ArticlePDF Available

Role of Smad- and Wnt-Dependent Pathways in Embryonic Cardiac Development

Authors:

Abstract and Figures

The development of the heart is essential for embryogenesis and precedes development of other organs. However, the mechanisms involved in embryonic cardiac development are ill-defined. Recent evidence suggests that Smad and Wnt signaling pathways are important in stem cell fate determination and their commitment to cardiovascular differentiation. We have previously reported that bone morphogenetic proteins (BMP)-2, -5, and -7 and fibroblast growth factors (FGF)-2 and -4 secreted from the adjoining endodermal cells favor cardiac differentiation in murine embryonic stem (ES) cells. Here, we demonstrate that BMP-2, -5, and -7 stimulate receptor-activated Smad1, 5, and 8, which in turn causes oligomerization of Smad4 in the nucleus. We further delineate the role of Wnt signaling pathway as evidenced by induction of Wnt3 and Wnt8b, stimulation of FRP-1, inhibition of GSK-B, accumulation of cytosolic beta-catenin, and transcription of target genes, including c-myc and cyclin-D1. We also ascertained the specificity of BMP- and Wnt-evoked activation of signaling cascades. Our data are consistent with the hypothesis that BMP-dependent activation of transcription factors including GATA-4, Nkx2.5, and MEF-2C augments cardiac differentiation mediated by cooperative control of Smad and Wnt signaling pathways. Our results provide a solid foundation for further study of the biochemistry of cardiac differentiation from stem cells.
Content may be subject to copyright.
STEM CELLS AND DEVELOPMENT 15:29–39 (2006)
© Mary Ann Liebert, Inc.
Original Research Report
Role of Smad- and Wnt-Dependent Pathways in Embryonic
Cardiac Development
RAJARSHI PAL and APARNA KHANNA
ABSTRACT
The development of the heart is essential for embryogenesis and precedes development of other or-
gans. However, the mechanisms involved in embryonic cardiac development are ill-defined. Recent
evidence suggests that Smad and Wnt signaling pathways are important in stem cell fate determi-
nation and their commitment to cardiovascular differentiation. We have previously reported that
bone morphogenetic proteins (BMP)-2, -5, and -7 and fibroblast growth factors (FGF)-2 and -4 se-
creted from the adjoining endodermal cells favor cardiac differentiation in murine embryonic stem
(ES) cells. Here, we demonstrate that BMP-2, -5, and -7 stimulate receptor-activated Smad1, 5, and
8, which in turn causes oligomerization of Smad4 in the nucleus. We further delineate the role of
Wnt signaling pathway as evidenced by induction of Wnt3 and Wnt8b, stimulation of FRP-1, inhi-
bition of GSK-B, accumulation of cytosolic -catenin, and transcription of target genes, including
c-myc and cyclin-D1. We also ascertained the specificity of BMP- and Wnt-evoked activation of sig-
naling cascades. Our data are consistent with the hypothesis that BMP-dependent activation of tran-
scription factors including GATA-4, Nkx2.5, and MEF-2C augments cardiac differentiation mediated
by cooperative control of Smad and Wnt signaling pathways. Our results provide a solid foundation
for further study of the biochemistry of cardiac differentiation from stem cells.
29
INTRODUCTION
E
MBRYONIC STEM
(ES)
CELLS
are a population of multi-
potent, self-renewing cells that are derived from the
epiblast of mammalian blastocyst embryos and retain this
developmental identity even after prolonged culture in vitro
(1). The recent isolation and culture of human embryonic
stem (hES) cells (2) attracts two important considerations.
The first is their potential application in regenerative med-
icine, and the second is their experimental usefulness in de-
lineating important developmental signaling pathways, and
the potential they offer for regenerative medicine, as well
as the delineation of the events involved in early human
development and the signaling pathways activated during
development. Pluripotent ES cells can be induced to dif-
ferentiate into a myriad of tissues belonging to the three
germ layers, including ectoderm, mesoderm and endoderm
in vitro after aggregation into three-dimensional structures
termed embryoid bodies (EBs) (3). EBs give rise to a va-
riety of specialized cell types, including cardiomyocytes,
that manifest by the appearance of spontaneously contract-
ing foci (4). Thus, cardiac differentiation of EBs is a suit-
able in vitro model to study the signaling pathways in heart
development from mouse ES cells.
The heart is one of the first organs to develop in a de-
veloping embryo and ensures the distribution of vital nu-
trients within the growing organism. Truly, the well-or-
chestrated morphological and molecular events that result
in the formation of this complex organ are intriguing. Al-
though the cardiac-promoting role of many growth fac-
tors has been thoroughly investigated, our current knowl-
edge about regulatory events leading to heart formation
is mostly based on previously characterized transcription
factors that belong to very different gene families. One
Embryonic Stem Cell Group, Reliance Life Sciences, Ltd., Navi Mumbai-400 701, India.
of the biggest challenges now is to identify molecules
that regulate these cardiac transcription factors and to un-
derstand their intertwined regulatory relationships.
Dissecting the genetic pathways involved in cardio-
genesis has revealed crucial roles for members of vari-
ous growth factor families in this process. For example,
the combination of bone morphogenetic proteins 2/4
(BMP-2/4) belonging to the transforming growth factor-
(TGF-) family, and fibroblast growth factor-4 (FGF-
4) is capable of converting avian posterior mesoderm,
which normally does not contribute to the heart, into car-
diac tissue (5,6). Similarly, FGF/FGFR signaling also
plays important functions in heart formation and devel-
opment (7).
Likewise, various growth factors appear to exert ef-
fects in early hepatogenesis through similar tissue inter-
actions. Studies have shown that the cardiogenic meso-
derm, which is transiently opposed to the prospective
hepatic endoderm, provides a signal that induces liver
progenitors in the endoderm (8). Although this inductive
property of the precardiac mesoderm was necessary for
determining the hepatic lineage commitment, it was not
sufficient for hepatocyte differentiation. Furthermore,
during induction of the endoderm with the intervention
of precardiac mesoderm via reciprocal signaling, GATA-
4 and HNF-3B are key transcription factors that lead to
induction of downstream effectors of hepatic differenti-
ation (9).
Members of the TGF-family control growth and dif-
ferentiation and have important functions during embry-
onic development (10,11). BMPs are a subclass of the
TGF- superfamily active in the developing heart (12).
BMPs bind to and activate different serine/threonine ki-
nase receptors (BMPR-I and BMPR-II). Upon activation,
BMP receptors recruit and phosphorylate several recep-
tor-regulated Smad transcription factors (Smad1, Smad5,
or Smad-8), which then interact with Smad4, followed
by translocation of the heteromeric Smad complex to the
nucleus where it associates with other transcription fac-
tors to activate specific BMP-responsive genes (13–15).
Three classes of Smads have been defined: the receptor-
regulated Smads (R-Smads); the co-Smads (Smad-4), and
the inhibitory Smads (I-Smads). Smad proteins are re-
cruited to specific target genes via their interactions with
distinct transcriptional cofactors. Investigating the mech-
anism that mediates the selective response of the embry-
onic mesoderm to BMP signaling is likely to provide in-
valuable insight into the molecular basis of cardiac
specification, which is poorly understood. Furthermore,
a separate pathway involving the TGF--activated kinase
1 (TAK1) and p38 mitogen-activated protein kinase
(MAPK) is reportedly activated by BMPs in some cells
(16,17).
In contrast to the well-investigated roles of the signal-
ing molecules described above, the function of Wnt fam-
ily members in vertebrate cardiogenesis is currently un-
der investigation. Wnt/Wg genes, related to wingless in
Drosophila, encode a number of secreted proteins that
play critical roles in the development of many organisms,
especially in cell fate and patterning (18–20). Once Wnt
molecules have bound to their receptors, the cytosolic
phosphoprotein Dsh or Frz becomes activated, which, in
turn, leads to inactivation of GSK-3. Inhibition of GSK-
3 leads to elevated levels of cytosolic -catenin. GSK-3
inhibits the Wnt pathway, by phosphorylating amino-ter-
minal -catenin residues, directing -catenin toward the
degradation pathway. In regulating the stability of cy-
tosolic -catenin, GSK-3 is accompanied by at least three
different molecules: adenomatous polyposis coli (APC),
Axin (also known as conductin), and GSK-3 binding pro-
tein (GBP). APC contains a -catenin, as well as a GSK-
3 binding domain (21). But the role of Wnt proteins in
heart development is complex, which arises from the ex-
istence of different Wnt signal transduction cascades. To
date, only Wnt-3, Wnt-8, and Wnt-11 have been ascribed
a promoting role in cardiac differentiation from the
mouse embryonic carcinoma stem cell line P19 (22,23).
However, this does not exclude positive regulatory roles
for other Wnt proteins expressed in cardiac tissue. Tar-
geted gene disruptions of different Wnt genes in mice
suggest that the functions of these proteins are inter-
changeable.
Almost all of the reports on the reciprocal signaling of
the endodermal derivatives in cardiogenesis are based on
tissue explant systems in chick, quail, or Xenopus. In our
previous study, we have shown a simple and effective
strategy for co-differentiation of mouse ES cells into car-
diac myocytes and hepatocyte-like cells without using
exemplary cardiotrophic factors and also without em-
ploying an in vitro co-culture system. We have also dem-
onstrated that BMPs and FGFs secreted from the sur-
rounding hepatocyte-like cells play the role of intrinsic
signals in differentiation of cardiomyocytes (24).
In light of the above considerations, we undertook the
present study to define further the obligatory role of Smad
molecules as effectors of downstream events leading to
cardiac differentiation from mouse ES cells. The activa-
tion of Wnt signaling pathway in cardiogenic develop-
ment is implicated by our results.
MATERIALS AND METHODS
Mouse ES cell culture, co-differentiation of
cardiomyocytes, and hepatocyte-like cells
J1 mouse ES cells (ATCC, Manassas, VA) were grown
on a mitotically inactivated (mitomycin-c) mouse embry-
onic fibroblast feeder layer. The medium consisted of Dul-
becco’s modified Eagle medium (DMEM) (no-pyruvate,
PAL AND KHANNA
30
high-glucose formulation; GIBCO-BRL, Grand Island, NY)
supplemented with 15% fetal bovine serum (FBS; Hyclone,
Logan, UT); 1 nonessential amino acids, 2.0 mM gluta-
mine, 1,000 U/ml mouse LIF/ESGRO
®
, 100 M 2-mer-
captoethanol, 100 U penicillin, and 100 g/ml streptomycin
(all from GIBCO-BRL). This medium inhibits differentia-
tion of ES cells and is used until differentiation is initiated.
To initiate differentiation, cells were first cultured on 0.1%
gelatin- (Sigma, St. Louis, MO) coated plates without feeder
layers. For differentiation of ES cells, EBs were generated
by the hanging drop method in suspension culture for 4 days
in the absence of leukemia inhibitory factor (LIF). After
their generation, 10–15 EBs were seeded onto 35-mm tis-
sue culture plates (Nunc, Roskilde, Denmark) precoated
with 0.5% poly-
L
-lysine (Sigma) in DMEM medium sup-
plemented with 15% FBS, 50 nM basic FGF (R&D Sys-
tems, Minneapolis, MN). Rhythmic beating of EB out-
growths, surrounded by oval-shaped hepatocyte-like cells,
was observed on days 8–9 of differentiation, and the cells
continued to beat for more than 30 days in culture.
RNA extraction and RT-PCR analysis
Total RNA was isolated by the TRIzol method (Invit-
rogen) according to the manufacturer’s protocol. A total
of 1 g of RNA treated with RNase-OUT ribonuclease in-
hibitor (Invitrogen, Carlsbad, CA) was used for cDNA syn-
thesis. Reverse-transcription using Superscript reverse
transcriptase-II (Invitrogen) and oligo(dT) (Invitrogen)
was carried out to prime the reaction. PCR primers were
selected to distinguish between cDNA and genomic DNA
by using individual primers specific for different exons.
cDNA (2 l) was amplified by PCR using Abgene 2
PCR master mix (Abgene: Advanced Biotechnologies,
Ltd., Epsom, Surrey, UK) and appropriate primers (see
Table 1). The expression of genes such as GAPDH, TTR,
AFP, albumin, cTnT, -MHC, MLC-2v, Smad-1 and -5,
Smad-4, GATA-4, Nkx2.5, MEF-2C, Wnt8b, Wnt3, -
catenin, c-myc, cyclin-D1, BMP-2, BMP-5, and FGFR-4
were checked. For all of the genes, PCR was performed
for 35 cycles, consisting of an initial denaturation at 94°C
for 1 min, then 94°C for 30 sec, an annealing temperature
of the respective gene primer for 45 sec (for T
m
values of
individual primers, see Table 1), 72°C for 1 min, and was
terminated by final extension at 72°C for 5 min.
Immunochemistry
The differentiated cells were tested for cytoplasmic and
nuclear markers by immunofluorescence analysis as de-
scribed in our earlier report (24). Briefly, EB outgrowths
were mechanically dissected, enzymatically dispersed us-
ing trypsin-EDTA (0.5% trypsin, 0.53 mM EDTA; Life
Technologies, Inc., Grand Island, NY) for 5 min at 37°C;
plated on poly-
L
-lysine-coated two-well-chambered glass
slides, and incubated for 48 h. After fixing the cells with
4% paraformaldehyde, the cells were permeabilized with
0.2% Triton X-100 for 5 min at room temperature. The cells
were then blocked with phosphate-buffered saline (PBS)
containing 1% bovine serum albumin (BSA) for 1 h at room
temperature and were incubated with primary antibody so-
lutions (Smad-4, Santa Cruz, Santa Cruz, CA; -catenin,
Santa Cruz; -MHC, Chemicon, Temecula, CA) diluted
with 1% BSA in PBS overnight at 4°C followed by incu-
bation with secondary antibody solution, coupled with a
fluorescent label fluorescein isothiocyanate (FITC) at room
temperature for 1 h on a rocker. Additionally, cells were
counterstained with DAPI (1 g/ml; Sigma). The slides
Smad AND Wnt SIGNALING IN CARDIAC DIFFERENTIATION
31
T
ABLE
1. D
ETAILS OF
P
RIMER
U
SED
Name of Annealing Product
gene temp (C°) Primer sequence (5-3) size (bp)
Smad-1/5 63 ATGAATGTGACCAGCTTGTTT 349
CTGCTTGGAACCAAATGGGAA
Smad-4 55 AAGGTGGGGAAAGTGAAAC 250
ATGCTTTAGTTCATTCTTGTG
Wnt-3 60 ACACTTGAGCAGAACGGATACA 207
TGGATACAGCAGGTTGGTAGG
Wnt-8b 55 AATGTCTGACTTGAAATGAAA 190
AATGGTTAGAAGAGGTTGGC
-Catenin 62 GCCTGCAGAACTCCAGAAAG 135
GTGGCAAAAACATCAACGTG
Cyclin-D1 62 TCTCCTGCTACCGCACAAC 749
TTCCTCCACTTCCCCCTC
c-myc 60 CGCGCCCAGTGAGGATATC 281
CCACATACAGTCCTGGATGAT
For details of the other primer pairs used, please refer to our previous report (24).
were then mounted with DPX mountant and examined un-
der an inverted fluorescence microscope (Nikon Eclipse
E600, Kanagawa, Japan).
Cell fractionation and western blotting of
EB outgrowths
Spontaneously beating EB outgrowths, each compris-
ing of at least 10
3
cells, were harvested, washed twice
with PBS, and then lysed using M-PER mammalian cell
extraction buffer (Pierce, Rockford, IL). Whole-cell
lysates were centrifuged at 120,000 g for 30 min at
4°C, and the resulting supernatant leaving the debris was
collected. Protein concentrations were determined using
Bradford method (BioRad, Hercules, CA), and 25-g
samples of soluble proteins were boiled in 2% sodium
dodecyl sulfate (SDS) sample buffer (with 1 mM -mer-
captoethanol) for 5 min, then separated on 12% SDS-
polyacrylamide gel electrophoresis (PAGE) gels and
electrically transferred to polyvinylidene fluoride (PVDF)
membranes (Pall Corp., Mumbai, India). Blots were
stained with Red Ponceau-S stain to verify the loading
and transfer of the proteins, blocked with 10% skim milk
in tris-buffered saline-Tween (TBST), and sequentially
probed with a 1:500 dilution of indicated primary anti-
bodies, including Smad-1, -5, and -8, Smad-4, Wnt-3, Wnt-
8b, FRP-1, GSK-3, -catenin (Santa Cruz) overnight at
4°C. Bound antibody was visualized using appropriate
secondary horseradish peroxidase (HRP) or alkaline
phosphatase (ALP) conjugates. The signals were devel-
oped using a 3,3-diaminobenzidine tetrahydrochloride
(DAB) kit (Vector Laboratories, Burlingame, CA) and or
BCIP/NBT alkaline phosphatase substrate tablets (Sigma).
RESULTS
Differentiation of cardiomyocytes interspersed
with hepatocyte-like cells
In our earlier report, we showed the generation of spon-
taneously beating cardiomyocytes surrounded by hepato-
cyte-like cells. Using the same methodology, we used
phase-contrast microscopy, which revealed the appearance
of spontaneously beating EB outgrowths at days 8–10 of
differentiation (Fig. 1A). The rate of the contraction of the
cells was recorded to be at its maximum on the 15th day
of differentiation, and the cardiomyocytes were able to re-
tain the contractility up to 30 days or longer in culture.
Polyhedral to oval-shaped hepatocyte-like cells were con-
firmed to be present surrounding the cardiac bodies (Fig.
1B). The identity of both of the cell types was ascertained
by reverse transcriptase (RT)-PCR (Fig. 1C) using tissue-
specific markers like transthyretin (TTR), alpha feto-pro-
tein (AFP), and albumin for liver-specific gene expression
and cTnT, -MHC, and MLC-2V for heart-specific gene
expression respectively.
Signaling mechanisms induced by BMP-2, -5, and
-7 are mediated through Smad-1, -5, and -8
Smad proteins are important mediators of signaling in-
duced by TGF- family members. It is also known that
Smad-1, -5, and -8 are involved in the TGF- pathway
evoked by BMP and it subsequently binds to the serine/thre-
onine kinase receptors BMPR types I and II. Therefore, we
looked for the activation status of these proteins in in vitro
cardiogenesis in response to the intrinsic signals like BMPs
and FGFs. In our earlier experiments, we have reported the
up-regulation in mRNA levels of BMP-2, -5, and -7 along
with FGFR-2 and -4 stimulated by the adjacent hepatocyte-
like cells. We further investigated the molecular mecha-
nisms behind the initial activation of these signaling mol-
ecules. In our study, we have used noggin, a potent inhibitor
of the BMP/Smad pathway. Noggin at high concentration
is capable of binding BMP ligand with high affinity and
can abolish BMP activity by blocking its binding to a cog-
nate cell-surface receptor (25). Therefore, we treated the
cells with 500 ng/ml of noggin/Fc chimeric protein (Sigma)
on the 4th day of differentiation to antagonize the effect of
PAL AND KHANNA
32
FIG. 1. Co-differentiation of cardiomyocytes and hepatocyte-
like cells from mouse ES cells. Phase-contrast micrographs
showing day-15 contracting cardiomyocytes (A) and oval-
shaped hepatocyte-like cells (B). Scale bars, 50 m. (C) Gene
expression analysis of differentiated cells with liver and cardiac
tissue-specific primers including TTR, AFP, albumin, cTnT,
-MHC, and MLC-2V, respectively. GAPDH is used as a
housekeeping gene control.
intrinsic BMP signaling and to examine the changes in the
downstream events. We had observed that lower concen-
trations of noggin (100 or 200 ng/ml) were not able to block
BMP activity (data not shown). The EB outgrowths were
harvested and used for immunofluorescence and gene ex-
pression analysis, as described in the earlier report. For
western blotting, the cells were lysed in M-PER mammalian
cell extraction buffer. Equal amounts of proteins were sep-
arated by electrophoresis and were transferred to a PVDF
membrane. The membranes were then probed with suitable
antibodies. The actin levels were used as loading controls.
We observed enhanced expression of the Smad-1, -5,
and -8 and Smad-4 on the 15th day of differentiation in
comparison to the undifferentiated mouse ES cells (Fig.
2A,B). After the treatment of the cells with noggin chi-
meric protein, the up-regulation of Smad proteins was re-
verted back to basal levels, as determined by both gene
expression and protein analysis (Fig. 2A,B). Furthermore,
a positive expression of cytoplasmic Smad-4 protein in day
15 differentiated cells was demonstrated by immunostain-
ing with less or no expression in the cells exposed to nog-
gin (Fig. 2C,D,E). Mouse embryonic fibroblast (MEF)
cells were used as a negative control (Fig. 2F).
The results clearly indicate that in response to BMP-
2, -5, and -7 secreted by the hepatocyte-like cells, BMP
receptors recruit Smad-1, -5, and -8, which in turn
translocate to the nucleus and activate the common Smad,
Smad-4. Blocking BMP signaling by application of ex-
ogenous noggin reverses the regulation of the Smad pro-
teins and its downstream effectors.
Activation of cardiogenic transcription factors by
Smad-4 in the nucleus
Upon activation, receptor-activated Smad proteins
(Smad-1, -5, -8) are translocated to the nucleus and in-
teract with the common Smad (Smad-4). In the next step,
Smad-4 oligomer binds to the DNA of the target genes
like GATA-4, Nkx2.5, and MEF-2C, which thereafter
play a pivotal role in specification of the cardiac fate and
morphogenesis of the heart.
Hence, we examined the levels of GAT-4, Nkx2.5, and
Smad AND Wnt SIGNALING IN CARDIAC DIFFERENTIATION
33
FIG. 2. Differential activation of Smad proteins in cardiac differentiation. (A) RT-PCR analysis shows the modulation of Smad-
1 and -5, Smad-4, and the subsequent up-regulation of their downstream effectors like GATA-4, Nkx2.5, and MEF-2C. (B) The
response of Smad-1, -5, and -8, and Smad-4 proteins to the intrinsic BMP signaling could be reversed by the application of nog-
gin, a potent inhibitor of BMP ligand as detected by western blotting. The intensities of the protein expression were normalized
against signals obtained with -actin. (C) Immunochemistry shows cytoplasmic localization of Smad-4 in day-15 differentiated
cells whereas cells treated with noggin (D) were devoid of positively stained cells. (E) Presence of cells was confirmed by coun-
terstaining with the nuclear stain DAPI (1 g/l). (F) MEF cells were stained with goat anti-Smad-4 immunoglobulinG (IgG)
to ascertain the specificity of the antibody. Scale bars, 50 m.
A
B
MEF-2C mRNA in the cells before and after exposure to
noggin. We observed that there was a transient increase
in the expression of these three transcription factors co-
incident with the onset of cardiac differentiation and was
reversible with the treatment of the BMP antagonist.
These data indicate that BMPs favor the commitment of
mouse ES cells into cardiac phenotype via Smad proteins.
Wnt/-catenin pathway is activated at the early
stage of differentiation
Upon Wnt signaling, the constitutive kinase activity of
GSK-3 is inhibited and allows the accumulation of -
catenin in the nucleus. The frizzled form of the trans-
membrane proteins functions as a receptor for the Wnt
family members and aids in the regulation of intracellu-
lar levels of -catenin. -Catenin is essential for the tran-
scriptional activity of TCF/lymphoid enhancing factor
(LEF). TCF-1 and LEF-1 have been shown to be ex-
pressed in largely overlapping, complex patterns during
embryogenesis. Among the number of TCF/LEF target
genes identified (26), we concentrated on c-myc and cy-
clin-D1, both of which have important implications in
understanding the role of Wnt signaling.
We first used RT-PCR analysis to detect the temporal
changes of Wnt-3 and Wnt-8b in the differentiated cells.
Wnt-3 and Wnt-8b exhibited early expression in the un-
differentiated cells. However, both were induced to ele-
vated levels within 15 days of differentiation (Fig. 3A).
Likewise, data from protein analysis by western blotting
support the gene expression results (Fig. 3B). Further-
more, FRP-1, a secreted frizzled-related protein, which
PAL AND KHANNA
34
FIG. 3. Wnt signaling mediates in vitro cardiogenesis from mouse ES cells. (A) RT-PCR analysis confirms the activation of
Wnt-3, Wnt-8b, and -catenin followed by subsequent upregulation of the TCF/LEF target genes like c-myc and cyclin-D1. (B)
Likewise, protein analysis by western blotting supported the transcriptional profiling. FRP-1, being a receptor to Wnt family
members, is up-regulated followed by the down-regulation of GSK-3 in two isoforms, GSK-3, and GSK-3 (52 and 40 kD).
Upon addition, Fz-8/Fc chimeric protein, being an inhibitor of Wnt, was able to block the Wnt signaling and the downstream
events thereafter, as determined by the identifying the reduced levels of the related proteins. (C) -Catenin, the key player in
canonical Wnt signaling, is shown to accumulate in the cells on the 15th day of differentiation and was reduced to lower levels
post Fz-8/Fc treatment as detected by immunochemistry (D). (E) Cells were similarly counterstained with DAPI (1 g/l). (F)
MEF was used to evaluate the specificity of goat anti--catenin IgG. Scale bars, 50 m.
A
B
acts as receptor to Wnt family of proteins, was shown to
be up-regulated on day 15 of differentiation (Fig. 3B).
We extended our examination to check the regulation of
GSK-3, a serine/threonine-directed kinase in this system.
Intriguingly, we observed the presence of two related
forms of GSK-3, GSK-3 and GSK-3 by western blot-
ting in the undifferentiated cells through the differentia-
tion of cardiomyocytes surrounded by hepatocyte-like
cells (Fig. 3B).
To assess whether functional signaling is activated at
the time of Wnt induction, we examined cytosolic -
catenin, the crux of the canonical Wnt signal transduc-
tion pathway (22,27). Wnt stimulated the accumulation
of soluble -catenin (Fig. 3A,B). Simultaneous treatment
with 500 ng/ml Fz-8/Fc chimeric protein (R&D Systems,
USA), an antagonist for Wnt-8a and potentially for other
Wnt proteins (22), decreased -catenin to a basal level
(Fig. 3A,B), as evidenced by RT-PCR and western blot-
ting of whole-cell lysates. This indicates that the accu-
mulation of -catenin is regulated by an autocrine or
paracrine circuit in the system, involving endogenous
Wnt proteins. Conversely, as expected, -catenin was de-
creased upon inhibition of Wnt signaling, as determined
by immunochemistry. As a third criterion, to confirm the
activation of the canonical Wnt pathway, TCF/LEF-de-
pendent transcription of the target genes like c-myc and
cyclin-D1 was evaluated. Recruitment of Wnt proteins
induced a considerable increase in the mRNA levels of
both the genes on day 15 of differentiation (Fig. 3A).
Wnt/-catenin signaling is required to
enhance cardiac differentiation
To understand the possible role of Wnt-mediated sig-
naling in early cardiogenesis, we further monitored dif-
ferentiation induced by the adjoining endodermal cells,
with or without Fz-8/Fc. Treatment with soluble Wnt in-
hibitor prevented GATA-4, Nkx2.5, MEF-2C, and -
MHC induction at least through day 15 of differentiation
(Fig. 4A). Likewise, Fz-8/Fc inhibited the expression of
BMP-2, BMP-5, and FGFR-4 (Fig. 4A), which indicates
that the Wnt pathway lies upstream to the induction of
these cardiac differentiation factors. We extended our ex-
amination to ascertain the direct implication of Wnt in-
duction in cardiac differentiation. We counted the num-
ber of beating regions and found that there was a
significant reduction in the total number of contracting
cardiac bodies on days 9 through 15 in differentiation af-
ter application of Fz-8/Fc (Fig. 4B). Although, there was
also a small decrease in the beating rate in terms of the
beats/minute, no major alteration in the phenotype of the
beating cardiomyocytes was observed. Furthermore, im-
munofluorescence analysis shows that -MHC, a marker
for mature cardiac muscle, is comprehensively expressed
in day-15 beating cardiomyocytes but disappears with the
exposure of Fz-8/Fc inhibitor (Fig. 4C,D). Thus, Wnt sig-
naling plays a critical role not only in early cardiogene-
sis but also in the formation of mature spontaneously
beating cardiomyocytes from ES cells.
Smad AND Wnt SIGNALING IN CARDIAC DIFFERENTIATION
35
FIG. 4. Activation of Wnt signaling modulates the regulation of its target genes. (A) By RT-PCR analysis, a transient increase
in the mRNA levels of the cardiogenic transcription factors like GATA-4, Nkx2.5, MEF-2C, and -MHC was demonstrated. In-
terestingly, Fz-8/Fc was also seen to have an inducing effect on BMP-2, BMP-5, and FGFR-4 expression. (B) Effect of
Fz-8/Fc inhibitor on the number of beating regions. (C) Immunofluorescence analysis shows presence of -MHC protein in day-
15 cardiomyocytes whereas (D) little or no positive expression of the same protein could be detected post Fz-8 treatment. Scale
bars, 50 m.
B
A
C
D
DISCUSSION
The heart is perhaps the first organ to be formed in the
vertebrate embryo. Development and acquisition of a car-
diac fate by embryonal mesodermal cells is a fundamen-
tal step in heart formation, which is crucial for further
development. Several discrete steps initiate heart devel-
opment in general. Commitment to cardiac fate results
from inductive interactions during gastrulation. In am-
phibian and avian embryos, the endoderm adjacent to the
mesodermal cardiac precursors is the source of instruc-
tive signaling capable of specifying a cardiac fate
(28–30). Likewise, most of the recent work on the
prospective role of endoderm-derived growth factors be-
longing to the TGF- superfamily in cardiac develop-
ment has been performed in avian species or primitive
species, such as chick, quail, and Xenopus (12,31). There-
fore, it is important to elucidate when, where, and how
mesodermal cells are instructed to assume the cardiac fate
for understanding the entire body of mechanisms that op-
erate later in heart development. To date, this problem
remains elusive and undefined.
In our earlier report, we demonstrated that during dif-
ferentiation of ES cells, the hepatocyte-like cells appear-
ing adjacent to the spontaneously beating “cardiac bod-
ies” play an important role in regulating the fate of the
ES cells toward the cardiac phenotype by providing nec-
essary intrinsic signals in form of BMPs and FGFs (24).
Prior to this, exogenous application of BMP-2 and -4 was
demonstrated to induce commitment of ES cells to car-
diac differentiation within EBs (32). Conversely, we
demonstrated the differentiation of spontaneously beat-
ing cardiomyocytes authenticated by the expression of
cardiac tissue-specific genes, including cTnT, -MHC,
and MLC-2V (Fig. 1A,C) without using exogenous
BMPs or any other cardiotropic factors.
The Smad signaling pathway is critical for transmit-
ting TGF- superfamily signals from the cell surface to
the nucleus (33,34). TGF-family members initiate their
cellular responses by binding to distinct receptors with
intrinsic serine/threonine kinase activity and activation of
specific downstream intracellular effects termed Smad
proteins. Smad proteins relay the signal from the cell
membranes to the nucleus, where they effect the tran-
scription of target genes by recruiting co-activators and
co-repressors to a wide array of DNA-binding partners
(Fig. 5). The different members of the TGF- family
make use of specific Smad proteins to achieve specificity
(15). BMP usually is shown to induce Smad-1, -5, and
-8 as its downstream effector, whereas TGF-
1
is com-
monly known to stimulate Smad-2 and -3. Thus, we
looked at the activity of Smad-1, -5, and -8 in our in vitro
system. As expected, we noticed increased levels of
Smad-7, -5, and -8 and the common Smad Smad-4 (Fig.
2A,B), which is in concurrence with the earlier reports.
Furthermore, we checked the mRNA and protein levels
of the cardiogenic transcription factors like GATA-4,
Nkx2.5, and MEF-2C as a downstream event. These tran-
scription factors are known to activate the promoters of
several cardiac genes, such as myosin light chain, tro-
ponin T, tropinin I, -MHC, and ANP. Interestingly, we
observed a significant increase in these target molecules,
which may be consequential in inducing formation of
beating cardiomyocytes.
As a third criterion, we verified the involvement of BMPs
in this signaling pathway. To do so, we used a noggin chi-
meric protein to block the BMP activity on day 4 of dif-
ferentiation and checked the endogenous BMP levels (data
not shown) and the downstream regulators of BMP at pro-
gressive days of differentiation. We observed that noggin
was able to recover the elevated levels of Smad-1, -5, and
-8, Smad-4, and the target genes such as GATA-4, Nkx2.5,
and MEF-2C to basal levels (Fig. 2A–C) as studied by RT-
PCR, western blotting, and immunofluorescence analyses.
Our results indicate that during differentiation in response
to BMPs secreted by the adjacent hepatocyte-like cells,
Smad proteins are recruited, thus activating the key car-
diogenic transcription factors favoring formation of spon-
taneously beating cardiomyocytes. However, experiments
are in progress to show the specific binding of Smad
oligomers to the DNA-binding proteins.
BMP has been reported to activate the transcription of
Wnt genes in certain cells (35,36). Recent studies in chick
embryos revealed that Wnt signaling could dictate bio-
logical fate in which BMP signaling is required but not
sufficient, such as neural inhibition of ectoderm, neural
crest induction, and apoptosis (37–39). In addition, Hus-
sein and co-workers have delineated the molecular mech-
anism underlying the induction of Msx2 promoter by co-
operative activation of Wnt and BMP signaling pathways
through the interaction of Smad-4 with the LEF1 tran-
scription factor (40). On the basis of these reports, we
have speculated that convergence of the BMP and Wnt
signaling pathways is also required to enhance the ex-
pression of cardiogenic transcription factors such as
GATA-4, Nkx2.5, and MEF-2C, which in turn may in-
duce cardiac differentiation.
Activation of the Wnt/-catenin signaling cascade was
an early event in cardiac differentiation of pluripotent J1
mouse ES cells, as measured by Wnt-3 and Wnt-8b in-
duction, FRP-1 stimulation, GSK-3 down-inhibition,
accumulation of -catenin, and transcription of TCF/LEF
target genes including c-myc and cyclin-D1 (Fig. 3A–C).
Blocking Wnt receptor interactions with soluble Fz pro-
teins largely or completely blocked the cardiogenic path-
way, including induction of the earliest markers of car-
diac differentiation like GATA-4, Nkx2.5, MEF-2C, and
even -MHC, a late cardiac muscle-specific marker (Fig.
4A). The effect of the activation of these early-stage car-
diac transcription factors on cardiac differentiation was
PAL AND KHANNA
36
ascertained by the decrease in the number of beating re-
gions (Fig. 4B) and subsequent depletion of -MHC
protein expression (Fig. 4C,D) with Fz-8/Fc chimeric
protein. Thus, endogenous Wnt proteins mediate cardio-
genesis in mouse ES cells and do so via the canonical -
catenin pathway. Fz-8 protein itself had an inducing ef-
fect on BMP-2, BMP-5, and FGFR-4, indicating an
indirect role of endogenous BMPs in this system. How-
ever, our conclusions differ from inhibitory roles found
for Wnt-3a and Wnt-8 in Xenopus and chicks (41–43).
We emphasize that apart from potential phylogenetic dis-
similarities, the studies also differ inherently (cultured
cells vs. explants and embryos) in the stage of matura-
tion, perturbations, and diversity of the cell types pres-
ent. But, our results are in agreement with the report by
Pandur and co-workers, showing the stimulation of car-
diogenesis by Wnt-11 via the noncanonical pathway in
Xenopus and P19 EC cells (23). Therefore, BMPs via
Smad signaling and Wnt proteins via accumulation of cy-
tosolic -catenin exert a tight control over mammalian
cardiogenesis at least in mouse ES cells.
Hence, this dual regulation of Wnt and BMP appears
to be frequent in mammalian development. Moreover,
Wnt proteins and BMPs are expressed in many overlap-
ping tissues and, being morphogens, this is consistent
with the idea that a formation of a gradient of ligand
would result in graded expression of common target
genes. But, given the extraordinary diversity and over-
lapping expression of the Wnt ligands and receptors, a
complete genetic analysis of the Wnt family, associated
downstream events and interactions, even confined to the
potential involvement of Wnt proteins in cardiogenesis
is far from being straightforward and comprehensive.
CONCLUSION
The present study was undertaken to understand the
molecular mechanisms behind the inductive role of en-
dodermal derivatives in early cardiogenesis of mouse ES
cells. In summary, our results demonstrate the BMP-2,
-5, and -7 and FGF-2 and -4 secreted by the hepatocyte-
like cells mediate the simultaneous activation of Smad
and Wnt signal transduction pathways and their down-
stream events, leading to enhancement of cardiac myo-
genesis. However, the function of endogenous Smad and
Wnt proteins emerge more clearly from inhibitor studies.
To our knowledge, this is the first report where we have
elucidated the obligatory role of interactive signaling
pathways in in vitro cardiogenesis, thereby posing im-
Smad AND Wnt SIGNALING IN CARDIAC DIFFERENTIATION
37
FIG. 5. Schematic illustration of representative signaling pathways in cardiac myogenesis. Sequential activation of various
genes/proteins at different stages of the Smad signaling cascade is elucidated, which ultimately leads to the regulation of the tar-
get genes in early cardiogenesis like GATA-4, Nkx2.5, and MEF-2C.
portant questions for ES cell biology and mammalian car-
diac development. Further experiments to identify other
players and their relationships in these pathways are in
progress.
ACKNOWLEDGMENTS
The authors are thankful to the Therapeutic Proteins
and Gene Therapy Group (R.L.S.) for providing ALP
conjugates and the developing reagent (Sigma, USA). We
thank Dr. Mahendra S. Rao for his critical review of the
manuscript and helpful suggestions. The authors grate-
fully acknowledge Reliance Life Sciences (http://www.
relbio.com) for providing an opportunity to work on this
project.
REFERENCES
1. Rossant J. (2001). Stem cells from mammalian blastocyst.
Stem Cells 19:477–482.
2. Thomson JA, J Itskovitz-Eldor, SS Shapiro, MA Waknitz,
JJ Swiergiel, VS Marshall and JM Jones. (1998). Embry-
onic stem cell lines derived from human blastocysts. Sci-
ence 282:1145–1147.
3. Desbaillets I, U Ziegler, P Groscurth and M Gassmann.
(2000). Embryoid bodies: an in vitro model of mouse em-
bryogenesis. Exp Physiol 85:645–651.
4. Maltsev VA, J Rohwedel, J Hescheler and AM Wobus.
(1993). Embryonic stem cells differentiate in vitro into car-
diomyocytes representing sinusnodal, atrial and ventricu-
lar cell types. Mech Dev 44:41–50.
5. Lough J, M Barron, M Brogley, Y Sugi, DL Bolender and
X Zhu. (1996). Combined BMP-2 and FGF-4, but neither
factor alone, induces cardiogenesis in non-precardiac em-
bryonic mesoderm. Dev Biol 178:198–202.
6. Barron M, J Gao and J Lough. (2000). Requirement for
BMP and FGF signaling during cardiogenic induction in
non-precardiac mesoderm is specific, transient, and coop-
erative. Dev Dyn 8:383–393.
7. Powers CJ, SW McLeskey and A Wellstein. (2000). Fi-
broblast growth factors, their receptors and signaling. En-
docr Relat Cancer 7:165–197.
8. Le Douarin NM and FV Jotereau. (1975). Tracing of cells
of the avian thymus through embryonic life in interspecific
chimeras. J Exp Med 142:17–40.
9. Zaret KS. (2002). Regulatory phases of early heart develop-
ment: paradigms of organogenesis. Nature Rev 3:499–512.
10. Derynck R, RJ Akhurst and A Balmain. (2001). TGF-beta
signaling in tumor suppression and cancer progression. Na-
ture Genet 29:117–129.
11. Massague J and D Woton. (2000). Transcriptional control
by the TGF-beta/Smad signaling system. EMBO J
19:1745–1754.
12. Schultheiss TM, JB Burch and AB Lassar. (1997). A role
of bone morphogenetic proteins in the induction of cardiac
myogenesis. Genes Dev 11:451–446.
13. Derynck R, Y Zhang and XH Fang. (1998). Smads: tran-
scriptional activators of TGF-beta responses. Cell 95:737–740.
14. Heldin CH, K Miyazono and P ten Dijke. (1997). TGF-
beta signalling from cell membrane to nucleus through
SMAD proteins. Nature 390:465–471.
15. Massague J and YG Chen. (2000). Controlling TGF-beta
signaling. Genes Dev 14:627–644.
16. Eggen BJ, GF Benus, S Folkertsma, LJ Jonk and W Krui-
jer. (2001). TAK1 activation of the mouse JunB promoter
is mediated through a CCAAT box and NF-Y. FEBS Lett
506:267–271.
17. Goswami M, AR Uzgare and AK Sater. (2001). Regula-
tion of MAP kinase by the BMP-4/TAK1 pathway in Xeno-
pus ectoderm. Dev Biol 236:259–270.
18. Arias AM, AM Brown and K Brennan. (1999). Wnt signalling:
pathway or network? Curr Opin Genet Dev 9:447–454.
19. Bejsovec A. (1999). Wnt signaling shows its versatility.
Curr Biol 9:R684–R687.
20. Moon RT, B Bowerman, M Boutros and N Perrimon.
(2002). The promise and perils of Wnt signaling through
beta-catenin. Science 296:1644–1646.
21. Molenaar Mand Destree O. (1999). A tight control over
Wnt action. Int J Dev Biol 43:675–680.
22. Nakamura T, M Sano, Z Songyang and MD Schneider.
(2003). A Wnt- and beta-catenin-dependent pathway for
mammalian cardiac myogenesis. Proc Natl Acad Sci USA
100:5834–5839.
23. Pandur P, M Lasche, LM Eisenberg and M Kuhl. (2002).
Wnt-11 activation of a non-canonical Wnt signalling path-
way is required for cardiogenesis. Nature 418:636–641.
24. Pal R and A Khanna. (2005). Role of hepatocyte-like cells
is differentiation of cardiomyocytes from mouse embryonic
stem cells. Stem Cells Dev 14:153–161.
25. McMahon JA, S Takada, LB Zimmerman, CM Fan, RM
Harland and AP McMahon. (1998). Noggin-mediated an-
tagonism of BMP signaling is required for growth and
patterning of the neural tube and somite. Genes Dev
12:1438–1452.
26. Giles RH, JH van Es and H Clevers. (2003). Caught up in
a Wnt storm: Wnt signaling in cancer. Biochim Biophys
Acta 1653:1–24.
27. Papkoff J, B Rubinfeld, B Schryver and P Polakis. (1996).
Wnt-1 regulates free pools of catenins and stabilizes APC-
catenin complexes. Mol Cell Biol 16:2128–2134.
28. Sugi Y and J Lough. (1994). Anterior endoderm is a spe-
cific effector of terminal cardiac myocyte differentiation of
cells from the embryonic heart-forming region. Dev Dyn
200:155–162.
29. Nascone M and M Mercola. (1995). An inductive role for
the endoderm in Xenopus cardiogenesis. Development
121:515–523.
30. Schultheiss TM, S Xydas and AB Lassar. (1995). Induc-
tion of avian cardiac myogenesis by anterior endoderm. De-
velopment 121:4203–4214.
31. Antin PB, TG Taylor and T Yatskievych. (1994). Precar-
diac mesoderm is specified during gastrulation in quail.
Dev Dyn 200:144–154.
PAL AND KHANNA
38
32. Behfar A, LV Zingman, DM Hodgson, JM Rauzier, GC
Kane, A Terzie and M Puccat. (2002). Stem cell differen-
tiation requires a paracrine pathway in the heart. FASEB J
16:1558–1566.
33. Ten Dijke P, MJ Goumans, F Itoh and S Itoh. (2002). Reg-
ulation of cell proliferation by Smad proteins. J Cell Phys-
iol 191:1–16.
34. Attisano L and JL Wrana. (2002). Signal transduction by
the TGF-beta superfamily. Science 296:1646–1647.
35. Hoppler S and RT Moon. (1998). BMP-2/-4 and Wnt-8 co-
operatively pattern the Xenopus mesoderm. Mech Dev
71:119–129.
36. Fischer L, G Boland and RS Tuan. (2002). Wnt-3A en-
hances bone morphogenetic protein-2-mediated chondro-
genesis of murine C3H10T1/2 mesenchymal cells. J Biol
Chem 277:30870–30878.
37. Kudoh T, SW Wilson and IB Dawid. (2002). Distinct roles
for Fgf, Wnt and retinoic acid in posteriorizing the neural
ectoderm. Development 129:4335–4346.
38. Garcia-Castro MI, C Marcelle and M Bronner-Fraser.
(2002). Ectodermal Wnt function as a neural crest inducer.
Science 297:848–851.
39. Ellies DL, V Church, P Francis-West and A Lumsden.
(2000). The WNT antagonist cSFRP2 modulates pro-
grammed cell death in the developing hindbrain. Develop-
ment 127:5285–5295.
40. Hussein SM, EK Duff and C Sirard. (2003). Smad4 and
beta-catenin co-activators functionally interact with lym-
phoid-enhancing factor to regulate graded expression of
Msx2. J Biol Chem 278:48805–48814.
41. Schneider M and M Mercola. (2001). Wnt antagonism
initiates cardiogenesis in Xenopus laevis. Genes Dev
15:304–315.
42. Marvin MJ, G Di Rocco, A Gardiner, SM Bush and AB
Lassar. (2001). Inhibition of Wnt activity induces heart for-
mation from posterior mesoderm. Genes Dev 15:316–327.
43. Tzahor E and AB Lassar. (2001). Wnt signals from the neural
tube block ectopic cardiogenesis. Genes Dev 15:255–260.
Address reprint requests to:
Dr. Aparna Khanna
Dhirubhai Ambani Life Sciences Center, South Block
R-282, TTC Industrial area of MIDC
Thane-Belapur Road, Rabale
Navi Mumbai-400 701
India
E-mail: aparna_khanna@relbio.com
Received September 5, 2005; accepted December 5,
2005.
Smad AND Wnt SIGNALING IN CARDIAC DIFFERENTIATION
39
This article has been cited by:
1. Lisa K. Martin , Nadejda V. Mezentseva , Momka Bratoeva , Ann F. Ramsdell , Carol A. Eisenberg , Leonard M. Eisenberg .
2011. Canonical WNT Signaling Enhances Stem Cell Expression in the Developing Heart Without a Corresponding Inhibition
of Cardiogenic Differentiation. Stem Cells and Development 20:11, 1973-1983. [Abstract] [Full Text] [PDF] [PDF Plus]
[Supplementary material]
2. Jin-Yong Yook , Min-Jeong Kim , Myung Jin Son , Seokyoung Lee , Yoonkey Nam , Yong-Mahn Han , Yee Sook Cho . 2011.
Combinatorial Activin Receptor-Like Kinase/Smad and Basic Fibroblast Growth Factor Signals Stimulate the Differentiation
of Human Embryonic Stem Cells into the Cardiac Lineage. Stem Cells and Development 20:9, 1479-1490. [Abstract] [Full
Text] [PDF] [PDF Plus] [Supplementary material]
3. Takatoshi Tsuchihashi, Jun Maeda, Chong H. Shin, Kathryn N. Ivey, Brian L. Black, Eric N. Olson, Hiroyuki Yamagishi,
Deepak Srivastava. 2011. Hand2 function in second heart field progenitors is essential for cardiogenesis. Developmental
Biology 351:1, 62-69. [CrossRef]
4. Santiago Roura, Jordi Farré, Leif Hove-Madsen, Cristina Prat-Vidal, Carolina Soler-Botija, Carolina Gálvez-Montón, Marta
Vilalta, Antoni Bayes-Genis. 2010. Exposure to cardiomyogenic stimuli fails to transdifferentiate human umbilical cord
blood-derived mesenchymal stem cells. Basic Research in Cardiology 105:3, 419-430. [CrossRef]
5. Min Young Lee, Hyun Woo Lim, Sang Hun Lee, Ho Jae Han. 2009. Smad, PI3K/Akt, and Wnt-Dependent Signaling Pathways
Are Involved in BMP-4-Induced ESC Self-Renewal. Stem Cells 27:8, 1858-1868. [CrossRef]
6. R. Buesen, E. Genschow, B. Slawik, A. Visan, H. Spielmann, A. Luch, A. Seiler. 2009. Embryonic Stem Cell Test Remastered:
Comparison between the Validated EST and the New Molecular FACS-EST for Assessing Developmental Toxicity In Vitro.
Toxicological Sciences 108:2, 389-400. [CrossRef]
7. L. Lin, L. Cui, W. Zhou, D. Dufort, X. Zhang, C.-L. Cai, L. Bu, L. Yang, J. Martin, R. Kemler, M. G. Rosenfeld, J. Chen, S.
M. Evans. 2007. beta-Catenin directly regulates Islet1 expression in cardiovascular progenitors and is required for multiple
aspects of cardiogenesis. Proceedings of the National Academy of Sciences 104:22, 9313-9318. [CrossRef]
8. Rajarshi Pal, Aparna Khanna. 2007. Similar pattern in cardiac differentiation of human embryonic stem cell lines, BG01V
and ReliCell # hES1, under low serum concentration supplemented with bone morphogenetic protein-2. Differentiation 75:2,
112-122. [CrossRef]
9. Rajarshi Pal, Geeta Ravindran. 2006. Assessment of pluripotency and multilineage differentiation potential of NTERA-2 cells
as a model for studying human embryonic stem cells. Cell Proliferation 39:6, 585-598. [CrossRef]
10. Deepak Srivastava, Kathryn N. Ivey. 2006. Potential of stem-cell-based therapies for heart disease. Nature 441:7097,
1097-1099. [CrossRef]
11. 2006. Correction. Stem Cells and Development 15:3, 471-471. [Citation] [PDF] [PDF Plus]
12. Denis English , Paul R. Sanberg . 2006. Neural Specification of Stem Cell Differentiation. Stem Cells and Development 15:2,
139-140. [Citation] [PDF] [PDF Plus]
... Given that cP1P promoted SMAD1/5/8 phosphorylation even after the S1P receptor had been blocked by VPC, we further assessed alternative receptors that are linked to the induction of SMAD1/5/8 signaling during cardiac differentiation. Bone morphogenetic protein receptor (BMPR)-mediated signaling has also been reported to activate the p-SMAD1/5/8 pathway during cardiogenesis [20]. ALK3/BMP receptor 1A-mediated signaling has been reported to be important during the development of the heart [22,23]. ...
... The mechanism by which cP1P contributes to cardiac differentiation remains unknown. We investigated an important signaling pathway regulated by SMAD proteins that has been reported to regulate cardiogenesis [20]. Activated SMAD proteins 1/5/8 relay signals from the cell membrane to the nucleus via the common mediator SMAD4, triggering the activation of cardiogenic transcription factors GATA4, NKX2.5, and MEF2C [20,[38][39][40]. ...
... We investigated an important signaling pathway regulated by SMAD proteins that has been reported to regulate cardiogenesis [20]. Activated SMAD proteins 1/5/8 relay signals from the cell membrane to the nucleus via the common mediator SMAD4, triggering the activation of cardiogenic transcription factors GATA4, NKX2.5, and MEF2C [20,[38][39][40]. In line with previous reports, we observed that the enhanced expressions of the CM markers NKX2.5, GATA4, and MEF2C could be explained by the upregulation of SMAD 1/5/8 phosphorylation ( Figure 2E). ...
Article
Full-text available
Adult human cardiomyocytes have an extremely limited proliferative capacity, which poses a great barrier to regenerative medicine and research. Human embryonic stem cells (hESCs) have been proposed as an alternative source to generate large numbers of clinical grade cardiomyocytes (CMs) that can have potential therapeutic applications to treat cardiac diseases. Previous studies have shown that bioactive lipids are involved in diverse cellular responses including cardiogenesis. In this study, we explored the novel function of the chemically synthesized bioactive lipid O-cyclic phytosphingosine-1-phosphate (cP1P) as an inducer of cardiac differentiation. Here, we identified cP1P as a novel factor that significantly enhances the differentiation potential of hESCs into cardiomyocytes. Treatment with cP1P augments the beating colony number and contracting area of CMs. Furthermore, we elucidated the molecular mechanism of cP1P regulating SMAD1/5/8 signaling via the ALK3/BMP receptor cascade during cardiac differentiation. Our result provides a new insight for cP1P usage to improve the quality of CM differentiation for regenerative therapies.
... LPCs (red box) that were specified in the background of BMP4 and (or) Wnt treatment (n = 3). *p < 0.05, **p < 0.01 vs control group; #p < 0.05, ##p < 0.01 vs BMP4 group; $p < 0.05, $$p < 0.01 vs CHIR-3 group The embryonic development of the heart is complex and intersecting, and is co-regulated by multiple signaling pathways (Cohen et al. 2008;Pal and Khanna 2006;van Wijk et al. 2007). The BMP and Wnt signaling pathways are relatively clear. ...
Article
Full-text available
The embryonic development of sinus nodes (SAN) is co-regulated by multiple signaling pathways. Among these, the bone morphogenetic protein (BMP) and Wnt signaling pathways are involved in the development of SAN. In this study, the effects of BMP and Wnt signaling on the differentiation of SAN-like pacemaker cells (SANLPCs) were investigated. Human induced pluripotent stem cells (hiPSCs) were divided into four groups: control, BMP4, CHIR—3, and BMP4 + CHIR (CHIR: a Wnt signaling activator). The samples were tested at day (D) 15 of differentiation. The final protocol for the activation of BMP signaling at D0–D3 and reactivation of Wnt signaling at D5–D7 in the differentiation of hiPSCs were determined. The results showed that the mRNA levels of pacemaker markers (TBX18, SHOX2, TBX3, HCN4, and HCN1) were higher in the BMP4 + CHIR group than in the control group, and working myocardial genes were downregulated. The immunofluorescence assay revealed that the expression of SHOX2 and HCN4 increased in the BMP4 + CHIR group compared to that in the other groups. In addition, the results of patch clamps revealed that a funny current of higher density and typical SAN action potentials were recorded, except in the control group, in which the L-type calcium current was higher in the BMP4 + CHIR group than in the other groups. Finally, the proportion of SANLPCs (cTnT⁺ NKX2.5⁻) was further enhanced by the combination of BMP4 and CHIR treatment. In summary, the combination of BMP and Wnt signaling promotes the differentiation of SANLPCs from hiPSCs.
... Specifically, as shown in Figure 1, GCs activate PI3K, and, subsequently, Akt-1 accelerates both p-GSK-3β and vascular endothelial growth factor (VEGF) expression [38]. Moreover, p-GSK-3β expression induced by Akt protein directly regulates downstream transcriptional factors, such as β-catenin and the zinc finger transcription factor, GATA-4 [42,43]. GATA-4 is related to cardiac myocyte hypertrophy [44,45]. ...
Article
Full-text available
Although the rate of preterm birth has increased in recent decades, a number of preterm infants have escaped death due to improvements in perinatal and neonatal care. Antenatal glucocorticoid (GC) therapy has significantly contributed to progression in lung maturation; however, its potential effects on other organs remain controversial. Furthermore, the effects of antenatal GC therapy on the fetal heart show both pros and cons. Translational research in animal models indicates that constant fetal exposure to antenatal GC administration is sufficient for lung maturation. We have established a premature fetal rat model to investigate immature cardiopulmonary functions in the lungs and heart, including the effects of antenatal GC administration. In this review, we explain the mechanisms of antenatal GC actions on the heart in the fetus compared to those in the neonate. Antenatal GCs may contribute to premature heart maturation by accelerating cardiomyocyte proliferation, angiogenesis, energy production, and sarcoplasmic reticulum function. Additionally, this review specifically focuses on fetal heart growth with antenatal GC administration in experimental animal models. Moreover, knowledge regarding antenatal GC administration in experimental animal models can be coupled with that from developmental biology, with the potential for the generation of functional cells and tissues that could be used for regenerative medical purposes in the future.
... BMP-7 plays a major role in upregulating the transcription factor osterix (Osx) or SP7 which has the ability to stimulate differentiation of osteoblasts both in vitro and in vivo [65,75,76]. These studies suggested the involvement of canonical cell signaling pathway in osteoblast differentiation and embryo skeletal formation induced by BMP-7 [76][77][78][79][80][81][82]. BMP-7 induced activation of Smad-1/5 leads to the activation of osterix resulting in increased osteogenic markers alkaline phosphatase (ALP) activity and mineralization [83]. ...
Article
Full-text available
Bone morphogenetic protein-7 is (BMP-7) is a potent anti-inflammatory growth factor belonging to the Transforming Growth Factor Beta (TGF-β) superfamily. It plays an important role in various biological processes, including embryogenesis, hematopoiesis, neurogenesis and skeletal morphogenesis. BMP-7 stimulates the target cells by binding to specific membrane-bound receptor BMPR 2 and transduces signals through mothers against decapentaplegic (Smads) and mitogen activated protein kinase (MAPK) pathways. To date, rhBMP-7 has been used clinically to induce the differentiation of mesenchymal stem cells bordering the bone fracture site into chondrocytes, osteoclasts, the formation of new bone via calcium deposition and to stimulate the repair of bone fracture. However, its use in cardiovascular diseases, such as atherosclerosis, myocardial infarction, and diabetic cardiomyopathy is currently being explored. More importantly, these cardiovascular diseases are associated with inflammation and infiltrated monocytes where BMP-7 has been demonstrated to be a key player in the differentiation of pro-inflammatory monocytes, or M1 macrophages, into anti-inflammatory M2 macrophages, which reduces developed cardiac dysfunction. Therefore, this review focuses on the molecular mechanisms of BMP-7 treatment in cardiovascular disease and its role as an anti-fibrotic, anti-apoptotic and anti-inflammatory growth factor, which emphasizes its potential therapeutic significance in heart diseases.
... Disturbances in TGF-β signaling in mice causes embryonic lethality and the development of VSDs, myocardial thinning, and double outlet right ventricle (DORV), a cardiac anomaly of which VSD is a feature [40]. Moreover, BMP-dependent activation of transcription factors including the cardiac zinc finger transcription factor GATA4, homeobox protein NKX2.5, and myocyte enhancer factor 2C (MEF2C) augment differentiation mediated by the SMAD signaling pathways that regulate cardiomyocyte proliferation [41]. ...
Article
Full-text available
Ventricular Septal Defect (VSD), the most common congenital heart defect, is characterized by a hole in the septum between the right and left ventricles. The pathogenesis of VSD is unknown in most clinical cases. There is a paucity of data relevant to epigenetic changes in VSD. The placenta is a fetal tissue crucial in cardiac development and a potentially useful surrogate for evaluating the development of heart tissue. To understand epigenetic mechanisms that may play a role in the development of VSD, genome-wide DNA methylation assay on placentas of 8 term subjects with isolated VSD and no known or suspected genetic syndromes and 10 unaffected controls was performed using the Illumina HumanMethylation450 BeadChip assay. We identified a total of 80 highly accurate potential CpGs in 80 genes for detection of VSD; area under the receiver operating characteristic curve (AUC ROC) 1.0 with significant 95% CI (FDR) p-values < 0.05 for each individual locus. The biological processes and functions for many of these differentially methylated genes are previously known to be associated with heart development or disease, including cardiac ventricle development (HEY2, ISL1), heart looping (SRF), cardiac muscle cell differentiation (ACTC1, HEY2), cardiac septum development (ISL1), heart morphogenesis (SRF, HEY2, ISL1, HEYL), Notch signaling pathway (HEY2, HEYL), cardiac chamber development (ISL1), and cardiac muscle tissue development (ACTC1, ISL1). In addition, we identified 8 microRNAs that have the potential to be biomarkers for the detection of VSD including: miR-191, miR-548F1, miR-148A, miR-423, miR-92B, miR-611, miR-2110, and miR-548H4. To our knowledge this is the first report in which placental analysis has been used for determining the pathogenesis of and predicting VSD.
... The GSK-3b inhibition by Akt protein is an important negative regulator of downstream transcriptional factors such as bcatenin and GATA-4. 36 GSK-3b is constitutively active under unstimulated conditions and induces cardiac development through Wnt signaling. The inhibition of GSK-3b activates Wnt/b-catenin signaling during the proliferation or differentiation of early beating cardiac myocytes and robustly enhances the proliferative capacity of early cardiomyocytes. ...
Article
Abstruct Background ATP synthesis and cardiac contraction‐related protein production are accelerated in the immature fetal heart by antenatal glucocorticoid (GC) administration. This study investigated the structural maturity of the myocardium and underlying signal pathway associated with cardiac growth in fetal rats that received antenatal GC. Methods and Results Dexamethasone (DEX) was administered to pregnant rats for 2 days from day 17 or day 19 of gestation and the hearts of 19‐ and 21‐day fetuses and 1‐day‐old neonates were analyzed. Although irregular myofibril orientation was observed morphologically in 19‐day fetal hearts, the myofibril components were organized in fetuses after DEX administration. The cross‐sectional area of the myocardium and Ki‐67‐positive cells were significantly increased in fetal DEX groups, suggesting that cardiac enlargement resulted from myocyte proliferation. Glycogen synthase kinase‐3β (GSK‐3β) protein levels were significantly decreased in fetal DEX groups. β‐catenin and vascular endothelial growth factor protein levels were also significantly increased. Furthermore, increased cardiomyocyte proliferation appeared to be mediated by GC receptors after culture with DEX in vitro. Conclusions These results suggest that antenatal DEX administration induces structural maturity accompanying cardiomyocyte proliferation in the premature fetal rat heart, and GSK‐3β, β‐catenin is thought to contribute to cardiac growth This article is protected by copyright. All rights reserved.
Preprint
Full-text available
The vertebrate head mesoderm provides the heart, the great vessels, smooth and most head skeletal muscle, and parts of the skull base. The ability to generate cardiac and smooth muscle is thought to be the evolutionary ground-state of the tissue, and initially the head mesoderm has cardiac competence throughout, even in the paraxial region that normally does not engage in cardiogenesis. How long this competence lasts, and what happens as cardiac competence fades, is not clear. Using a wide palette of marker genes in the chicken embryo, we show that the paraxial head mesoderm has the ability to respond to Bmp, a known cardiac inducer, for a long time. However, Bmp signals are interpreted differently at different time points. Bmp triggers cardiogenesis up to early head fold stages; the ability to upregulate smooth muscle markers is retained slightly longer. Notably, as cardiac competence fades, Bmp activates the head skeletal muscle programme instead. Summary statement The head mesoderm has generic cardiac competence until head fold stages. Thereafter, cardiac competence fades in the paraxial region, and Bmp activates head skeletal muscle programmes instead of cardiac programmes.
Preprint
Full-text available
Ventricular Septal Defect (VSD), the most common congenital heart defect, is characterized by a hole in the septum between the right and left ventricles. The pathogenesis of VSD is unknown in most clinical cases. There is a paucity of data relevant to epigenetic changes in VSD. The placenta is a fetal tissue and is a potentially useful surrogate for the evaluation of fetal organ development. To understand epigenetic mechanisms that may play a role in the development of VSD, a genome-wide DNA methylation assay of the placentas of 8 term subjects with isolated VSD and no known or suspected genetic syndromes and 10 normal controls was performed using the Illumina HumanMethylation450 BeadChip assay. The study identified a total of 80 highly accurate potential epigenomic markers in 80 genes for the detection of VSD; area under the receiver operating characteristic curve (AUC ROC) = 1.0 with significant 95% CI (FDR) p-values < 0.05. The biological processes and functions for these differentially methylated genes are known to be associated with heart development or heart disease, including cardiac ventricle development (HEY2, ISL1), heart looping (SRF), cardiac muscle cell differentiation (ACTC1, HEY2), cardiac septum development (ISL1), heart morphogenesis (SRF, HEY2, ISL1, HEYL), Notch signaling pathway (HEY2, HEYL), cardiac chamber development (ISL1), and cardiac muscle tissue development (ACTC1, ISL1). The study also identified eight microRNA genes that have the potential to be biomarkers for the early detection of VSD including miR-191, miR-548F1, miR-148A, miR-423, miR-92B, miR-611, miR-2110, and miR-548H4. To our knowledge this is the first report in which placental analysis has been used for determining the pathogenesis of and predicting CHD.
Article
During cardiac development, DNA binding transcription factors and epigenetic modifiers regulate gene expression in cardiac progenitor cells (CPCs). We have previously shown that YY1 is essential for the commitment of mesodermal precursors into CPCs. However, the role of YY1 in the maintenance of CPC phenotype and their differentiation into cardiomyocytes is unknown. In this study, we found, by genome-wide transcriptional profiling and phenotypic assays, that YY1 overexpression prevents cardiomyogenic differentiation and maintains the proliferative capacity of CPCs. We show further that the ability of YY1 to regulate CPC phenotype is associate with its ability to modulate histone modifications specifically at a developmentally critical enhancer of Nkx2-5 and other key cardiac transcription factor such as Tbx5. Specifically, YY1 overexpression helps to maintain markers of gene activation such as the acetylation of histone H3 at lysine 9 (H3K9Ac) and lysine 27 (H3K27Ac) as well as tri-methylation at lysine 4 (H3K4Me3) at the Nkx2-5 cardiac enhancer. Furthermore, transcription factors associated proteins such as PoIII, p300, and Brg1 are also enriched at the Nkx2-5 enhancer with YY1 overexpression. The biological activities of YY1 in CPCs appear to be cell autonomous, based co-culture assays in differentiating embryonic stem cells. Altogether, these results demonstrate that YY1 overexpression is sufficient to maintain a CPC phenotype through its ability to sustain the presence of activating epigenetic/chromatin marks at key cardiac enhancers. This article is protected by copyright. All rights reserved.
Article
Full-text available
Embryonic patterning in vertebrates is dependent upon the balance of inductive signals and their specific antagonists. We show that Noggin, which encodes a bone morphogenetic protein (BMP) antagonist expressed in the node, notochord, and dorsal somite, is required for normal mouse development. Although Noggin has been implicated in neural induction, examination of null mutants in the mouse indicates that Noggin is not essential for this process. However, Noggin is required for subsequent growth and patterning of the neural tube. Early BMP-dependent dorsal cell fates, the roof plate and neural crest, form in the absence of Noggin. However, there is a progressive loss of early, Sonic hedgehog (Shh)-dependent ventral cell fates despite the normal expression of Shh in the notochord. Further, somite differentiation is deficient in both muscle and sclerotomal precursors. Addition of BMP2 or BMP4 to paraxial mesoderm explants blocks Shh-mediated induction of Pax-1, a sclerotomal marker, whereas addition of Noggin is sufficient to induce Pax-1. Noggin and Shh induce Pax-1 synergistically. Use of protein kinase A stimulators blocks Shh-mediated induction of Pax-1, but not induction by Noggin, suggesting that induction is mediated by different pathways. Together these data demonstrate that inhibition of BMP signaling by axially secreted Noggin is an important requirement for normal patterning of the vertebrate neural tube and somite.
Article
Full-text available
WNT signaling has been shown to influence the development of the heart. Although recent data suggested that canonical WNTs promote the emergence and expansion of cardiac progenitors in the pregastrula embryo, it has long been accepted that once gastrulation begins, canonical WNT signaling needs to be suppressed for cardiac development to proceed. Yet, this latter supposition appears to be odds with the expression of multiple canonical WNTs in the developing heart. The present study examining the effect of ectopic canonical WNT signaling on cardiogenesis in the developing frog was designed to test the hypothesis that heart formation is dependent on the inhibition of canonical WNT activity at the onset of gastrulation. Here we report that cardiac differentiation of explanted precardiac tissue from the dorsal marginal zone was not suppressed by exposure to WNT1 protein, although expression of Tbx5, Tbx20, and Nkx2.5 was selectively reduced. Pharmacological activation of WNT signaling in intact embryos using the GSK3 inhibitor SB415286 did not prevent the formation of an anatomically normal and functionally sound heart, with the only defect observed being lower levels of the cardiac transcription factor Nkx2.5. In both the explant and whole embryo studies, expression of muscle genes and proteins was unaffected by ectopic canonical WNT signaling. In contrast, canonical Wnt signaling upregulated expression of the cardiac stem cell marker c-kit and pluripotency genes Oct25 and Oct60. However, this regulatory stimulation of stem cells did not come at the expense of blocking cardiac progenitors from differentiating.
Article
The Wnt signaling pathway, named for its most upstream ligands, the Wnts, is involved in various differentiation events during embryonic development and leads to tumor formation when aberrantly activated. Molecular studies have pinpointed activating mutations of the Wnt signaling pathway as the cause of approximately 90% of colorectal cancer (CRC), and somewhat less frequently in cancers at other sites, such as hepatocellular carcinoma (HCC). Ironically, Wnts themselves are only rarely involved in the activation of the pathway during carcinogenesis. Mutations mimicking Wnt stimulation-generally inactivating APC mutations or activating beta-catenin mutations-result in nuclear accumulation of beta-catenin which subsequently complexes with T-cell factor/lymphoid enhancing factor (TCF/LEF) transcription factors to activate gene transcription. Recent data identifying target genes has revealed a genetic program regulated by beta-catenin/TCF controlling the transcription of a suite of genes promoting cellular proliferation and repressing differentiation during embryogenesis, carcinogenesis, and in the post-embryonic regulation of cell positioning in the intestinal crypts. This review considers the spectra of tumors arising from active Wnt signaling and attempts to place perspective on recent data that begin to elucidate the mechanisms prompting uncontrolled cell growth following induction of Wnt signaling.
Article
Wnt signalling controls many different cell fate choices in a wide variety of animal species. Recent studies have revealed that regulatory interactions at several steps in the pathway can modify its outcome, helping to explain how the same pathway can, in different contexts, have very different characteristics and consequences.
Article
We previously reported that combined treatment with bone morphogenetic protein-2 (BMP-2) and fibroblast growth factor-4 (FGF-4) induces cardiogenic events culminating in full cardiac differentiation of non-precardiac mesoderm explanted from stage 6 avian embryos (Lough et al. [1996] Dev. Biol. 178:198–202.). To elucidate the respective functions of BMP and FGF in initiating and maintaining the cardiogenic process, we have used these ectopic cells as a cardiac specification model to ascertain requirements for growth factor specificity and extent of application, as well as induction of cardiac transcription factors. The inability of some BMP isoforms to replace the inductive activity of BMPs-2/4 indicated a specific requirement for this signaling pathway; moreover, neither activin-A nor insulin, which support terminal differentiation of precardiac mesoderm, nor leukocyte inhibitory factor (LIF), which promotes hypertrophy in cardiac myocytes, could replace BMP's cardiogenic activity. A similarly specific requirement for FGF-2/4 signaling was revealed since neither FGF-7, activin-A nor insulin could replace this activity. The effect of both factors was concentration-dependent; maximal incidence of explant differentiation for each occurred at 50 ng/ml. Surprisingly, the majority of explants treated with high BMP levels (250 ng/ml) exhibited a non-cardiac phenotype that was characterized by intense expression of alkaline phosphatase, suggesting differentiation toward an alternative mesodermal phenotype. Experiments to assess the duration of exposure to each factor that was required revealed that while exposure to BMP and FGF during only the initial 30 min of a 48-hr culture period was sufficient to induce cardiogenesis in a significant percentage of explants, 100% incidence of explant differentiation was obtained only when FGF treatment was restricted to the first 30 min and BMP was continuously present during the 48-hr culture period. Treatment with both growth factors was required to induce the cardiac transcription factors cNkx-2.5 and SRF; neither mRNA was induced by BMP or FGF alone. These findings indicate that: (1) specific members of the BMP and FGF families are required to induce cardiogenesis in non-precardiac mesoderm; (2) BMPs-2/4 may function as a morphogen; (3) brief application of both factors can induce cardiogenesis in a modest number of explants whereas (4) 100% incidence of explant differentiation can only be attained by brief FGF treatment combined with continuous BMP treatment and (5) both factors are necessary to induce downstream cardiac transcription factors. These findings are interpreted in terms of these factors' possible roles during cardiac specification and differentiation. Dev Dyn;218:383–393. © 2000 Wiley-Liss, Inc.
Article
Establishment of the dorsoventral axis is central to animal embryonic organization. In Xenopus two different classes of signaling molecules function in the dorsoventral patterning of the mesoderm. Both the TGF-beta-related products of the BMP-2 and BMP-4 genes and the Wnt molecule encoded by Xenopus Wnt-8 specify ventral fate and appear to inhibit dorsal mesodermal development. The similar functions of these molecularly very different classes of signaling molecules prompted us to study their mutual regulation and to closely compare their roles in mesoderm patterning. We find that Wnt-8 and BMP-4 are indistinguishable in their abilities to induce expression of ventral genes. Although BMP-2/-4 signaling regulates Wnt-8 expression, these genes do not function in a linear pathway because Wnt-8 overexpression cannot compensate for an inhibition of BMP-2/-4 function, but rather BMP-4 overexpression rescues ventral gene expression in embryos with inhibited Wnt-8 function. We further find that Wnt-8 and BMP-2/-4 differ in their abilities to regulate dorsal gene expression. While BMP-4 appears to generally inhibit the expression of dorsal genes, XenopusWnt-8 only inhibits the expression of the notochord marker Xnot. Whereas the inhibitory effect of BMP-2/-4 localizes dorsal mesodermal fate, our results suggest that Xenopus Wnt-8 functions in the further patterning of the dorsal mesoderm into the most dorsal sector from which the notochord develops and the dorsolateral sector from where the somites differentiate.
Article
Cardiogenesis involves the contributions of multiple progenitor pools, including mesoderm-derived cardiac progenitors known as the first and second heart fields. Disruption of genetic pathways regulating individual subsets of cardiac progenitors likely underlies many forms of human cardiac malformations. Hand2 is a member of the basic helix loop helix (bHLH) family of transcription factors and is expressed in numerous cell lineages that contribute to the developing heart. However, the early embryonic lethality of Hand2-null mice has precluded lineage-specific study of its function in myocardial progenitors. Here, we generated and used a floxed allele of Hand2 to ablate its expression in specific cardiac cell populations at defined developmental points. We found that Hand2 expression within the mesoderm-derived second heart field progenitors was required for their survival and deletion in this domain recapitulated the complete Hand2-null phenotype. Loss of Hand2 at later stages of development and in restricted domains of the second heart field revealed a spectrum of cardiac anomalies resembling forms of human congenital heart disease. Molecular analyses of Hand2 mutant cells revealed several genes by which Hand2 may influence expansion of the cardiac progenitors. These findings demonstrate that Hand2 is essential for survival of second heart field progenitors and that the graded loss of Hand2 function in this cardiac progenitor pool can cause a spectrum of congenital heart malformation.