a member of the
interleukin-6 (IL-6) family, is crucial for uterine prepara-
tion for implantation. It binds to the LIF receptor and
shares
gp130 as a common signal-transduction partner
with other cytokines. In mice,
Lif is expressed first in uter-
ine glands on the morning of day 4, and then in stromal
cells that surround the blastocyst during attachment
47,48
.
This indicates that LIF has a dual role: initially in uterine
preparation and later in attachment. Blastocysts remain
dormant’ in Lif
–/–
mice and do not implant, an effect
that depends on the uterine LIF mutant status
47,48
. The
molecular mechanism by which LIF executes its effects
on implantation is still unclear, although inactivation
of the gp130 protein by deleting its signal transducer
and activator of transcription (STAT) binding sites also
results in implantation failure
49
. Uterine Lif expression is
high around the time of implantation in other species,
including humans
2
.
Homeobox proteins. Several homeobox transcription
factors are crucial to uterine receptivity and implan-
tation. In mice, two Abdominal-B-like Hox genes,
Hoxa10 and Hoxa11, are expressed in uterine stromal
cells during the receptive phase. This expression per-
sists during postimplantation decidualization, which
might indicates an overlapping role for the two genes
in uterine receptivity, implantation and decidualiza-
tion
50–53
. Most Hoxa10
–/–
females are infertile, primarily
due to a reduced stromal-cell proliferation and the
consequent failure to decidualize
50,52
. However, Hoxa10
does not seem crucial for uterine receptivity, because
initial uterine attachment of blastocysts can occur in
Hoxa10
–/–
mice, and Hoxa10 expression is normal in
Lif
–/–
uteri
54
. In contrast, Hoxa11
–/–
uteri are hypoplastic,
have fewer glands and show a more severe phenotype
than Hoxa10
–/–
mice
55
. More importantly, the absence
of Lif expression in Hoxa11
–/–
uteri indicates that
Hoxa11 might be crucial to uterine receptivity and later
events of implantation
55
. Both Hoxa10 and Hoxa11 are
upregulated in the human uterus during the secretory
phase, which indicates that they might have a role in
uterine receptivity
56
. Gene-targeting experiments show
that blastocysts fail to implant in
Hmx3
–/–
mice, but the
reason for this failure remains unknown because Hmx3,
which belongs to a different homeobox gene family to
Hox genes, is mainly expressed in the
myometrium
57
.
Another homeobox gene,
Msx1, is transiently
expressed in the mouse uterine epithelium during the
receptive period, but disappears at the time of blastocyst
attachment or when the uterus enters the nonreceptive
phase
54
. Sustained expression of Msx1 in Lif
–/–
mice fur-
ther reinforces the importance of Msx1 in uterine recep-
tivity. It is interesting that Msx1 is downregulated in the
receptive human
endometrium
58–62
(TABLE 1). However, a
definitive role for Msx1 in uterine receptivity requires
conditional uterine deletion, because offspring that are
missing Msx1 die shortly after birth due to craniofacial
defects
63
.
Morphogens. One less-explored area is the role of
morphogens in uterine receptivity and implantation.
Embryo–uterine interactions during implantation
share many features with reciprocal epithelial–
mesenchymal interactions during embryogenesis, and
both involve evolutionarily conserved signalling path-
ways. The importance of hedgehog (HH), WNT and
bone-morphogenetic-protein (BMP) signalling in uter-
ine receptivity was recently explored. The genes encoding
the components of the HH signalling pathway, namely
Indian hedgehog (
Ihh), HH-binding protein/receptor
Patched (Ptc) and the transcription factors Gli1–3 are
expressed in the mouse uterus
64,65
. Ihh expression is
P
4
-dependent and reaches high levels in epithelial cells
on day 4, while that of Ptc, Gli1 and Gli2 is upregulated in
the underlying stroma. In day 4 uterine-explant cultures,
recombinant N-sonic hedgehog (N-SHH) stimulates
mesenchymal-cell proliferation, a characteristic of the
receptive phase
65
. These findings indicate that epithelial
IHH functions as a paracrine growth factor for stromal
cells and that this epithelial–mesenchymal signalling is
important for uterine receptivity.
The roles of WNT and BMP signalling in preserv-
ing tissue boundaries in the adult uterus remain largely
unknown.
sfrp4, a WNT antagonist and a member of the
secreted Frizzled-related proteins (sFRPs), and
Noggin,
an anti-BMP, are expressed in the uterine stroma during
the receptive phase
66
. Wnt4 and Bmp2 are not expressed
at this time, but are induced in the stroma with the onset
of blastocyst attachment, and thereafter with disappear-
ing expression of the antagonists
54,66
. These findings
indicate that while HH signalling participates in uterine
receptivity, WNT4 and BMP2 are involved in the attach-
ment reaction and postimplantation events.
Why, then, are sfrp4 and Noggin expressed in the
absence of their ligands? Do they have functions that
are independent of their ligands? Are other members
of the ligand family expressed in the uterus? A marked
downregulation of sfrp4 in Lif
–/–
uteri indicates that
a WNT-signalling component is important in uter-
ine preparation
54
. Alternatively, this downregulation
might be a consequence of compromised uterine func-
tion in the absence of Lif. Of the WNT family,
Wnt7a
is expressed in the LE in adult females, and deletion of
the Wnt7a gene shows global posterior shifting of the
Hypoplastic
Refers to an underdeveloped
tissue or organ.
Decidualization
Transformation of stromal cells
into morphologically and
functionally distinct cells. Part
of decidualized tissue is shed
at parturition.
Attachment
A process by which the
blastocyst trophectoderm is
brought into physical and
physiological contact with the
uterine luminal epithelium.
Myometrium
The muscular outer layer of
the uterus, which is comprised
of longitudinal and circular
muscle fibers.
Endometrium
The inner lining of the uterus;
it is primarily comprised of
stromal cells (the supporting
tissue of an organ) and
epithelial cells of both luminal
and glandular types. Part of the
endometrium is shed during
menstruation.
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
191
© 2006 Nature Publishing Group
Table 2 | Genes critical to uterine biology and implantation: results of mouse knockout models
Genes Molecule encoded (Putative function) HomoloGene
No. (NCBI)
Knockout phenotype in females Refs
Uterine patterning during postnatal growth
Hoxa10 Homeobox A10 (TF) 7365 Homeotic transformation of anterior uterus to oviduct 50,53,
114
Hoxa11 Homeobox A11 (TF) 4033 No uterine glands; partial homeotic transformation of
uterus to oviduct
51
Wnt5a WNT-5a protein (SP) 20720 No morphologically
defined cervix; no uterine glands 115
Wnt7a WNT-7a protein (SP) 20969 Abnormal oviduct and uterine development*; infertility 67
Uterine physiology in adult life
Adamts1 A disintegrin-like and metalloprotease with
thrombospondin-type-1 motif-1 (Enzyme, tissue
remodelling
21381 Impaired follicular development and fertilization;
uterine cysts; subfertility
116
Bteb1 Basic transcription-element-binding protein-1 (TF) 931, 79195 Uterine hypoplasia; compromised uterine P
4
function;
impaired embryo implantation; subfertility (ME)
117
Cenpb Centromere protein B (Centromere assembly) 1370 Disrupted luminal and glandular uterine epithelia;
subfertility (genetic-background-dependent)
118
Cyp27b1
25-hydroxyvitamin D 1α–hydroxylase enzyme
(Vitamin D metabolism)
37139 Uterine hypoplasia; absence of corpus luteum;
infertility
119
Esr1
Oestrogen receptor-α (NR,TF)
47906 Ovarian cysts; uterine hypoplasia; infertility 42
Igf1 Insulin-like growth factor-1 (GF) 515 Ovulation failure; uterine myometrial hypoplasia;
infertility
120
Pgr Progesterone receptor (NR,TF) 713 Unopposed oestrogen action; uterine hyperplasia;
infertility
45
Ube3a
Ubiquitin-protein ligase E3A (Protein modification,
proteolysis and peptidolysis)
7988 Impaired follicular development and uterine
hypoplasia; subfertility
121
Vdr Vitamin D receptor (R,TF) 37297 Uterine hypoplasia with impaired folliculogenesis;
infertility
122
Uterine preparation for initiating implantation
Bsg Basigin (Immunoglobulin) 1308, 45225 Defective fertilization; no implantation 123,124
Esr1
Oestrogen receptor-α (NR,TF)
47906 No uterine attachment, but uterine responsiveness to
decidualization persists with P
4
priming
42,44
Fkbp52 FK506-binding protein-4 (Immunophilin co-
chaperone for steroid hormone NRs)
36085, 43060 Compromised P
4
function; no uterine receptivity (ME) 69
Gp130/
Stat
GP130/Signal Ttransducer and activator of
transcription (Cytokine-receptor signalling)
1645 No implantation (ME) 49
Hmx3 H6 homeobox-3 (TF) 40612 No implantation (ME) 57
LpA3 Lysophosphatidic acid receptor-3 (LPA signalling) 8123 Deferred, on-time implantation; aberrant embryo
spacing; postimplantation defects; small litter size (ME)
80
Lif Leukaemia inhibitory factor (Cytokine) 1734 No implantation (ME) 48
Pgr Progesterone receptor (NR,TF) 713 No implantation or decidualization (ME) 45
Pla2g4a Phospholipase A2, group IVA
§
(Arachidonic-acid-
releasing enzyme)
32059 Deferred on-time implantation; aberrant embryo
spacing; postimplantation defects; small litter size (ME)
79
Ppard
Peroxisome proliferator-activated receptor-δ
(NR,TF)
4544 4–6 h delay in initiating embryo attachment; placental
defects; subfertility
76,125
Ptgs2
Prostaglandin-endoperoxide synthase-2
(Prostaglandin synthesis)
31000 Multiple reproductive failures, including defective
attachment reaction; genetic-background-dependent
75,77
Uterine decidualization
Fkbp52 FK506-binding protein-4 (Immunophilin co-
chaperone for steroid hormone NRs)
36085, 43060 Compromised P
4
function; defective decidualization
(ME)
69
Hoxa10 Homeobox A10 (TF) 7365 Defective decidualization; reduced fertility (ME) 50,52,53
Hoxa11 Homeobox A11 (TF) 4033 Defective implantation and decidualization; infertility 51
Il11ra1
Interleukin-11 receptor-α1 (Cytokine signalling)
3316 Impaired decidualization; infertility 126,127
Pgr Progesterone receptor (NR,TF) 713 Lack of decidual response even after P
4
priming 45
Ptgs2
Prostaglandin-endoperoxide synthase-2
(Prostaglandin synthesis)
31000 Defective decidualization; reduced angiogenic
response
75,85
*Also, reduced stromal tissue, lack
of uterine glands and disorganized myometrium.
Ube3a, also known as E6AP ubiquitin-protein ligase.
§
Cytosolic, calcium-
dependent.
Also known as cyclooxygenase-2 (COX2). GF, growth factor; ME, maternal effect; NR, nuclear receptor; R, receptor, SP, signalling protein; TF,
transcription factor.
REVIEWS
192
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
M
Blastocyst
activation
Tr
HB-EGF
ErbB
PRA
PRA
LPA3
Adhesion
molecules
ErbB1/4
CB1
S
LE
GE
V
MYO
ERα
LE
Em
AM
ZP
COX2
cPLA2α
PPARδ
WNT4
BMP2
LIF
FKBP52
FKBP52
LIF
AttachmentApposition
b
Postimplantation
a
Bmp7; Dan; Crim1
Fgf2; sFRP4
Noggin; Fgf10
Wnt4
Bmp2
ICM
ICM
Basal lamina
A thin sheet of proteoglycans
and glycoproteins that are
secreted by cells as an
extracellular matrix. It is also
called the basement membrane
and influences cell polarity,
differentiation and migration.
Decidual cells
In the mouse, the cells that
surround the implanting
blastocyst.
Oedema
Fluid accumulation in the
intercellular tissue spaces.
Luminal closure
The closure of the uterine
lumen, resulting in closer
contact between the luminal
epithelial linings; this step is
essential for blastocyst
attachment.
reproductive tract, with the loss of Hoxa10 and Hoxa11.
Wnt7a
–/–
females are infertile, with uteri that lack glands
and disorganized myometria, which indicates that Wnt7a
might be crucial for normal uterine cellular architec-
ture
67
. Of the Bmp genes that have been studied, Bmp4–7
and 8a do not show the same highly localized expression
pattern as Bmp2 during attachment
66
. An investigation
that spans other members of the WNT and BMP fami-
lies, their receptors and putative antagonists is warranted
to better understand the roles of these morphogens in
uterine receptivity.
Signalling during implantation
The process of implantation is classified into three
stages: apposition, attachment (adhesion) and penetra-
tion. During apposition, the Tr becomes closely apposed
to the LE. This is followed by the attachment stage, when
the association of the Tr and LE is sufficiently intimate to
resist dislodging of the blastocyst by flushing the uterine
lumen. The first sign of the attachment reaction occurs
on the evening of day 4 in mice, and coincides with a
localized increase in stromal vascular permeability at
the site of blastocyst attachment. Penetration involves
invasion of the embryo through the LE and
basal lamina
into the stroma, to establish a vascular relationship with
the mother. At this stage, stromal-cell differentiation into
decidual cells (decidualization) is extensive and leads to
the loss of the LE at the site of the implanting blastocyst.
(Note, however, that stromal-cell decidualization also
occurs in women during the luteal phase of the men-
strual cycle, in the absence of an embryo.) The dynamic
and overlapping expression of signalling molecules dur-
ing these three stages makes it difficult to assign the con-
tribution of specific signalling pathways to a particular
stage (
FIG. 2; TABLE 2).
Apposition. In rodents, a generalized stromal
oedema
leads to uterine
luminal closure, resulting in interdigita-
tion of microvilli of the Tr and LE (apposition). Luminal
closure occurs in pregnant or pseudopregnant uteri, and
therefore does not require the presence of blastocysts. P
4
priming, however, is essential for closure. This is sup-
ported by the absence of luminal closure in pregnant mice
that are missing FK506 binding protein-4 (FKBP52), a
co-chaperone that is required for appropriate uterine
PR function
68,69
. Fkbp52 expression overlaps with that
of PR in the stroma before the attachment reaction,
and Fkbp52
–/–
females show implantation failure and
Figure 2 | Gene products participating in embryo implantation. a | Signalling pathways that are known to coordinate
blastocyst apposition and attachment in the mouse uterus. Apposition and attachment are key steps in implantation and
absolutely depend on the synchronized development of the blastocyst to implantation competency and differentiation
of the uterus to the receptive stage. Ovarian oestrogen and progesterone, acting through their cognate nuclear
receptors, influence several locally produced growth factors, adhesion molecules, cytokines, transcription factors and
vasoactive mediators and their receptors in the uterus and/or blastocyst to coordinate blastocyst–uterine crosstalk. This
crosstalk further influences some of the signalling pathways to ensure the successful execution of the implantation
process. b | Region-specific expression patterns of morphogens in the mouse deciduum during the postimplantation
period. This scheme is based on in situ hybridization of the indicated genes in a representative cross-section of an
implantation chamber on day 7 of pregnancy. AM, antimesometrial pole; BMP2, bone morphogenetic protein-2; CB1,
brain-type cannabinoid receptor-1; COX2, cyclooxygenase-2; cPLA2α, cytosolic phospholipase A
2α
; Crim1, cysteine-rich
transmembrane BMP-regulator-1; Dan, differential screening-selected gene aberrative in neuroblastoma; Em, embryo;
ERα, nuclear oestrogen receptor-α; ErbB, EGF-receptor family; FGF, fibroblast growth factor; FKBP52, FK506 binding
protein-4; GE, glandular epithelium; HB-EGF, heparin-binding EGF-like growth factor; ICM, inner cell mass; LE, luminal
epithelium; LIF, leukaemia inhibitory factor; LPA3, lysophosphatidic-acid receptor-3; M, mesometrial pole; MYO,
myometrium; PPARδ, peroxisome-proliferator-activated receptor-δ; PRA; nuclear progesterone receptor A; S, stroma;
sFRP4, secreted Frizzled-related protein-4; Tr, trophectoderm; V, blood vessels; ZP, zona pellucida.
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
193
© 2006 Nature Publishing Group
Integrins
A family of receptors for
various extracellular-matrix
ligands that modulate cell–cell
adhesion and signal
transduction. Each integrin has
two subunits, α and β, and
each αβ combination has a
unique binding specificity and
unique signalling properties.
Selectins
A group of cell-adhesion
molecules, including L-selectin,
E-selectin and P-selectin, that
bind to carbohydrates.
Galectins
A family of lectins with
galactose-binding ability.
Trophinin–tastin–bystin
complex
A homophilic cell-adhesion
complex that is comprised of
membrane–cytoplasmic
proteins.
Prostaglandins
(PG). Vasoactive lipid mediators
that are implicated in various
pathophysiological processes,
including vascular permeability,
angiogenesis and cell migration.
downregulation of the P
4
-responsive genes Areg (which
encodes amphiregulin), Hoxa10 and Ihh in the uterus.
However, although P
4
priming via PR is essential for
luminal closure and apposition, blastocyst attachment
cannot occur unless the P
4
-primed uterus is exposed
to oestrogen.
The signalling pathway that is initiated by HB-EGF has
been studied extensively during apposition and attach-
ment because HB-EGF is an early molecular marker of
embryo–uterine crosstalk
70
. Heg f1 is expressed in the
mouse LE at the site of blastocyst apposition several hours
before attachment, and this persists through the early
attachment phase. HB-EGF is produced as soluble and
transmembrane forms. Molecular and genetic evidence
show that it influences embryonic functions as a para-
crine and/or juxtacrine factor by interacting with ErbB1
and/or ErbB4, which are expressed on the blastocyst cell
surface
30,31,38
. Most Hegf1
–/–
mice die during prenatal and
early postnatal life due to cardiac defects
71
, precluding an
examination of the implantation phenotype.
Implantation-competent blastocysts that also express
Hegf1 induce expression of the gene in the uterus in a
paracrine manner
29
. This auto-induction loop is per-
haps the first example of molecular crosstalk between
the blastocyst and uterus, initiating the attachment reac-
tion. HB-EGF also has a role in human implantation. Its
expression is maximal in the receptive endometrium,
and cells that express transmembrane HB-EGF adhere
to blastocysts that display cell-surface ErbB4
(REF. 72).
Attachment. It is correctly assumed that adhesive-
signalling systems are required for the attachment phase.
Indeed, numerous glycoproteins and carbohydrate
ligands and their receptors are expressed in LE and
Tr cells around the time of implantation. The most
important adhesion molecules that are implicated in
this process are
integrins, selectins, galectins, heparan
sulfate proteoglycans (HSPGs), mucin-1, cadherins
and the
trophinin–tastin–bystin complex
1,2,4
. Integrins
and selectins are of special interest because of their
unique functional features. In the human uterus,
integrin αvβ3 is localized to the LE during the recep-
tive phase, and its aberrant expression is correlated
with infertility and recurrent pregnancy loss
1
. Recent
evidence shows that selectin signalling is also important
in human implantation. While selectin oligosaccharide
ligands are expressed in the receptive LE,
L-selectin
molecules are displayed on the Tr cell surface
73
. More
importantly, beads that are coated with specific selectin
ligands adhere to trophoblast cells and, conversely, iso-
lated trophoblast cells bind preferentially to the recep-
tive uterine surface. These findings indicate that the
selectin-adhesion system constitutes an initial step
in human implantation. However, apparently normal
fertility in mice that lack L-selectin indicates a species-
specific variation in the adhesion cascade during implan-
tation. As stated before, Lif also seems to be important
for the attachment process, because Lif
–/–
mice show a
lack of HB-EGF and aberrant cyclooxygenase-2 (
Cox2)
expression in blastocysts during the anticipated time
of attachment
47,74
.
Penetration. One key event in implantation is an
increased endometrial vascular permeability at the
site of blastocyst attachment and penetration
(FIG. 2),
a process that involves the action of
prostaglandins
(PGs). COX1 and COX2 mediate PG synthesis and
are encoded by Ptgs1 and Ptgs2, respectively. Ptgs2
expression is unique in the mouse uterus, and shows
expression in the LE and underlying stromal cells at
the site of blastocyst attachment
75
. It is speculated that
HB-EGF that is produced in the uterus and embryo
induces uterine Ptgs2 expression. Ptgs2
–/–
females are
largely infertile, with defective ovulation, fertiliza-
tion, implantation and decidualization
75
. COX2-
derived prostacyclin (PGI
2
) is the primary PG that is
produced at the implantation site, and implantation
defects are improved in Ptgs2
–/–
mice by PG admin-
istration
76
. Evidence indicates that PGI
2
participates
in implantation via the activation of peroxisome-
proliferator-activated receptor-δ (
PPARδ), the expression
of which overlaps with Ptgs2 at the implantation site
76
.
However, depending on the genetic background,
COX1 can compensate for COX2 to improve infertil-
ity in Ptgs2
–/–
females
77
. Cox2 is also expressed in the
uterus and/or blastocyst during implantation in several
species, including primates
78
, indicating a conserved
function for COX2 in implantation.
The function of PG is further illustrated by the
reduced fertility of mice that lack cytoplasmic phos-
pholipase A
2α
(cPLA2α), which generates a precursor
for PG synthesis. Compromised fertility is due to the
deferral of on-time implantation, which leads to inap-
propriate embryo spacing, retarded feto-placental
development and reduced litter size
79
. These results
reveal that the cPLA2α–COX2 signalling axis is crucial
to implantation. Signalling by lysophosphatidic acid
(LPA), which belongs to a lysophospholipid group, also
influences blastocyst attachment in mice by activating
the G-protein-coupled receptor LPA3
(REF. 80). Like the
cPla2
α
/–
mice, lpA3
–/–
females show deferred implanta-
tion and its associated defects. The treatment of both
cPla2
α
–/–
and lpA3
–/–
mice with PGs resumes on-time
implantation, but embryo crowding persists. Phenotypic
similarities between lpA3-
and cPla2
α
-deficient mice
and reduced levels of uterine COX2 in lpA3
–/–
mice
identify COX2 as a common signalling pathway.
Implications for human fertility. From the results
discussed above, one important finding is that a short
delay in blastocyst attachment creates an adverse
ripple effect throughout the course of pregnancy,
which leads to defective feto–placental development
and poor pregnancy outcome. This indicates a new
concept in which embryo–uterine interactions dur-
ing implantation set up subsequent developmental
programming. This idea is supported by the clinical
finding that implantation beyond the normal window
of receptivity is associated with a higher risk of early
pregnancy loss in women
81
. The downstream pathways
of PG signalling that participate in the ripple effect
remain unknown. In light of these findings, one can
assume that many previous studies that describe early
REVIEWS
194
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
Placenta previa
A condition in humans in which
the placenta is situated close
to or covering the cervix.
Hemochorial placentation
The process by which maternal
blood comes in direct contact
with the trophoblast.
or mid-gestational embryonic lethality arising from
specific mutations might have originated at the time
of implantation.
Another unresolved issue is embryo spacing in the
uterus. While the LPA3–cPLA2α–COX2
signalling axis
is important for normal embryo spacing, attachment and
penetration, no molecule has been found to rescue the
spacing defect in mice that are mutant for the processes
involved. Answers to the question of how embryo spac-
ing is regulated might provide insights into the aetiology
of
placenta previa in humans. Is it possible that embryo
spacing is regulated by local factors that are associated
with PG signalling? BMPs are required for the spacing
of tissue structures during development
82
, and local
delivery of BMP2 or BMP4 in the uterus causes aberrant
embryo spacing
66
. There is also genetic evidence that
BMP5 and NODAL are important for this process
83
. As
there is a relationship between BMP and PG signalling
in other systems
84
, these pathways might work together
to influence embryo spacing.
Postimplantation uterine development
Uterine stromal cells that surround the blastocyst
undergo decidualization following attachment, even-
tually embedding the embryo in the antimesometrial
stromal bed. One function of the deciduum is to provide
nutritional support to the developing embryo before
the establishment of a functional placenta. Numerous
signalling molecules, including cytokines, homeobox
transcription factors, cell-cycle molecules, extracellular-
matrix remodelling factors and lipid mediators, are
expressed in the endometrium during decidualization
and are crucial to this process
2
. Here, we focus on the
less-explored areas, such as uterine angiogenesis and
establishment of the uterine–embryonic axis during the
postimplantation period
(FIG. 2b).
Uterine angiogenesis. Under physiological conditions
in adult females, angiogenesis primarily occurs in the
uterus and ovary during the reproductive cycle and preg-
nancy. Angiogenesis is essential to normal implantation
and placentation, and is profoundly influenced by vas-
cular endothelial growth factor (
VEGF) and angiopoi-
etins. PGs, because of their role in angiogenesis in other
systems, are also thought to participate in uterine angio-
genesis during pregnancy. But what is the link between
VEGF, angiopoietin and PG signalling?
The VEGF
receptor FLK1 (also known as KDR, kinase-insert-
domain protein receptor) is a marker of endothelial
cells during angiogenesis. Using Ptgs2
–/–
x Flk1
+/–LacZ
reporter mice, it was shown that COX2-derived PGs
markedly influence uterine angiogenesis during
decidualization by differentially regulating VEGF and
angiopoietin signalling cascades
85
. Uterine angiogen-
esis in Ptgs2
–/–
mice is severely compromised, owing to
defective VEGF, but not angiopoietin, signalling and
this defect is rescued by exogenous PG. Because PGs
coordinate VEGF signalling with that of angiopoietins
during decidual angiogenesis, one cause of compromised
implantation and decidualization in Ptgs2
–/–
mice could
be dysregulated vascular events.
Establishment of the uterine–embryonic axis. The
adult uterus undergoes dynamic cellular and molecu-
lar changes during pregnancy, but how these changes
are coordinated to specify the allocation of new cell
types, for example, decidual cells and their boundaries,
remains largely unknown. Decidualization is initiated
at the antimesometrial pole, subsequently extend-
ing to the mesometrial pole, the presumptive site of
placentation. This orients the implantation chamber
in an antimesometrial–mesometrial (AM–M) direc-
tion, in alignment with the embryonic axis. It is still
unclear how the implantation chamber is oriented
and grows in an AM–M direction, with the decidual
reaction spreading in the same direction. It is also not
known how decidual cell growth is restricted, leaving a
layer of undifferentiated stromal cells underneath the
myometrium.
It is speculated that WNT signalling, in collabora-
tion with those of BMP and fibroblast growth factor
(FGF), helps to orient the implantation chamber in
the AM–M direction and specifies these boundaries
during decidualization
(FIG. 2b); in particular, differen-
tial WNT4 signalling seems to participate in making
this boundary
54
. An inverse relationship with respect
to Bmp2 and Noggin expression that is observed dur-
ing implantation and decidualization also indicates
differential BMP signalling during early pregnancy
66
.
However, the expression of
Dan (differential screen-
ing-selected gene aberrative in neuroblastoma), a
member of the Dan/Dante Bmp-antagonist gene family,
and
Crim1 (cysteine-rich transmembrane BMP-
regulator-1), which encodes a protein that is thought to
bind BMP, partially overlap with that of Bmp2 expres-
sion. Furthermore, antimesometrial expression of
Fgf2,
in contrast to mesometrial expression of
Fgf10, adds to
evidence that the AM–M orientation of the uterus dur-
ing early pregnancy is influenced by differential gene
expression
66
. We speculate that uterine orientation helps
to establish embryonic orientation during development,
and that the failure of the implantation chamber to
orient itself in an AM–M direction is likely to disrupt
embryonic orientation. Therefore, these developmental
genes are not only important for establishing bound-
aries and polarities during embryogenesis, but also for
establishing the orientation of the growing implantation
chamber and creating boundaries to prevent undifferen-
tiated stromal cells from decidualizing (the undifferen-
tiated stromal cells might serve to replenish the stroma
after parturition).
How mice can help humans
Studies in mice have provided insights into the molecu-
lar basis of human implantation because of their shared
features. Both mouse and human embryos can develop
in vitro in simple, defined media. In both species,
embryo implantation leads to stromal decidualization
— embryos embed in the antimesometrial stroma and
placentation is
hemochorial.
Mouse embryos grow more effectively when they
are cultured in a small volume
86
. This protocol, which is
practiced by some clinics, has shown improved embryo
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
195
© 2006 Nature Publishing Group
MALDI mass spectrometry
(Matrix-assisted laser
desorption/ionization mass
spectrometry). It is based on
the co-crystallization of a test
compound with an ultraviolet-
light-absorbing matrix, which
allows ionization using laser
excitation to determine the
mass of the test compound.
Preeclampsia
The development of
hypertension with proteinuria
(excess protein in urine) and/or
oedema during pregnancy;
early onset occurs from
defective trophoblast function.
development in human IVF programs. However, the
pregnancy success rate remains poor (~30%) due to
the transfer of IVF-derived embryos into nonrecep-
tive uteri. One cause of nonreceptivity might be high
oestrogen levels; this results from the gonadotropin
treatment that is given to women to stimulate the
ovaries to produce multiple eggs. Indeed, the range of
oestrogen levels that determines uterine receptivity in
mice is narrow
26
. This remarkable uterine sensitivity
to oestrogen might be important to ensure the correct
timing of implantation, which is crucial to pregnancy
outcome. Understanding the cause of uterine non-
receptivity at higher oestrogen levels might make it
possible to extend uterine receptivity by using an aro-
matase inhibitor to neutralize excess oestrogen during
gonadotropin stimulation.
Early onset of intrauterine growth restriction, recur-
rent abortion,
preeclampsia and preterm delivery are
important reproductive health issues, and are associated
with placental deficiencies. A transient postponement of
blastocyst attachment in mice produces a detrimental
ripple effect throughout pregnancy, which indicates that
these end results are due to defective implantation. Such
defects could be corrected, because signalling by LIF,
HB-EGF, COX2 and HOX family members, which are
important at different stages of implantation in mice, are
also thought to be important for human implantation. In
fact, downregulation of HB-EGF expression in the human
trophoblast is associated with preeclampsia
87
. Further
insights into these and the recently identified pathways
that are described above might improve pregnancy
success and could also help in designing new and improved
contraceptives. There is a need to develop nonsteroidal
contraceptives so that women are spared the complica-
tions of hormonal imbalances and the risk of developing
gynecological cancers. Molecular approaches to disrupt
LIF–STAT, FKBP52 or LPA3–COX2 signalling pathways
might be considered for potential contraceptives.
Conclusion
Implantation is an incredibly useful biological system,
a better understanding of which will advance our
know ledge in several basic physiological processes.
These include: the paracrine and juxtacrine epithelial–
epithelial interactions that occur between the Tr and LE
during attachment; the epithelial–mesenchymal interac-
tion between the LE and the stroma; Tr–epithelium–stroma
interactions, involving cell migration and invasion; vas-
cular permeability and adult angiogenesis; and regulated
growth (proliferation, differentiation, polyploidy and
apoptosis) during stromal decidualization. Despite the
large strides that have been made by applying genom-
ics and proteomic approaches to rodents, the field faces
many important challenges
(BOX 4).
Implantation involves numerous signalling pathways
that are common to other systems under either normal or
pathological conditions. Therefore, research on implanta-
tion should appeal to a broader range of scientists, not
solely to reproductive or developmental biologists.
For
example, many of the features and signalling pathways
that are required during implantation are also active
during tumourigenesis — the difference being that tight
Box 4 | Future challenges for reproductive biology and reproductive medicine
There is a need to identify reliable markers of uterine receptivity and to develop the means to extend uterine receptivity
or treat nonreceptivity to improve the pregnancy rate in in vitro fertilization and embryo-transfer programmes.
Overcoming these challenges will lessen the need to transfer multiple embryos to increase the pregnancy rate and the
resulting complications of multiple pregnancies.
Although various signalling pathways operate during implantation, it is still unclear whether they work independently, in
parallel or converge on a common pathway.
Suitable animal models must continue to be developed to define the molecular communication between the uterus and
embryo. Such studies require information about the contribution that is made by each of the two tissues, a task not easily
achievable in humans because of experimental difficulties and ethical restrictions on research with human embryos.
Another challenge is to identify gene promoters for creating inducible Cre-transgenic mice for conditional gene deletion
specifically in uterine cells; genome-wide deletion of many implantation-associated genes leads to embryonic lethality,
precluding studies on implantation. This approach will also help to elucidate the long-term versus acute effects of a gene
during implantation. This is particularly important in the context of the adaptation of animals to a new make-up. For
example, deletion of one gene that does not affect pregnancy under normal conditions shows adverse effects under a
stress situation
109
.
Efforts should continue in establishing a relevant in vitro model of implantation to study the hierarchy of events that are
triggered by the embryo, and the function of specific signalling molecules.
There is a need to properly annotate the markers of uterine receptivity that are derived from microarray experiments in
rodents and humans
58–62,110,111
with results from functional analyses. Comparative proteomics between wild-type and
mutant uteri (an assay that is under-exploited in the field) should provide unique information
68
.
Another approach that shows great promise is the direct analysis of tissue sections by MALDI mass spectrometry to
identify the spatial localization of proteins
112
. This approach is attractive for comparing proteome profiles of implantation
versus interimplantation sites or between different regions within an implantation site.
The nature of embryonic signals that influence uterine functions is mostly unknown. In rodents and humans, the limiting
factor is the availability of adequate amounts of tissues for analysis. With the advent of microscale proteomics and
genomics, it might now be possible to identify embryonic signals during implantation. Once potential molecules are
identified, their functions could be assessed by local application into the uterus using blastocyst-sized beads as carriers
66
.
REVIEWS
196
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
1. Carson, D. D. et al. Embryo implantation. Dev. Biol.
223, 217–237 (2000).
2. Dey, S. K. et al. Molecular cues to implantation.
Endocr. Rev. 25, 341–373 (2004).
3. Paria, B. C., Reese, J., Das, S. K. & Dey, S. K.
Deciphering the cross-talk of implantation: advances
and challenges. Science 296, 2185–2188 (2002).
4. Red-Horse, K. et al. Trophoblast differentiation during
embryo implantation and formation of the maternal-
fetal interface. J. Clin. Invest. 114, 744–754 (2004).
5. Enders, A. C. & Schlafke, S. A morphological analysis
of early implantation stages in the rat. Am. J. Anat.
120, 195–226 (1967).
6. Nothias, J. Y., Majumder, S., Kaneko, K. J. &
DePamphilis, M. L. Regulation of gene expression at
the beginning of mammalian development. J. Biol.
Chem. 270, 22077–22080 (1995).
7. Latham, K. E., Garrels, J. I., Chang, C. & Solter, D.
Quantitative analysis of protein synthesis in mouse
embryos. I. Extensive reprogramming at the one- and
two-cell stages. Development 11 2 , 921–932 (1991).
8. Shi, C. Z. et al. Protein databases for compacted
eight-cell and blastocyst-stage mouse embryos.
Mol. Reprod. Dev. 37, 34–47 (1994).
9. Zimmermann, J. W. & Schultz, R. M. Analysis of gene
expression in the preimplantation mouse embryo: use
of mRNA differential display. Proc. Natl Acad. Sci.
USA 91, 5456–5460 (1994).
10. Ko, M. S. et al. Large-scale cDNA analysis reveals
phased gene expression patterns during
preimplantation mouse development. Development
127, 1737–1749 (2000).
11. Hamatani, T., Carter, M. G., Sharov, A. A. & Ko, M. S.
Dynamics of global gene expression changes during
mouse preimplantation development. Dev. Cell 6,
117–131 (2004).
This study, together with the work described in
reference 12, shows that mouse preimplantation
embryo development is a dynamic molecular
process that is governed by waves of gene
expression.
12. Wang, Q. T. et al. A genome-wide study of gene
activity reveals developmental signaling pathways in
the preimplantation mouse embryo. Dev. Cell 6,
133–144 (2004).
13. Zeng, F., Baldwin, D. A. & Schultz, R. M. Transcript
profiling during preimplantation mouse development.
Dev. Biol. 272, 483–496 (2004).
14.
Tong, Z. B. et al. Mater, a maternal effect gene
required for early embryonic development in mice.
Nature Genet. 26, 267–268 (2000).
15. Johnson, M. H. & McConnell, J. M. Lineage allocation
and cell polarity during mouse embryogenesis.
Semin. Cell Dev. Biol. 15, 583–597 (2004).
16. Rossant, J. Lineage development and polar
asymmetries in the peri-implantation mouse
blastocyst. Semin. Cell Dev. Biol. 15, 573–581
(2004).
17. Nichols, J. et al. Formation of pluripotent stem cells
in the mammalian embryo depends on the POU
transcription factor Oct4. Cell 95, 379–391 (1998).
This study shows that expression of Oct4 in inside
cells of mouse preimplantation embryos is required
for the generation of pluripotent cells. The other
key molecules for cell-lineage differentiation during
preimplantation development are described in
references 18–24.
18. Avilion, A. A. et al. Multipotent cell lineages in early
mouse development depend on SOX2 function. Genes
Dev. 17, 126–140 (2003).
19. Niwa, H., Miyazaki, J. & Smith, A. G. Quantitative
expression of Oct-3/4 defines differentiation,
dedifferentiation or self-renewal of ES cells. Nature
Genet. 24, 372–376 (2000).
20. Mitsui, K. et al. The homeoprotein Nanog is required
for maintenance of pluripotency in mouse epiblast and
ES cells. Cell 11 3, 631–642 (2003).
21. Chambers, I. et al. Functional expression cloning of
Nanog, a pluripotency sustaining factor in embryonic
stem cells. Cell 11 3, 643–655 (2003).
22. Boyer, L. A. et al. Core transcriptional regulatory
circuitry in human embryonic stem cells. Cell 122,
947–956 (2005).
23. Strumpf, D. et al. Cdx2 is required for correct cell fate
specification and differentiation of trophectoderm in
the mouse blastocyst. Development 132, 2093–2102
(2005).
24. Russ, A. P. et al. Eomesodermin is required for mouse
trophoblast development and mesoderm formation.
Nature 404, 95–99 (2000).
25. Paria, B. C., Huet-Hudson, Y. M. & Dey, S. K.
Blastocyst’s state of activity determines the
‘‘window’’ of implantation in the receptive mouse
uterus. Proc. Natl Acad. Sci. USA 90, 10159–10162
(1993).
This study, which uses a delayed-implantation
mouse model, showed for the first time that
the receptive state of the uterus alone is not
sufficient for successful implantation, but that
blastocysts must also achieve implantation
competency. The differential roles of oestrogen
and catecholoestrogens in establishing the
window of implantation are highlighted in
references 26 and 32.
26. Ma, W. G., Song, H., Das, S. K., Paria, B. C. & Dey, S. K.
Estrogen is a critical determinant that specifies the
duration of the window of uterine receptivity for
implantation. Proc. Natl Acad. Sci. USA 100,
2963–2968 (2003).
27. Lopes, F. L., Desmarais, J. A. & Murphy, B. D.
Embryonic diapause and its regulation. Reproduction
128, 669–678 (2004).
28. Renfree, M. B. & Shaw, G. Diapause. Annu. Rev.
Physiol. 62, 353–375 (2000).
29. Hamatani, T. et al. Global gene expression analysis
identifies molecular pathways distinguishing blastocyst
dormancy and activation. Proc. Natl Acad. Sci. USA
101, 10326–10331 (2004).
This study analyses global gene expression in
dormant and activated mouse blastocysts,
providing evidence that gene-expression patterns
are distinct at these two different physiological
states of the embryo.
30. Paria, B. C., Das, S. K., Andrews, G. K. & Dey, S. K.
Expression of the epidermal growth factor receptor
gene is regulated in mouse blastocysts during delayed
implantation. Proc. Natl Acad. Sci. USA 90, 55–59
(1993).
31. Raab, G. et al. Mouse preimplantation blastocysts
adhere to cells expressing the transmembrane form of
heparin-binding EGF-like growth factor. Development
122, 637–645 (1996).
32. Paria, B. C. et al. Coordination of differential effects of
primary estrogen and catecholestrogen on two distinct
targets mediates embryo implantation in the mouse.
Endocrinology 139, 5235–5246 (1998).
33. Guo, Y. et al. N–acylphosphatidylethanolamine-
hydrolyzing phospholipase D is an important
determinant of uterine anandamide levels during
implantation. J. Biol. Chem. 280, 23429–23432
(2005).
34. Paria, B. C., Das, S. K. & Dey, S. K.
The preimplantation mouse embryo is a target for
cannabinoid ligand-receptor signaling. Proc. Natl
Acad. Sci. USA 92, 9460–9464 (1995).
This work provided the first evidence for the
presence of the G-protein-coupled cannabinoid
receptors CB1 and CB2 in preimplantation mouse
embryos. The differential roles of
endocannabinoids in embryo–uterine interactions
during implantation are further illustrated in
references 33 and 35–37.
35. Wang, H. et al. Aberrant cannabinoid signaling
impairs oviductal transport of embryos. Nature Med.
10, 1074–1080 (2004).
36. Wang, H. et al. Differential G protein-coupled
cannabinoid receptor signaling by anandamide directs
blastocyst activation for implantation. Proc. Natl
Acad. Sci. USA 100, 14914–14919 (2003).
37. Paria, B. C. et al. Dysregulated cannabinoid signaling
disrupts uterine receptivity for embryo implantation.
J. Biol. Chem. 276, 20523–20528 (2001).
38. Wang, J., Mayernik, L., Schultz, J. F. & Armant, D. R.
Acceleration of trophoblast differentiation by
heparin-binding EGF-like growth factor is dependent
on the stage-specific activation of calcium influx by
ErbB receptors in developing mouse blastocysts.
Development 127, 33–44 (2000).
39. Stachecki, J. J. & Armant, D. R. Transient release of
calcium from inositol 1,4,5-trisphosphate-specific
stores regulates mouse preimplantation development.
Development 122, 2485–2496 (1996).
40. Wang, Y. et al. Entire mitogen activated protein kinase
(MAPK) pathway is present in preimplantation mouse
embryos. Dev. Dyn. 231, 72–87 (2004).
41. Riley, J. K. et al. The PI3K/Akt pathway is present and
functional in the preimplantation mouse embryo.
Dev. Biol. 284, 377–386 (2005).
42. Lubahn, D. B. et al. Alteration of reproductive
function but not prenatal sexual development after
insertional disruption of the mouse estrogen receptor
gene. Proc. Natl Acad. Sci. USA 90, 11162–11166
(1993).
43. Curtis, S. W., Clark, J., Myers, P. & Korach, K. S.
Disruption of estrogen signaling does not prevent
progesterone action in the estrogen receptor α
knockout mouse uterus. Proc. Natl Acad. Sci. USA 96,
3646–3651 (1999).
44. Paria, B. C., Tan, J., Lubahn, D. B., Dey, S. K. & Das, S. K.
Uterine decidual response occurs in estrogen
receptor-α-deficient mice. Endocrinology 140,
2704–2710 (1999).
regulation occurs during implantation, while dysregu-
lation of the same pathways occurs in tumourigenesis.
Therefore, understanding the intricacies of implanta-
tion might help to better understand the complexities of
tumourigenesis, and might one day reveal that ‘life and
death are linked by a common thread’.
Note added in proof
The discoveries that CDX2 and OCT3/4 are crucial
for specifying Tr and ICM cells, respectively, in pre-
implantation embryos have now been expanded to
show their roles in stem-cell biology. A report shows
that embryos that are missing CDX2 develop to blas-
tocysts, but fail to implant because they lack Tr. These
blastocysts have ICMs and generate ES cells when
grown in culture
128
. Another recent report shows that
ES cells can be forced to differentiate to trophoblast
stem (TS) cells by repressing OCT3/4
(REF. 129). This
study shows that CDX2 is dispensable for Tr differen-
tiation, but is required for TS-cell self renewal, which
indicates that the reciprocal inhibition of transcription
factors OCT3/4 and CDX2 participates in lineage dif-
ferentiation in mammalian embryos
129
. Although the
cell-lineage specification in mammalian embryos is
primarily thought to occur between the 8-cell and
blastocyst stages, the question of how embryonic
polarity is established is still a subject of debate. Gore
et al.
130
now show that maternal Squint, a mammalian
NODAL-related morphogen, localizes to two blast-
omeres at the 4-cell stage and specifies the dorsal axis,
which indicates that NODAL could also be involved in
determining polarity in early mammalian embryos.
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
197
© 2006 Nature Publishing Group
45. Lydon, J. P. et al. Mice lacking progesterone receptor
exhibit pleiotropic reproductive abnormalities. Genes
Dev. 9, 2266–2278 (1995).
46. Mulac-Jericevic, B., Mullinax, R. A., DeMayo, F. J.,
Lydon, J. P. & Conneely, O. M. Subgroup of
reproductive functions of progesterone mediated by
progesterone receptor-B-isoform. Science 289,
1751–1754 (2000).
47. Song, H., Lim, H., Das, S. K., Paria, B. C. & Dey, S. K.
Dysregulation of EGF family of growth factors and
COX-2 in the uterus during the preattachment and
attachment reactions of the blastocyst with the
luminal epithelium correlates with implantation failure
in LIF-deficient mice. Mol. Endocrinol. 14, 1147–1161
(2000).
48. Stewart, C. L. et al. Blastocyst implantation depends
on maternal expression of leukaemia inhibitory factor.
Nature 359, 76–79 (1992).
This study provided the first evidence that Lif is
expressed in mouse uterine glands and is essential
for implantation. The stromal expression of Lif
surrounding the blastocyst at the time of
attachment was also found to be important for
implantation, as described in reference 47.
49. Ernst, M. et al. Defective gp130-mediated signal
transducer and activator of transcription (STAT)
signaling results in degenerative joint disease,
gastrointestinal ulceration, and failure of uterine
implantation. J. Exp. Med. 194, 189–203 (2001).
50. Benson, G. V. et al. Mechanisms of reduced fertility in
Hoxa-10 mutant mice: uterine homeosis and loss of
maternal Hoxa-10 expression. Development 122,
2687–2696 (1996).
51. Hsieh-Li, H. M. et al. Hoxa 11 structure, extensive
antisense transcription, and function in male and
female fertility. Development 121, 1373–1385 (1995).
52. Lim, H., Ma, L., Ma, W. G., Maas, R. L. & Dey, S. K.
Hoxa-10 regulates uterine stromal cell responsiveness
to progesterone during implantation and
decidualization in the mouse. Mol. Endocrinol. 13,
1005–1017 (1999).
53. Satokata, I., Benson, G. & Maas, R. Sexually
dimorphic sterility phenotypes in Hoxa10-deficient
mice. Nature 374, 460–463 (1995).
This paper was the first to show female infertility in
mice that lack Hoxa10. It was later shown that
defective decidualization is the cause of this female
infertility, as described in references 50 and 52.
54. Daikoku, T. et al. Uterine Msx-1 and Wnt4 signaling
becomes aberrant in mice with the loss of leukemia
inhibitory factor or Hoxa-10: evidence for a novel
cytokine-homeobox-Wnt signaling in implantation.
Mol. Endocrinol. 18, 1238–1250 (2004).
This work was the first to provide evidence that
cytokines, homeotic proteins and morphogens in
the mouse uterus constitute a molecular circuitry
that is crucial to implantation.
55. Gendron, R. L. et al. Abnormal uterine stromal and
glandular function associated with maternal
reproductive defects in Hoxa-11 null mice. Biol.
Reprod. 56, 1097–1105 (1997).
56. Taylor, H. S., Arici, A., Olive, D. & Igarashi, P.
HOXA10 is expressed in response to sex steroids at
the time of implantation in the human endometrium.
J. Clin. Invest. 101, 1379–1384 (1998).
57. Wang, W., Van De Water, T. & Lufkin, T. Inner ear and
maternal reproductive defects in mice lacking the
Hmx3 homeobox gene. Development 125, 621–634
(1998).
58. Borthwick, J. M. et al. Determination of the transcript
profile of human endometrium. Mol. Hum. Reprod. 9,
19–33 (2003).
59. Carson, D. D. et al. Changes in gene expression during
the early to mid-luteal (receptive phase) transition in
human endometrium detected by high-density
microarray screening. Mol. Hum. Reprod. 8, 871–879
(2002).
60. Kao, L. C. et al. Global gene profiling in human
endometrium during the window of implantation.
Endocrinology 143, 2119–2138 (2002).
61. Mirkin, S. et al. In search of candidate genes critically
expressed in the human endometrium during the
window of implantation. Hum. Reprod. 20,
2104–2117 (2005).
62. Riesewijk, A. et al. Gene expression profiling of human
endometrial receptivity on days LH+2 versus LH+7
by microarray technology. Mol. Hum. Reprod. 9,
253–264 (2003).
63. Satokata, I. & Maas, R. Msx1 deficient mice exhibit
cleft palate and abnormalities of craniofacial and tooth
development. Nature Genet. 6, 348–356 (1994).
64. Takamoto, N., Zhao, B., Tsai, S. Y. & DeMayo, F. J.
Identification of Indian hedgehog as a progesterone-
responsive gene in the murine uterus. Mol. Endocrinol.
16, 2338–2348 (2002).
65. Matsumoto, H., Zhao, X., Das, S. K., Hogan, B. L. &
Dey, S. K. Indian hedgehog as a progesterone-
responsive factor mediating epithelial-mesenchymal
interactions in the mouse uterus. Dev. Biol. 245,
280–290 (2002).
66. Paria, B. C.
et al. Cellular and molecular responses of
the uterus to embryo implantation can be elicited by
locally applied growth factors. Proc. Natl Acad. Sci.
USA 98, 1047–1052 (2001).
This article provides a comprehensive account of
the expression of morphogens, including HH, BMP,
WNT and FGF signalling in the mouse uterus during
the periimplantation period. The evidence that HH
signalling in the uterine epithelial–mesenchymal
interaction is important for implantation was later
reported in references 64 and 65.
67. Parr, B. A. & McMahon, A. P. Sexually dimorphic
development of the mammalian reproductive tract
requires Wnt-7a. Nature 395, 707–710 (1998).
68. Daikoku, T. et al. Proteomic analysis identifies
immunophilin FK506 binding protein 4 (FKBP52) as a
downstream target of Hoxa10 in the periimplantation
mouse uterus. Mol. Endocrinol. 19, 683–697 (2005).
69. Tranguch, S. et al. Cochaperone immunophilin
FKBP52 is critical to uterine receptivity for embryo
implantation. Proc. Natl Acad. Sci. USA 102,
14326–14331 (2005).
70. Das, S. K. et al. Heparin-binding EGF-like growth
factor gene is induced in the mouse uterus temporally
by the blastocyst solely at the site of its apposition: a
possible ligand for interaction with blastocyst EGF-
receptor in implantation. Development 120,
10711083 (1994).
The role of HB-EGF as an early initiator of
molecular crosstalk between the blastocyst and
uterus before attachment was first illustrated in
this study.
71. Iwamoto, R. et al. Heparin-binding EGF-like growth
factor and ErbB signaling is essential for heart function.
Proc. Natl Acad. Sci. USA 100, 3221–3226 (2003).
72. Chobotova, K. et al. Heparin-binding epidermal
growth factor and its receptor ErbB4 mediate
implantation of the human blastocyst. Mech. Dev.
119 , 137–144 (2002).
73. Genbacev, O. D. et al. Trophoblast L-selectin-mediated
adhesion at the maternal-fetal interface. Science 299,
405–408 (2003).
This study shows that, in humans, selectin
oligosaccharide ligands are expressed in the
receptive uterine lining, while the Tr cell surface is
decorated with L-selectin. Further evidence
indicates that this ligand–receptor signalling is
important for human implantation.
74. Fouladi-Nashta, A. A. et al. Characterization of the
uterine phenotype during the peri-implantation period
for LIF-null, MF1 strain mice. Dev. Biol. 281, 1–21
(2005).
75. Lim, H. et al. Multiple female reproductive failures in
cyclooxygenase 2-deficient mice. Cell 91, 197–208
(1997).
This study shows that ovulation, fertilization,
implantation and decidualization are defective in
mice lacking COX2-derived prostaglandins.
76. Lim, H. et al. Cyclo-oxygenase-2-derived prostacyclin
mediates embryo implantation in the mouse via
PPARδ. Genes Dev. 13
, 1561–1574 (1999).
77. Wang, H. et al. Rescue of female infertility from the
loss of cyclooxygenase-2 by compensatory
up-regulation of cyclooxygenase-1 is a function of
genetic makeup. J. Biol. Chem. 279, 10649–10658
(2004).
78. Kim, J. J. et al. Expression of cyclooxygenase-1 and-2
in the baboon endometrium during the menstrual
cycle and pregnancy. Endocrinology 140, 2672–2678
(1999).
79. Song, H. et al. Cytosolic phospholipase A2α is crucial
for ‘‘on-time’’ embryo implantation that directs
subsequent development. Development 129,
2879–2889 (2002).
This work shows that mouse uteri that lack cPLA2α
transiently defer on-time implantation, creating an
adverse ripple effect throughout the course of
pregnancy and leading to poor pregnancy outcome.
A similar phenotype is observed in lpA3-null mice,
as reported in reference 80. The importance of on-
time implantation in human pregnancy outcome is
presented in reference 81.
80. Ye, X. et al. LPA3-mediated lysophosphatidic acid
signalling in embryo implantation and spacing. Nature
435, 104–108 (2005).
81. Wilcox, A. J., Baird, D. D. & Weinberg, C. R. Time of
implantation of the conceptus and loss of pregnancy.
N. Engl. J. Med. 340, 1796–1799 (1999).
82. Hogan, B. L. Bone morphogenetic proteins:
multifunctional regulators of vertebrate development.
Genes Dev. 10, 1580–1594 (1996).
83. Pfendler, K. C., Yoon, J., Taborn, G. U., Kuehn, M. R. &
Iannaccone, P. M. Nodal and bone morphogenetic
protein 5 interact in murine mesoderm formation and
implantation. Genesis 28, 1–14 (2000).
84. Arikawa, T., Omura, K. & Morita, I. Regulation of bone
morphogenetic protein-2 expression by endogenous
prostaglandin E2 in human mesenchymal stem cells. J.
Cell. Physiol. 200, 400–406 (2004).
85. Matsumoto, H. et al. Cyclooxygenase-2 differentially
directs uterine angiogenesis during implantation in
mice. J. Biol. Chem. 277, 29260–29267 (2002).
This study shows that COX2-derived prostaglandins
coordinate VEGF and angiopoietin signalling during
angiogenesis in the mouse deciduum — a process
that is required for the establishment of pregnancy.
86. Paria, B. C. & Dey, S. K. Preimplantation embryo
development in vitro: cooperative interactions among
embryos and role of growth factors. Proc. Natl Acad.
Sci. USA 87, 4756–4760 (1990).
87. Leach, R. E. et al. Pre-eclampsia and expression of
heparin-binding EGF-like growth factor.
Lancet 360,
1215–1219 (2002).
88. National Institutes of Health. Stem Cells: Scientific
Progress and Future Research Directions. Stem Cell
Information [online], http://stemcells.nih.gov/info/
scireport (2001).
89. Gardner, R. L. Specification of embryonic axes begins
before cleavage in normal mouse development.
Development 128, 839–847 (2001).
90. Fujimori, T., Kurotaki, Y., Miyazaki, J. & Nabeshima, Y.
Analysis of cell lineage in two- and four-cell mouse
embryos. Development 130, 5113–5122
(2003).
91. Piotrowska, K., Wianny, F., Pedersen, R. A. &
Zernicka-Goetz, M. Blastomeres arising from the first
cleavage division have distinguishable fates in normal
mouse development. Development 128, 3739–3748
(2001).
92. Piotrowska, K. & Zernicka-Goetz, M. Role for sperm in
spatial patterning of the early mouse embryo.
Nature 409, 517–521 (2001).
93. Gardner, R. L. The early blastocyst is bilaterally
symmetrical and its axis of symmetry is aligned with
the animal–vegetal axis of the zygote in the mouse.
Development 124, 289–301 (1997).
This article proposes the concept of the embryonic
axis and cell polarity during mouse preimplantation
development. The ongoing debate on this subject is
further highlighted in references
89–92,96,101,107 and 108.
94. Piotrowska-Nitsche, K., Perea-Gomez, A., Haraguchi, S.
& Zernicka-Goetz, M. Four-cell stage mouse
blastomeres have different developmental properties.
Development 132, 479–490 (2005).
95. Piotrowska-Nitsche, K. & Zernicka-Goetz, M. Spatial
arrangement of individual 4-cell stage blastomeres
and the order in which they are generated correlate
with blastocyst pattern in the mouse embryo. Mech.
Dev. 122, 487–500 (2005).
96. Plusa, B. et al. The first cleavage of the mouse zygote
predicts the blastocyst axis. Nature 434, 391–395
(2005).
97. Surani, M. A. & Barton, S. C. Spatial distribution of
blastomeres is dependent on cell division order and
interactions in mouse morulae. Dev. Biol. 102,
335–343 (1984).
98. Garbutt, C. L., Johnson, M. H. & George, M. A. When
and how does cell division order influence cell
allocation to the inner cell mass of the mouse
blastocyst? Development 100, 325–332 (1987).
99. Alarcon, V. B. & Marikawa, Y. Unbiased contribution of
the first two blastomeres to mouse blastocyst
development. Mol. Reprod. Dev. 72
, 354–361
(2005).
100. Chroscicka, A., Komorowski, S. & Maleszewski, M.
Both blastomeres of the mouse 2-cell embryo
contribute to the embryonic portion of the blastocyst.
Mol. Reprod. Dev. 68, 308–312 (2004).
101. Motosugi, N., Bauer, T., Polanski, Z., Solter, D. &
Hiiragi, T. Polarity of the mouse embryo is established
at blastocyst and is not prepatterned. Genes Dev. 19,
1081–1092 (2005).
REVIEWS
198
|
MARCH 2006
|
VOLUME 7 www.nature.com/reviews/genetics
© 2006 Nature Publishing Group
102. Rossant, J. & Tam, P. P. Emerging asymmetry and
embryonic patterning in early mouse development.
Dev. Cell 7, 155–164 (2004).
103. Zernicka-Goetz, M. First cell fate decisions and spatial
patterning in the early mouse embryo. Semin. Cell
Dev. Biol. 15, 563–572 (2004).
104. Plusa, B., Grabarek, J. B., Piotrowska, K., Glover, D. M.
& Zernicka-Goetz, M. Site of the previous meiotic
division defines cleavage orientation in the mouse
embryo. Nature Cell Biol. 4, 811–815 (2002).
105. Plusa, B., Piotrowska, K. & Zernicka-Goetz, M.
Sperm entry position provides a surface marker for
the first cleavage plane of the mouse zygote. Genesis
32, 193–198 (2002).
106. Davies, T. J. & Gardner, R. L. The plane of first
cleavage is not related to the distribution of sperm
components in the mouse. Hum. Reprod. 17,
2368–2379 (2002).
107. Hiiragi, T. & Solter, D. First cleavage plane of the
mouse egg is not predetermined but defined by the
topology of the two apposing pronuclei. Nature 430,
360–364 (2004).
108. Louvet-Vallee, S., Vinot, S. & Maro, B. Mitotic spindles
and cleavage planes are oriented randomly in the two-
cell mouse embryo. Curr. Biol. 15, 464–469 (2005).
109. Ain, R., Dai, G., Dunmore, J. H., Godwin, A. R. &
Soares, M. J. A prolactin family paralog regulates
reproductive adaptations to a physiological stressor.
Proc. Natl Acad. Sci. USA 101, 16543–16548
(2004).
110. Cheon, Y. P. et al. A genomic approach to identify
novel progesterone receptor regulated pathways in the
uterus during implantation. Mol. Endocrinol. 16,
2853–2871 (2002).
111. Reese, J. et al. Global gene expression analysis to
identify molecular markers of uterine receptivity and
embryo implantation. J. Biol. Chem. 276,
44137–44145 (2001).
112. Reyzer, M. L. & Caprioli, R. M. MALDI mass
spectrometry for direct tissue analysis: a new tool for
biomarker discovery. J. Proteome Res. 4, 1138–1142
(2005).
113 . Ralston, A. & Rossant, J. Genetic regulation of stem
cell origins in the mouse embryo. Clin. Genet. 68,
106–112 (2005).
114. Branford, W. W., Benson, G. V., Ma, L., Maas, R. L. &
Potter, S. S. Characterization of Hoxa-10/Hoxa-11
transheterozygotes reveals functional redundancy and
regulatory interactions. Dev. Biol. 224, 373–387
(2000).
115. Mericskay, M., Kitajewski, J. & Sassoon, D. Wnt5a
is required for proper epithelial–mesenchymal
interactions in the uterus. Development 131,
2061–2072 (2004).
116. Shindo, T. et al. ADAMTS-1: a metalloproteinase-
disintegrin essential for normal growth, fertility, and
organ morphology and function. J. Clin. Invest. 105,
1345–1352 (2000).
117. Simmen, R. C. et al. Subfertility, uterine hypoplasia,
and partial progesterone resistance in mice lacking the
Kruppel-like factor 9/basic transcription element-
binding protein-1 (Bteb1) gene. J. Biol. Chem. 279,
29286–29294 (2004).
118. Fowler, K. J. et al. Uterine dysfunction and genetic
modifiers in centromere protein B-deficient mice.
Genome Res 10, 30–41 (2000).
119. Panda, D. K. et al. Targeted ablation of the
25-hydroxyvitamin D 1α-hydroxylase enzyme:
evidence for skeletal, reproductive, and immune
dysfunction. Proc. Natl. Acad Sci. USA 98,
7498–7503 (2001).
120. Baker, J. et al. Effects of an Igf1 gene null mutation on
mouse reproduction. Mol. Endocrinol. 10, 903–918
(1996).
121. Smith, C. L. et al. Genetic ablation of the steroid
receptor coactivator-ubiquitin ligase, E6-AP, results in
tissue-selective steroid hormone resistance and
defects in reproduction. Mol. Cell Biol. 22, 525–535
(2002).
122. Yoshizawa, T. et al. Mice lacking the vitamin D receptor
exhibit impaired bone formation, uterine hypoplasia
and growth retardation after weaning. Nature Genet.
16, 391–396 (1997).
123. Igakura, T. et al. A null mutation in basigin, an
immunoglobulin superfamily member, indicates its
important roles in peri-implantation development
and spermatogenesis. Dev. Biol. 194, 152–165
(1998).
124. Kuno, N. et al. Female sterility in mice lacking the
basigin gene, which encodes a transmembrane
glycoprotein belonging to the immunoglobulin
superfamily. FEBS Lett. 425, 191–194 (1998).
125. Barak, Y. et al. Effects of peroxisome proliferator-
activated receptor δ on placentation, adiposity, and
colorectal cancer. Proc. Natl Acad. Sci USA 99,
303–308 (2002).
126. Robb, L. et al. Infertility in female mice lacking the
receptor for interleukin 11 is due to a defective uterine
response to implantation. Nature Med 4, 303–308
(1998).
127. Bilinski, P., Roopenian, D. & Gossler, A.
Maternal IL-11Rα function is required for normal
decidua and fetoplacental development in mice.
Genes Dev. 12, 2234–2243 (1998).
128. Meissner, A. & Jaenisch, R. Generation of nuclear
transfer-derived pluripotent ES cells from cloned Cdx2-
deficient blastocysts. Nature 439, 212–215 (2006).
129. Niwa, H. et al. Interaction between Oct3/4 and Cdx2
determines trophectoderm differentiation. Cell 123,
917–929 (2005).
130. Gore, A. V. et al. The zebrafish dorsal axis is apparent at
the four-cell stage. Nature 438, 1030–1035 (2005).
Acknowledgements
We regret that page limitations precluded us from citing
numerous relevant references. The authors’ work embodied
in this article was supported in parts by NIH Grants to S.K.D.
S.K.D. is the recipient of Method to Extend Research in Time
(MERIT) Awards from the National Institute on Drug Abuse
(NIDA) and the National Institute of Child Health and Human
Development (NICHD). H.W. is the recipient of Solvay/Mortola
Research Award from the Society for Gynecologic
Investigation. We thank S. Tranguch for critical reading of the
manuscript.
Competing interests statement
The authors declare no competing financial interests.
DATABASES
The following terms in this article are linked online to:
Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.
fcgi?db=gene
Bmp2 | Cb1 | Cdx2 | Cox1 | Cox2 | cPla2
α
| Crim1 | Dan | Eomes
| Er
α
| Er
β
| ErbB1 | ErbB4 | Fgf2 | Ffg10 | Fkbp52 | Flk1 | Hbegf |
Hmx3 | Hoxa10 | Hoxa11 | Ihh | L-selectin | Lif | lpA3 | Msx1 |
Nanog | Noggin | Oct4 | PRA | PRB | sfrp4 | Wnt4 | Wnt7a
UniProtKB: http://ca.expasy.org/sprot/
gp130 | PPARδ | VEGF
FURTHER INFORMATION
Sudhansu K. Dey’s web page: http://www.mc.vanderbilt.
edu/reproductionlab/index.html
NIA Mouse cDNA Project: http://lgsun.grc.nia.nih.gov/
cDNA/CitationFinder.html
SUPPLEMENTARY INFORMATION
See online article: S1 (table)
Access to this links box is available online
REVIEWS
NATURE REVIEWS
|
GENETICS VOLUME 7
|
MARCH 2006
|
199
... Embryo implantation is a highly coordinated maternalembryonic communication process (1). Successful embryo implantation requires an implantable blastocyst and a receptive uterus. ...
... The maximum receptive period of the endometrium for the embryo is known as the implantation window (1). In mice, the implantation window typically opens on 4.5-5 dpc and lasts approximately 24 h. ...
... In mice, the implantation window typically opens on 4.5-5 dpc and lasts approximately 24 h. During this period, the embryo undergoes positioning, adhesion, and invasion, ultimately completing the implantation process (1,55,56). Even minor alterations in the uterine microenvironment during the implantation window can disrupt the process of embryo implantation. ...
Article
Full-text available
Introduction Superovulation is a critical step in assisted reproductive technology, but the use of human chorionic gonadotropin (hCG) as a trigger for superovulation can result in ovarian hyperstimulation. Thus, the use of Gonadotropin-releasing hormone agonist (GnRHa) trigger has been increasingly adopted, although it has been associated with a higher rate of pregnancy failure compared to natural cycles. This study aimed to investigate the effect of GnRHa trigger on embryo implantation in a mouse model. Methods Mice in the superovulation (PG) group were administered 7.5 IU of PMSG, followed by the injection of 3.5 μg of GnRHa (Leuprorelin) 48 h later, while mice in the control group (CTR) mated naturally. We compared the number of oocytes, blastocysts, and corpus luteum between the two groups and the implantation sites after the transfer of natural blastocysts. Ovaries, uterus, and serum 2 and 4 days after mating were collected for qRT-PCR, transcriptome sequencing, and hormone assays. Results The PG group had more oocytes, blastocysts, and corpus luteum after superovulation than the CTR group. However, the mRNA expression of leukemia inhibitory factor (Lif) and the number of implantation sites were reduced in the PG group. The ELISA assay revealed that superovulation increased ovarian estrogen secretion. The transcriptome analysis showed that superphysiological estrogen led to a response of the uterus to a high estrogen signal, resulting in abnormal endometrium and extracellular matrix remodeling and up-regulation of ion transport and inflammation-related genes. Conclusion Our findings suggest that a combination of PMSG and GnRHa trigger impaired embryo implantation in mice, as the excessive uterine response to superphysiological estrogen levels can lead to the change of gene expression related to endometrial remodeling, abnormal expression of uterine ion transport genes and excessive immune-related genes.
... Success requires synchronization between a competent embryonic blastocyst and a receptive uterus. There is a temporally restricted "implantation window" regulated by proliferation and differentiation of endometrial epithelium and stroma under the control of progesterone (P 4 ) and estrogen (E 2 ) (3,4). Uterine epithelial-stromal crosstalk involves endocrine, paracrine, and juxtacrine interactions that are critical for successful implantation (1,3). ...
... There is a temporally restricted "implantation window" regulated by proliferation and differentiation of endometrial epithelium and stroma under the control of progesterone (P 4 ) and estrogen (E 2 ) (3,4). Uterine epithelial-stromal crosstalk involves endocrine, paracrine, and juxtacrine interactions that are critical for successful implantation (1,3). Developmental programs are precisely controlled by chromatin regulators that maintain specific gene expression through epigenetic modification of the genome. ...
... ualization that is essential for normal pregnancy (3). Employing an oil-induced decidualization assay (26), the Pbrm1 mutant uteri showed a remarkably reduced decidual response (Supplemental Figure 1, A and B). ...
Article
Full-text available
Early gestational loss occurs in approximately 20% of all clinically recognized human pregnancies and is an important cause of morbidity. Either embryonic or maternal defects can cause loss, but a functioning and receptive uterine endometrium is crucial for embryo implantation. We report that the switch/sucrose nonfermentable (SWI/SNF) remodeling complex containing polybromo-1 (PBRM1) and Brahma-related gene 1 (BRG1) is essential for implantation of the embryonic blastocyst on the wall of the uterus in mice. Although preimplantation development is unaffected, conditional ablation of Pbrm1 in uterine stromal cells disrupts progesterone pathways and uterine receptivity. Heart and neural crest derivatives expressed 2 (Hand2) encodes a basic helix-loop-helix (bHLH) transcription factor required for embryo implantation. We identify an enhancer of the Hand2 gene in stromal cells that requires PBRM1 for epigenetic histone modifications/coactivator recruitment and looping with the promoter. In Pbrm1cKO mice, perturbation of chromatin assembly at the promoter and enhancer sites compromises Hand2 transcription, adversely affects fibroblast growth factor signaling pathways, prevents normal stromal-epithelial crosstalk, and disrupts embryo implantation. The mutant female mice are infertile and provide insight into potential causes of early pregnancy loss in humans.
... Embryo implantation is a critical step in embryo development and pregnancy outcome, and succesful implantation is highly depend on uterine receptivity. During implantation, the trophectoderm cells of the blastocyst interact with the luminal epithelium of the uterus in a series of defined events (Dey et al. 2004;Wang and Dey 2006). The purpose of implantation is to attach the developing embryo to the endometrial stroma, which is regulated by ovarian hormones and locally produced signaling molecules, including cytokines and growth factors (Dey et al. 2004). ...
Article
Full-text available
Obesity is defined by increased adipose tissue volume and has become a major risk factor for reproduction. Recent studies have revealed a substantial link between obesity and epigenetics. The epigenome is dynamically regulated mainly by DNA methylation. DNA methylation, which is controlled by DNA methyltransferases (Dnmts), has been widely studied because it is essential for imprinting and regulation of gene expression. In our previous study, we showed that the levels of Dnmt1, Dnmt3a and global DNA methylation was dramatically altered in the testis and ovary of high-fat diet (HFD)-induced obese mice. However, the effect of HFD on Dnmts and global DNA methylation in mouse uterus has not yet been demonstrated. Therefore, in the present study, we aimed to evaluate the effect of HFD on the level of Dnmt1, Dnmt3a, Dnmt3b, Dnmt3l and global DNA methylation in uterus. Our results showed that HFD significantly altered the levels of Dnmts and global DNA methylation in the uterus. The total expression of Dnmt1, Dnmt3a and Dnmt3b was significantly upregulated, while level of Dnmt3l and global DNA methylation were dramatically decreased (p < 0.05). Furthermore, we observed that the expression of Dnmt3b and Dnmt3l was significantly increased in endometrium including gland and epithelium (p < 0.05). Although Dnmt3b was the only protein whose expression significantly increased, the level of global DNA methylation and Dnmt3l significantly decreased in stroma and myometrium (p < 0.05). In conclusion, our results show for the first time that obesity dramatically alters global DNA methylation and expression of Dnmts, and decreased DNA methylation and Dnmt expression may cause abnormal gene expression, especially in the endometrium.
... In the proliferative phase, estrogen (E 2 ) stimulates the proliferation of endometrial epithelial cells (EEC) and stromal cells (ESCs) and the expression of the progesterone receptor (PGR) in these cells. A surge in luteinizing hormone secreted from the pituitary gland triggers ovulation and increases serum levels of progesterone (P4), which transitions the endometrium from the proliferative to the secretory phase [3] . After ovulation, the circulating P4 and intracellular cyclic adenosine monophosphate (cAMP) surge induces decidualization of ESCs and remodeling of the endometrial tissue during pregnancy preparation [4] . ...
Article
Full-text available
Endometrial stromal cell decidualization is a crucial step in endometrial remodeling during pregnancy. Decidualization is controlled by orchestrated ovarian hormones, followed by the activation of various downstream signaling pathways. Accumulating evidence has shown multiple functions of decidualized endometrial stromal cells during embryo implantation, including tissue remodeling, antioxidative stress, angiogenesis, and immune tolerance. The distinct secretomes of decidualized stromal cells also reveal their intensive interactions with epithelial, endothelial, and immune cells. However, aberrant decidualization leads to pregnancy failures, such as recurrent pregnancy loss and repeated implantation failure. This review aimed to provide an overview of the molecular mechanisms underlying the divergent functions of decidualized endometrial stromal cells and their potential clinical applications. Moreover, the use of single-cell RNA sequencing data further enhances our understanding of these biological processes. This review discusses decidualization-related signaling pathways that serve as potential therapeutic targets for treating implantation failure in in vitro fertilization and provides novel approaches to investigate the underlying causes of female infertility.
... ). It has been demonstrated that mTOR-deficient embryos die immediately after implantation (29).In contrast to our results, some studies showed that glucocorticoids, such as dexamethasone, could inhibit the mTOR signaling pathways in hypothalamic organotypic cultures as validated by decreased phosphorylation of 4E-BP1, as a downstream mediator of the mTOR protein, during implantation(74)(75)(76). In opposite to these results, another study reported that 4E-BP1 would not be altered by the treatment with dexamethasone(77). ...
... Because of our past human studies [33,35], our initial objective in using the Plzf d/d mouse was to determine the in vivo importance of PLZF in murine endometrial stromal cell decidualization, which normally occurs at GD6 onwards [77]. To bypass the earlier Plzf d/d endometrial receptivity phenotype, the well-described murine artificial decidual response assay was used to determine PLZF's endometrial intrinsic role in the decidualization of this tissue [48]. ...
Article
Full-text available
Using an established human primary cell culture model, we previously demonstrated that the promyelocytic leukemia zinc finger (PLZF) transcription factor is a direct target of the progesterone receptor (PGR) and is essential for progestin-dependent decidualization of human endometrial stromal cells (HESCs). These in vitro findings were supported by immunohistochemical analysis of human endometrial tissue biopsies, which showed that the strongest immunoreactivity for endometrial PLZF is detected during the progesterone (P4)-dominant secretory phase of the menstrual cycle. While these human studies provided critical clinical support for the important role of PLZF in P4-dependent HESC decidualization, functional validation in vivo was not possible due to the absence of suitable animal models. To address this deficiency, we recently generated a conditional knockout mouse model in which PLZF is ablated in PGR-positive cells of the mouse (Plzf d/d). The Plzf d/d female was phenotypically analyzed using immunoblotting, real-time PCR, and immunohistochemistry. Reproductive function was tested using the timed natural pregnancy model as well as the artificial decidual response assay. Even though ovarian activity is not affected, female Plzf d/d mice exhibit an infertility phenotype due to an inability of the embryo to implant into the Plzf d/d endometrium. Initial cellular and molecular phenotyping investigations reveal that the Plzf d/d endometrium is unable to develop a transient receptive state, which is reflected at the molecular level by a blunted response to P4 exposure with a concomitant unopposed response to 17-β estradiol. In addition to a defect in P4-dependent receptivity, the Plzf d/d endometrium fails to undergo decidualization in response to an artificial decidual stimulus, providing the in vivo validation for our earlier HESC culture findings. Collectively, our new Plzf d/d mouse model underscores the physiological importance of the PLZF transcription factor not only in endometrial stromal cell decidualization but also uterine receptivity, two uterine cellular processes that are indispensable for the establishment of pregnancy.
Article
Full-text available
Cellular responses to the steroid hormones, estrogen (E2), and progesterone (P4) are governed by their cognate receptor’s transcriptional output. However, the feed-forward mechanisms that shape cell-type-specific transcriptional fulcrums for steroid receptors are unidentified. Herein, we found that a common feed-forward mechanism between GREB1 and steroid receptors regulates the differential effect of GREB1 on steroid hormones in a physiological or pathological context. In physiological (receptive) endometrium, GREB1 controls P4-responses in uterine stroma, affecting endometrial receptivity and decidualization, while not affecting E2-mediated epithelial proliferation. Of mechanism, progesterone-induced GREB1 physically interacts with the progesterone receptor, acting as a cofactor in a positive feedback mechanism to regulate P4-responsive genes. Conversely, in endometrial pathology (endometriosis), E2-induced GREB1 modulates E2-dependent gene expression to promote the growth of endometriotic lesions in mice. This differential action of GREB1 exerted by a common feed-forward mechanism with steroid receptors advances our understanding of mechanisms that underlie cell- and tissue-specific steroid hormone actions.
Article
Full-text available
Regulated cell death (RCD) plays a fundamental role in placental development and tissue homeostasis. Placental development relies upon effective implantation and invasion of the maternal decidua by the trophoblast and an immune tolerant environment maintained by various cells at the maternal-fetal interface. Although cell death in the placenta can affect fetal development and even cause pregnancy-related diseases, accumulating evidence has revealed that several regulated cell death were found at the maternal-fetal interface under physiological or pathological conditions, the exact types of cell death and the precise molecular mechanisms remain elusive. In this review, we summarized the apoptosis, necroptosis and autophagy play both promoting and inhibiting roles in the differentiation, invasion of trophoblast, remodeling of the uterine spiral artery and decidualization, whereas ferroptosis and pyroptosis have adverse effects. RCD serves as a mode of communication between different cells to better maintain the maternal-fetal interface microenvironment. Maintaining the balance of RCD at the maternal-fetal interface is of utmost importance for the development of the placenta, establishment of an immune microenvironment, and prevention of pregnancy disorders. In addition, we also revealed an association between abnormal expression of key molecules in different types of RCD and pregnancy-related diseases, which may yield significant insights into the pathogenesis and treatment of pregnancy-related complications.
Article
Embryo implantation is composed of three steps: blastocyst apposition, adhesion/attachment and invasion. Blastocyst invasion has been studied less extensively than the other two events. Historically, studies conducted using electron microscopy have shown the removal of epithelial cells in the vicinity of the attached blastocysts in rodents, although the underlying mechanisms have remained unclear. Here, we describe recent studies using mice with uterine‐specific gene deletion that demonstrated important roles for nuclear proteins such as progesterone receptor, hypoxia inducible factor and retinoblastoma in the regulation of embryo invasion. In these mouse models, the detachment of the endometrial luminal epithelium, decidualization in the stroma, and the activation of trophoblasts have been found to be important in ensuring embryo invasion. This review summarizes the molecular signaling associated with these cellular events, mainly evidenced by mouse models.
Article
Full-text available
Cyclooxygenase (COX) is the rate-limiting enzyme in the biosyn- thesis of PGs. PGs together with ovarian steroids play important regulatory roles in the establishment and maintenance of pregnancy in a number of different species. In the primate, little is known about the role of PGs in these processes. In this study, the uterine expression of COX-1 and COX-2 throughout the menstrual cycle (late follicular, day 5 postovulation (PO), day 10 PO, and day 14 PO) and pregnancy (days 12-18, day 39, day 51, and near term) was analyzed using semiquantitative RT-PCR, in situ hybridization, and immunocyto- chemistry. During the menstrual cycle, the highest expression of COX-1 occurred in luteal phase endometrium and was localized to the surface and glandular epithelium. The stromal cells did not express detectable levels of COX-1 at any time. COX-2 messenger RNA (mRNA) expression, as measured by RT-PCR, was evident at all stages of the menstrual cycle, and in situ hybridization showed spe- cific localization for this mRNA in the epithelial cells during the cycle. Treatment of animals with the antiprogestin (ZK 137.316) for 9 days (beginning on the day of the LH surge) inhibited COX-1 expression in the epithelium when the tissue was analyzed on day 10 PO, whereas COX-2 expression disappeared in the epithelium and increased in the stroma. With the onset of pregnancy, COX-1 expression in epithelial cells decreased dramatically. In contrast, COX-2 continued to be de- tected on the surface epithelium and was also strongly expressed specifically in the stromal cells at the site of implantation. Immuno- cytochemical staining for COX-2 showed the same pattern of expres- sion for the protein as the message. Finally, near-term decidua ex- pressed very little COX-1 or COX-2 mRNA. These studies suggest that in the baboon endometrium, COX-1 expression is regulated primarily by progesterone, whereas regulation of COX-2 expression may involve additional mediators of embryonic origin at the site of implantation. (Endocrinology 140: 2672-2678, 1999)
Article
Full-text available
Embryo-uterine interactions leading to the attachment reaction is followed by stromal cell proliferation and differentiation into decidual cells (decidualization) at the sites of blastocyst apposition. In rodents, decidualization is also induced by application of an artificial stimulus (intraluminal oil infusion) in a pseudopregnant uterus, or to one that has been appropriately prepared by exogenous progesterone (P4) and estrogen. The process of decidualization is under the control of these steroids in the presence of blastocysts or deciduogenic stimuli. Al- though it is well known that estrogen is required for the induction of progesterone receptors in the uterus, the functional importance of estrogen in the process of decidualization is poorly understood. To better understand the role of estrogenic actions in decidualization, we used wild-type and estrogen receptor-a knock-out (ERKO) mice for induction of decidualization employing a defined steroid hormonal treatment schedule. Our results demonstrate that P4 alone induces decidualization in ovariectomized wild-type or ERKO mice in re- sponse to intraluminal oil infusion in the absence of estrogen. A combined treatment of either estradiol-17b (E2) or its catecholme- tabolite 4-hydroxyestradiol-17b (4-OH-E2) with P4 does not potentiate the decidual response produced by P4 treatment alone in either ovari- ectomized wild-type or ERKO mice. The induction of decidual re- sponse was associated with up-regulation of decidual cell marker genes, such as progesterone receptor, metallothionein-1, and cyclo- oxygenase-2. The results suggest that the stromal cell sensitivity to decidualization is critically dependent on P4-regulated events, and estrogenic induction of progesterone receptor via classical nuclear ER-a is not critical for this process. (Endocrinology 140:2704 -2710, 1999)
Article
Full-text available
The earliest cell fate decision in the mammalian embryo separates the extra-embryonic trophoblast lineage, which forms the fetal portion of the placenta, from the embryonic cell lineages. The body plan of the embryo proper is established only later at gastrulation, when the pluripotent epiblast gives rise to the germ layers ectoderm, mesoderm and endoderm. Here we show that the T-box gene Eomesodermin performs essential functions in both trophoblast development and gastrulation. Mouse embryos lacking Eomesodermin arrest at the blastocyst stage. Mutant trophoectoderm does not differentiate into trophoblast, indicating that Eomesodermin may be required for the development of trophoblast stem cells. In the embryo proper, Eomesodermin is essential for mesoderm formation. Although the specification of the anterior- posterior axis and the initial response to mesoderm-inducing signals is intact in mutant epiblasts, the prospective mesodermal cells are not recruited into the primitive streak. Our results indicate that Eomesodermin defines a conserved molecular pathway controlling the morphogenetic movements of germ layer formation and has acquired a new function in mammals in the differentiation of trophoblast.
Article
High density cDNA microarray screening was used to determine changes in gene expression occurring during the transition between the early luteal (prereceptive) and mid-luteal (receptive) phases in human endometrium. Of ~12 000 genes profiled, 693 (5.8%) displayed >2-fold differences in relative levels of expression between these stages. Of these, 370 genes (3.1%) displayed decreases ranging from 2- to >100-fold while 323 genes (2.7%) displayed increases ranging from 2- to >45-fold. Many genes correspond to mRNAs encoding proteins previously shown to change in a similar manner between the proliferative and mid-luteal phases, serving as one validation of the microarray screening results. In addition, novel genes were identified. Genes encoding cell surface receptors, adhesion and extracellular matrix proteins and growth factors accounted for 20% of the changes. Several genes were studied further by Northern blot analyses. These results confirmed that claudin-4/Clostridium perfringens enterotoxin (CPE) receptor and osteopontin (OPN) mRNA increased ~4- and 12-fold respectively, while betaig-H3 (BIGH3) decreased >80% during the early to mid-luteal transition. Immunostaining also revealed strong specific staining for claudin-4/CPE, EP1 and prostaglandin receptor in epithelia, and leukotriene B4 receptor in both epithelia and stroma, at the mid-luteal stage. Collectively, these studies identify multiple new candidate markers that may be used to predict the receptive phase in humans. Some of these gene products, e.g. OPN, may play direct roles in embryo–uterine interactions during the implantation process.
Article
In the mouse, estrogen is essential for blastocyst implantation in the progesterone (P4)-primed uterus. The mechanism(s) by which estrogen initiates this response still remains elusive. The present investigation, using delayed implantation in the mouse, examined the differential role of estradiol-17β (E2) and its catechol metabolite 4-hydroxy-E2 (4-OH-E2) in uterine and blastocyst activation for implantation. The conditions of delayed implantation were induced by ovariectomizing mice on day 4 (day 1 = vaginal plug) of pregnancy or pseudopregnancy and maintaining them with P4 from days 5–7. The binding of EGF to blastocysts was used as a marker for blastocyst activation. Our results show that whereas E2 fails to activate dormant blastocysts (with respect to EGF binding in vitro), 4-OH-E2, cAMP, or prostaglandin E2, is effective in this response. Further, whereas 4-OH-E2 induced-activation is not blocked by an antiestrogen, an inhibitor of PG synthesis, adenylyl cyclase or protein kinase A effectively blocks this activation. These results suggest that 4-OH-E2 effects on blastocysts are mediated by PGs, which, in turn, stimulate cAMP production and thus activation of protein kinase A. Two-fluoro-E2 is a poor substrate and an inhibitor of catecholestrogen synthesis, but it is estrogenic, with respect to uterine growth and gene expression. Using blastocyst transfer experiments, we observed that dormant blastocysts incubated with 4-OH-E2 in vitro, but not with E2, are capable of implanting in P4-treated delayed implanting mice receiving two-fluoro-E2. The results suggest that whereas E2 is necessary for preparation of the uterus, uterine-derived catecholestrogen is important for blastocyst activation for implantation. Indeed, the receptive uterus has the capacity to synthesize 4-OH-E2. Collectively, we demonstrate that the primary ovarian estrogen E2, via its interaction with nuclear estrogen receptors, participates in the preparation of the P4-primed uterus to the receptive state in an endocrine manner, whereas its metabolite 4-OH-E2, produced from E2 in the uterus, mediates blastocyst activation for implantation in a paracrine manner. Our results also establish that these target-specific effects of primary estrogen and catecholestrogen are both essential for implantation and that successful implantation occurs only when the activated stage of the blastocyst coincides with the receptive state of the uterus.
Article
The response of the human endometrium to the ovarian hormones, estrogen and progesterone, has been the focus of decades of research. In order to understand this critical aspect of endometrial physiology, we undertook a genome-wide analysis of transcript abundance and changes in transcript level between normal endometrium in the proliferative and secretory phases of the menstrual cycle. A high-density, oligonucleotide gene array, comprising 60 000 gene targets, was used to define the gene expression profile of proliferative and secretory phase endometrium. Results from the arrays were verified using real-time PCR. The expression levels of 149 transcripts differed significantly between the two phases of the cycle determined by stringent range limits (99.99%), calculated using local variance values. These transcripts include previously documented steroidally responsive genes (such as placental protein 14 and stromelysin-3) and novel transcripts not previously linked to either endometrial physiology or steroid regulation (such as intestinal trefoil factor and a number of expressed sequence tags). Examination of the 5¢ promoter regions of these genes identified many putative estrogen and progesterone receptor DNA binding domains, suggesting a direct response of these genes to the ovarian hormones.
Article
Cyclooxygenase (COX) is the rate-limiting enzyme in the synthesis of prostaglandins (PGs) and exists in two isoforms, COX-1 and COX-2. In spite of long-standing speculation, definitive roles of PGs in various events of early pregnancy remain elusive. We demonstrate herein that the targeted disruption of COX-2, but not COX-1, in mice produces multiple failures in female reproductive processes that include ovulation, fertilization, implantation, and decidualization. Using multiple approaches, we conclude that these defects are the direct result of target organ–specific COX-2 deficiency but are not the result of deficiency of pituitary gonadotropins or ovarian steroid hormones, or reduced responsiveness of the target organs to their respective hormones.