You are on page 1of 11

This article is the introduction of a new thematic series on Wnts in Cardiovascular Development and Disease, which

includes the following articles:


An Updated Overview on Wnt Signaling Pathways: A Prelude for More [Circ Res. 2010;106:1798 –1806]
The Multiple Phases and Faces of Wnt Signaling During Cardiac Differentiation and Development [Circ Res.
2010;107:186 –199]
The Role of Wnt Signaling in Physiological and Pathological Angiogenesis

Wnt Signaling in Cardiac Hypertrophy and Remodeling: Lessons Learned From Cardiac Development

Wnt Signaling and Aging-Related Heart Disorders


Wnt Signaling and Stem Cells Michael Kühl, Guest Editor

WNT Signaling in Adult Cardiac Hypertrophy and Remodeling


Lessons Learned From Cardiac Development
Martin W. Bergmann
Abstract: On pathological stress, the heart reactivates several signaling pathways that traditionally were thought
to be operational only in the developing heart. One of these pathways is the WNT signaling pathway. WNT
controls heart development but is also modulated during adult heart remodeling. This review summarizes the
currently available data regarding WNT signaling during left ventricular (LV) remodeling. Upstream, soluble
frizzled-related proteins (sFRPs) block WNT-dependent activation of the canonical WNT pathway. By inhibition
of WNT activation, these factors also reduce ␤-catenin– dependent transcription by altering the ratio of
cytoplasmic/nuclear ␤-catenin. In experimental settings, sFRPs injected into the heart attenuated LV remodeling.
Downloaded from http://ahajournals.org by on June 13, 2021

sFRPs are secreted from autologous bone marrow– derived mononuclear cells. Disheveled is a signaling
intermediate of both the canonical and noncanonical WNT pathway. Similarly to the effect of sFRP, depletion of
a disheveled isoform attenuated LV remodeling. In contrast, disheveled activation led to progressive dilated
cardiomyopathy. Inhibition of nuclear ␤-catenin signaling downstream of the canonical WNT pathway
significantly reduced postinfarct mortality and functional decline of LV function following chronic left anterior
descending coronary artery ligation. WNT signaling also affects mobilization and homing of bone marrow–
derived vasculogenic progenitor cells. Finally, heart-specific WNT/␤-catenin interaction partners have been
identified that will possibly allow targeting this pathway in a tissue-specific manner. In summary, the WNT
pathway plays a pivotal role in adult cardiac remodeling and may be suitable for therapeutic interventions.
Currently, several molecular and cellular mechanisms whereby WNT inhibition attenuates LV remodeling are
proposed. Reactivation of the developmental program to restore functional LV myocardium from resident
precursor cells may significantly contribute to this process. (Circ Res. 2010;107:1198-1208.)
Key Words: heart failure 䡲 progenitor cells 䡲 angiogenesis 䡲 WNT 䡲 ␤-catenin

O n pathological stress, the heart reactivates several sig-


naling pathways that traditionally were thought to be
operational only in the developing heart.1 Recently published
progenitor cells (CPCs) appear similar to prespecified cardiac
stem cells in the embryo.3 Canonical WNT signaling controls
the proliferation and differentiation of embryonic cardiac
data prove the adult heart to be capable of limited regenera- precursors toward mature cardiomyocytes.4,5 Both positive
tion from cardiac-endogenous precursors.2 Such adult cardiac and negative WNT signaling is observed during cardiac

Original received July 10, 2010; revision received September 14, 2010; accepted September 21, 2010. In September 2010, the average time from
submission to first decision for all original research papers submitted to Circulation Research was 13.1 days.
From the Experimental and Clinical Research Center, Charité Campus Buch & Max Delbrück Center for Molecular Medicine, Berlin, Germany. Present
address: Department of Cardiology, ASKLEPIOS Klinik St Georg, Hamburg, Germany.
Correspondence to Priv-Doz Dr med Martin W. Bergmann, Department of Cardiology, ASKLEPIOS Klinik St Georg, Lohmühlenstr. 5, 20099
Hamburg, Germany. E-mail docbergmann@mac.com
© 2010 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.110.223768

1198
Bergmann WNT and Cardiac Remodeling 1199

remodeling of the adult heart on stress or injury: WNT


Non-standard Abbreviations and Acronyms
factors, as well as frizzled-related proteins inhibiting WNTs,
are detected.6 – 8 This review summarizes the available data Ang II angiotensin II
regarding WNT signaling in the adult heart. Because the role BMP bone morphogenic protein
of WNT in cardiac development may allow understanding of CPC cardiac progenitor cell
the mechanisms also relevant in adult cardiac remodeling, the CT control (mice)
review begins with a brief summary of the existing knowl- cTnT cardiac troponin T
edge regarding WNT signaling in cardiac precursor cells. DKK dickkopf
E embryonic day
WNT/␤-Catenin Signaling Axis in Embryonic Frz frizzled (WNT receptor)
and Adult Cardiac Precursor Cells IGF insulin-like growth factor
The heart develops from a common precursor. One of the IGFBP insulin-like growth factor– binding protein
earliest cardiac-specific transcription factors expressed both Isl-1 islet-1
in cultured embryonic stem cells, as well as in situ, is the KLF Krüppel-like factor
basic helix–loop– helix factor MesP1 (Figure 1).9 It is appear- LAD left anterior descending coronary artery
ing at approximately embryonic day (E)5.5 in the murine LV left ventricular
cardiac development and vanishes by E8.5.10 In vitro, MesP1 MHC myosin heavy chain
is required and sufficient to induce cardiovasculogenesis
MSC mesenchymal stem cell
from embryonic stem cells. The key factor downstream of
NLK nemo-like kinase
MesP1 is the inhibitor of the WNT/␤-catenin canonical
PKC protein kinase C
signaling cascade, dickkopf-1. Whether dickkopf-1 is the sole
PLC phospholipase C
transcriptional target of MesP1 responsible for cardiac cell
sFRP soluble frizzled related protein
fate determination remains a matter of debate: following
TG transgenic (mice)
MesP1 activation, the whole panel of cardiac transcription
machinery is found to be upregulated.9 –11 More precisely,
forced expression of MesP1 in ES cells leads to the appear-
ance of cardiomyocytes with intact gap junctions, direct
expression of Nkx2.5, MEF2c, and GATA4, as well as early differentiation of several independent cardiac precursor
sprouting of vasculogenic cells positive for von Willebrand cell pools from a common cardiac stem cell marked by
Downloaded from http://ahajournals.org by on June 13, 2021

factor (vWF) and CD31.11 In summary, inhibition of the MesP1 expression (Figure 1). These cells have the capacity to
canonical WNT- pathway via dickkopf appears to mediate restore functional myocardium in the adult heart following

Figure 1. Schematic summary of WNT/␤-catenin signaling regarding developmental and adult CPC proliferation and differentia-
tion. CPCs expressing markers of first and second heart field progenitors were identified in both the murine embryo and adult heart. To
date, no MesP1pos multipotent CPCs have been described in the adult heart. WNT/␤-catenin activation is required for amplification of
CPCs in both the embryo and the adult heart. WNT/␤-catenin inhibition, together with activation of other signaling pathways (ie, BMP),
not depicted in the scheme controls cell fate determination (ie, first vs second heart field or vasculogenic) and differentiation toward
contractile LV cardiomyocytes.
1200 Circulation Research November 12, 2010

damage: a population of CPCs from rhesus monkey pluripo- second heart field progenitors. Negative regulation of
tent embryonic stem cells was able to integrate into an infarct ␤-catenin appears to drive CPC cell fate and differentiation.22
scar without forming teratomas. This maneuver replaced Similarly, the nuclear protein Chibby was found to facili-
⬇20% of the scar tissue in rhesus monkey heart with tate cardiomyocyte differentiation from ES cells. Chibby
contractile myocardial tissue.12 directly binds to ␤-catenin and antagonizes its transcriptional
Similarly to the role of dickkopf downstream of MesP1, the activity (Figure 3). Chibby expression was associated with
insulin-like growth factor binding protein (IGFBP)-4 was upregulation of Nkx2.5, ␤-myosin heavy chain (␤MHC), and
found to induce cardiomyocyte differentiation from P19CL6 Mef2c. Chibby expression is also found at high levels in adult
cells. Independent of IGF, IGFBP-4 leads to the expression of cardiomyocytes. Apparently, the cardiac-specific transcrip-
cardiac troponin T and other markers of cardiomyocyte tion factor Nkx2.5 controls Chibby expression. The authors
differentiation in P19CL6 cells. IGFBP-4 attenuated activa- suggested the clinical perspective that antagonizing the
tion of a ␤-catenin– dependent reporter gene on stimulation WNT/␤-catenin pathway by this cardiac signaling molecule
with Frizzled 8. Several independent experiments then proved will allow to promote CPC differentiation toward a cardio-
IGFBP-4 to inhibit WNT signaling directly at the cell myocyte phenotype similar to cardiac development.23
membrane receptor complex by binding to Frz8 and LRP6. Several other independent lines of evidence find positive
Other pathways known to be involved in cardiomyocyte regulation of WNT/␤-catenin to be required for CPC ampli-
differentiation, namely the bone morphogenic protein (BMP) fication, whereas negative regulation of WNT/␤-catenin is
signaling cascade, were not affected. IGFBP-4 was not only required for CPC differentiation toward adult cardiomyocytes
sufficient but also required for cardiomyocyte differentiation (Figure 1). In the murine embryo development, upregulation
in this system: knockdown of IGFBP-4 abrogated cardiomyo- of WNT/␤-catenin is required at the time of MesP1 activation
cyte differentiation.13 Interestingly, we identified IGFBP-5 to (approximately E5.5) to start amplification of the different
be a target for ␤-catenin– dependent transcription: gene array CPC pools. Most prominently, this was observed for first and
experiments with adult left ventricular (LV) tissue from mice second heart field CPCs marked by expression of Tbx5 and
with heart-specific, conditional ␤-catenin stabilization (␤- Isl1, respectively.24 At later stages (from approximately E7.5)
catenin␣MHC-⌬exon3) revealed IGFBP-5 to be a transcriptional
negative regulation of the WNT/␤-catenin pathway is re-
target of ␤-catenin.14 The role of this observation is currently
quired to drive differentiation of the 2 CPC populations to
unclear because IGFBP-1, -2, -4, and -6 but not IGFBP-5 and
adult cardiomyocytes.18,25 Other proteins regulated at this
-3 were found to inhibit WNT-dependent signaling. Possibly
time point of cardiac development include NKx2.5, the first
the IGF pathway itself is involved; forced, cardiac-specific
peak of ␣MHC, and GATA4 and eHand (first heart field) as
Downloaded from http://ahajournals.org by on June 13, 2021

expression of mIGF was found to attenuate LV remodeling


well as dHand (second heart field).26,27 It is often overlooked
following cryoinjury. This was not mediated by the AKT
that ␣MHC is long known to have 2 expression peaks: the
pathway but via the alternate pathway of phosphoinositide-
first peak is observed around E7.5 in the developing left
dependent protein kinase (PDK1) and serum glucocorticoid
ventricle.28,29 Subsequently, ␣MHC is downregulated and is
kinase (SGK)1. The authors suggested endogenous regener-
replaced by ␤MHC. After birth, ␣MHC is upregulated again
ation to be involved in this process.15
In cardiac development, WNT/␤-catenin has a stage- and is replacing ␤MHC.30
specific, at least biphasic, role.4,5 This state of affairs is We have developed a technique that allows for flow
covered in more detail in another review of this series.16 cytometric analysis of adult heart CPCs in the noncardiomyo-
Experiments with embryonic stem cell cultures revealed cyte cell fraction of either murine heart tissue or human left
WNT/␤-catenin inhibition plus activation of the BMP path- atrial appendage.31 In brief, heart tissue is mechanically
way to enhance differentiation toward a cardiomyocyte phe- minced and then subjected to 2 steps of size filtration where
notype but block hematopoietic and vascular lineage devel- the smallest mesh will filter for cells larger than 30 ␮m. This
opment in the early stage (Figure 1).17 Similar observations technique is almost completely filtering adult cardiomyo-
were made in vivo, where at approximately E5.5, upregula- cytes. This allows analyzing the noncardiomyocyte cell
tion of WNT/␤-catenin is required to amplify the cardiom- fraction of the adult heart without alteration of protein
yogenic precursor cell pool of both the first and second heart expression associated with the use of collagenase.16 Analysis
fields, giving rise to the left and right ventricle, respectively for several markers known from the developing heart to be
(Figure 1).18 The segregation of these 2 distinct cardiogenic characteristic for CPCs revealed the results depicted in Figure
lineages appears early and results in very distinct populations 2. In the adult heart, to date, no MesP1pos CPCs have been
of CPCs possibly also relevant to adult heart CPCs (Figure described. Similarly, only a few CPCs of the second heart
2).19 The second heart field is marked by the expression of field marked by islet-1 expression have been detected. How-
Islet (Isl)1.20 Using conditional mice, where Cre expression is ever, we readily detected CPCs positive for several markers
driven by the Isl1 promoter, Notch1 was found to negatively of the first heart field (Tbx5, ␣MHC, eHand) in adult murine
regulate ␤-catenin– dependent transcription.21 Inhibition of left ventricle, as well as human left atrial appendage (Figure
␤-catenin resulted in upregulation of cardiac transcription 2). Modulation of proliferation and differentiation of first
factors Myocd and Smyd1. In turn, these 2 factors were heart field CPCs in the adult heart may allow boosting
required to drive differentiation of Isl1 CPCs to a cardiomyo- endogenous regeneration. The precise knowledge of these
cyte phenotype (Figure 3). In summary, ␤-catenin upregula- developmental pathways has implications for adult cardiac
tion causes expansion of different CPC lines including Isl1pos remodeling as discussed below.
Bergmann WNT and Cardiac Remodeling 1201
Downloaded from http://ahajournals.org by on June 13, 2021

Figure 2. Qualitative and quantitative analysis of adult heart cardiac progenitor cells. Murine adult heart tissue or human adult left
atrial appendage tissue was homogenized and size-filtrated for depletion of adult cardiac myocytes as described.31 The flow-through
containing the noncardiomyocyte cell fraction of heart tissue was analyzed by flow cytometry and immunofluorescence for expression
of cardiac progenitor cell markers. A, Quantitative assessment of CPC marker expression in the noncardiomyocyte cell fraction from
murine adult heart as analyzed by flow cytometry. Tbx5, ␣MHC, and eHand are specific for the developmental first heart field, giving
rise to the left ventricle. Islet-1 and dHand are specific for the second heart field, giving rise to the right ventricle and LV outflow tract.
cTnT is specific for mature adult cardiomyocytes. Nkx2.5 is expressed in both first heart field (FHF) and second heart field (SHF) pro-
genitor cells. Sca-1 is an unspecific marker of mesenchymal stem cells. Data summarize analysis of nⱖ6 animals. B, Qualitative analy-
sis of murine adult heart CPCs. FACS analysis finds ⬇14% of ␣MHCpos nonadult cardiomyocyte cells to have proliferative capacity as
detected by coexpression of Ki67. The cytoplasm/nuclear ratio is close to 1; also, immunofluorescence detects cells with costaining of
Ki67 as a marker of cell proliferation and ␣MHC as a marker of cardiomyogenic cell commitment. This confirms the presence of first
heart field CPCs in the adult heart. C, Qualitative analysis of human adult heart CPCs isolated from left atrial appendage (LAA). ␣MHCpos/
Tbx5pos/eHandpos CPCs with a cytoplasm/nuclear ratio of close to 1 and evidence for cell division by depicting an S-phase CPC can
be found. This suggests the presence of first heart field CPCs to be present also in human adult heart.

Soluble Frizzled-Related Proteins Attenuate upregulated following experimental infarct predominantly in the
Adult Cardiac Remodeling Following infarct border zone together with frizzled receptors 1, 2, 5, and
10, as well as WNT 10b. These data suggest sFRPs to be
Experimental Infarct
involved in infarct healing and tissue homeostasis following
WNT proteins are secreted glycoproteins that act locally in a
injury.7
paracrine fashion. WNT proteins bind to frizzled receptors.
FrzA/sFRP1 was proven to regulate vascular cell prolifer-
These are 7-pass transmembrane proteins characterized by an
ation and to induce an angiogenic response.34 Moreover, a
extracellular N-terminal domain.32 sFRPs lack the transmem- genome-wide screen revealed sFRP2 to be the key stem cell
brane domain and compete locally for WNT binding (Figure paracrine factor that mediates myocardial survival and repair
3). Interestingly, sFRP-3 and sFRP-4 levels were found to be after experimental myocardial infarct treated with intracardial
elevated in hearts of patients with both dilated cardiomyop- injection of AKT-modified mesenchymal stem cells. This
athy and coronary heart disease.33 FrzA, related to mouse paracrine secretion of sFRP2 lead to a dramatic reduction of
sFRP1, has been detected during cardiovascular maturation infarct size and restoration of cardiac function. These effects
and in the adult heart. More specifically, sFRP1 is transiently were observed as early as 72 hours after AKT–mesenchymal
1202 Circulation Research November 12, 2010

Figure 3. Schematic summary of sig-


naling molecules described to inhibit
WNT/␤-catenin signaling in the car-
diac compartment. Inhibition of WNT
and several downstream signaling
components including ␤-catenin– de-
pendent transcription was found to
attenuate LV remodeling and improve
adult cardiac function following ische-
mia or pressure overload. This scheme
therefore summarizes both positive
and negative signaling components of
the WNT pathway in the cardiovascular
system. These signaling intermediates
may be suitable for therapeutic inter-
ventions. For example, sFRPs were
found to be released from bone mar-
row– derived mononuclear cells homing
to the heart after injury. Experimentally
injected or genetically upregulated
sFRPs generate similar responses.
sFRPs scavenge paracrine-secreted
WNTs, preventing them from binding to
membrane-bound Frz receptors. sFRPs
therefore inhibit both the canonical, as
well as the noncanonical, WNT path-
way. Disheveled binds to FrzR and is
required for activation of the nonca-
nonical and probably also the canoni-
cal pathway (see text). Dickkopf blocks
canonical WNT signaling upstream of GSK3␤. KLF15, Notch1, and Chibby are nuclear interaction partners of ␤-catenin blocking
␤-catenin/Tcf3-activated gene transcription. (Illustration Credit: Cosmocyte/Ben Smith).

stem cell implantation, clearly demonstrating the paracrine compartments like the bone marrow important for mobilization
secretion of sFRP2 to be the crucial mechanism whereby of endothelial progenitor cells that indirectly could influence LV
AKT-modified mesenchymal stem cells attenuate maladap- remodeling following experimental infarct.37
Downloaded from http://ahajournals.org by on June 13, 2021

tive LV remodeling following myocardial infarct.35 Transgenic mice with cardiac-specific overexpression of FrzA
Similar to the observation described above concerning genet- (similar to mouse sFRP1) resembled the phenotype described
ically modified mesenchymal stem cells, an endogenous repair above: ␣MHC-dependant FrzA expression increased survival by
mechanism was identified by comparing 2 different mice strains preventing myocardial rupture following experimental infarct.
inbred for the capability of accelerated healing following tissue Mechanistically, this phenotype was associated with reduced
injury (“superhealer”). These mice are capable of increased infarct size, less apoptosis, increased collagen deposition, and
myocardial regeneration following cryoinjury in association reduced activity of metalloproteinase-9 activity. This latter point
with a 3-fold increase of bone marrow– derived mononuclear is in line with the finding in global sFRP2 knockout mice,
cells found in the heart. Comparing bone marrow– derived although the functional phenotype is opposite.37 Interestingly,
CD45neg, CD11bneg, Sca-1pos, CD44pos mesenchymal stem cells the percentage of muscularized vessels was significantly higher
from the “superhealer” strain to a comparable wild type strain in mice with ␣MHC-dependent FrzA/sFRP1 overexpression,
identified sFRP2 to be the key paracrine factor secreted by together with an increase in open vessel area.7 In addition,
superhealer-derived mesenchymal stem cells. Wild-type mesen- FrzA/sFRP1 is sufficient and required for the effect of precon-
chymal stem cells transduced with sFRP2 and injected into the ditioning on infarct area: in the absence of FrzA/sFRP1, no
periinfarct region recapitulated the superhealer phenotype fol- effect of preconditioning is observed, which can be restored by
lowing experimental myocardial infarct.8 In conclusion, several the conditional, ␣MHC-dependent FrzA expression.38 Similarly,
lines of evidence suggest sFRP2 to be required and sufficient to direct injection of sFRP2-secreting mesenchymal stem cells in to
mediate myocardial repair following ischemic injury. In line the border zone of an experimental infarct decreased infarct area,
with the general function of sFRPs known to sequester WNTs increased ejection fraction and reduced dilation of the left
away from the active receptor complex, sFRP2 was shown to ventricle in association with increased vascular density. No
significantly inhibit ␤-catenin transcriptional activity as assessed increase in angiogenic-positive (PECAM-1) or cardiomyocyte
by reporter gene activation and target gene expression.36 marker–positive (anti-␣-actinin) cells was observed, implicating
Global knockout of sFRP2 lead to reduced cardiac fibrosis a paracrine mechanism rather than any form of direct transdif-
and improved function after experimental myocardial infarction. ferentiation of mesenchymal stem cells.8 Direct treatment of
The proposed mechanism was increased activation of metallo- isolated adult rat ventricular cardiomyocytes with sFRP2 re-
proteinases that are inhibited in the presence of sFRP2. It duced caspase activity and exerted a cytoprotective effect via
remains unclear in this study whether knockout of sFRP2 upregulation of Birc1b.35
affected WNT pathway activation. In addition, a global knock- In summary, the majority of data finds expression of sFRPs
out of sFRP2 might affect WNT signaling in other tissue in the heart following experimental infarct to be beneficial for
Bergmann WNT and Cardiac Remodeling 1203

available data regarding the WNT signaling intermediate


protein disheveled and ␤-catenin in LV remodeling are
summarized below.

Disheveled Is Required and Sufficient to


Induce Maladaptive Adult Heart Remodeling
Disheveled is an intracellular protein that is an essential
positive signal intermediate of both the WNT-initiated canon-
ical but also noncanonical signal transduction cascade.45
These branches are interconnected as described in detail in
another part of this review series.46 Some data support the
notion that the canonical/noncanonical branches separate at
the level of disheveled where activation of disheveled is
specific for the noncanonical pathway.32 Mutants in drosoph-
ila however indicate disheveled to be part of the membrane-
Figure 4. Schematic summary of cellular mechanisms con- bound protein complex of frizzled receptor regarding all
tributing to the observed attenuation of LV remodeling on downstream signaling pathways (Figure 3). Additional stud-
inhibition of WNT signaling at different levels. Several mecha-
nisms have been proposed whereby WNT signaling inhibition
ies may clarify this point in mammalian cells.
improves cardiac function in the adult following damage. Inhibi- More precisely, downstream of the frizzled receptor 3
tion of cardiac hypertrophy is possibly related to the Ca2⫹- branches are activated: the canonical WNT/␤-catenin axis
dependent pathways as part of noncanonical WNT signaling. and the 2 noncanonical pathways Rho/Rac/JNK and WNT/
WNT signaling may influence cardiomyocyte apoptosis and sur-
vival via PKB/AKT and G␣q. Mobilization of vasculogenic pro- Ca2⫹ (Figure 3). In the noncanonical Jun-kinase pathway,
genitors via canonical ␤-catenin may enhance angiogenesis. disheveled activates Rho/Rac via Daam1. This pathway has
Increased differentiation of cardiac-endogenous first heart field been linked to cytoskeletal organization and cell polarity.
CPCs toward functional adult cardiomyocytes may contribute to
improved contraction. In addition, WNT signaling affects secre-
Disheveled also links the intracellular domain of the Frizzled
tion of metalloproteinases, thereby altering cardiac fibrosis and receptor to activation of phospholipase C (PLC). Downstream
extracellular matrix composition. of PLC and the subsequent intracellular calcium release,
Calcium/calmodulin kinase II is activated linking Ca-
cardiac remodeling (Figure 4).7,8,34 –36,38 – 40 The mechanism is dependent signaling to frizzled/disheveled stimulation.
Downloaded from http://ahajournals.org by on June 13, 2021

not entirely clear, and the published literature suggests a CaMKII is required and sufficient to drive the hypertrophic
cardiomyocyte cytoprotective effect, as well as increased response in adult LV remodeling on injury and stress.47
vascular density in the infarct area (Figure 4). However, the Disheveled expression was shown to be upregulated on
general role of WNTs in other tissue compartments always transaortic banding (rat) and atrial-fibrillation induced heart
found activation rather than inhibition of the WNT pathway failure (porcine).6 Cardiac-specific overexpression of dishev-
to mediate antiapoptotic effects via activation of PKB/AKT.41 eled under the control of an ␣-MHC promoter results in
In addition, WNT activation is crucial for angiogenesis in severe cardiomyopathy at 3 months of age with significantly
heart development.42 The mechanism whereby WNT inhibi- enhanced mortality attributable to pulmonary congestion. The
tion by secreted frizzled related proteins improves cardiac phenotype resembles a dilated cardiomyopathy with grossly
remodeling therefore remains unclear. A detailed analysis of enlarged hearts (end-diastolic diameter of 5.5⫾0.2 mm [TG]
signaling molecules downstream of WNT/frizzled interaction versus 4.4⫾0.1 mm [CT]) and reduced ejection fraction. A
may allow identifying this missing link. compensatory increase in myocyte cross-sectional area and
WNT activates the noncanonical pathway including the myocardial fibrosis was observed. Downstream of dishev-
axis of phospholipase C (PLC). PLC in turn leads to calcium eled, the 2 noncanonical pathways of JNK and CaMKII, as
release within the cell. Calcium release induces Ca/calmod- well as c-Myc, were constitutively activated. Mechanistically,
ulin kinase (CaMK)II and calcineurin activation (Figure 3).32 cell culture experiments with small interfering RNA–medi-
Inhibition of CaMKII and calcineurin including downstream ated knockdown of disheveled suggest the protein to be
transcription factors like NF-AT and MEF2 was demonstrated to required and sufficient for ␤-adrenergic (isoproterenol)-
inhibit cardiac hypertrophy and attenuate LV remodeling.43 induced cardiomyocyte hypertrophy. Activation of the canon-
However, the dramatic effects exerted by sFRP-expression on ical WNT pathway with ␤-catenin as the downstream target
LV-remodeling following experimental infarct are significantly may also contribute to this phenotype.48 Interestingly, myo-
more pronounced than those observed in transgenic mice with fibroblasts known to contribute to myocardial wound healing
CaMKII, NF-AT, or MEF2 deletion. Inhibition of cardiac are also regulated by WNT signaling. Migration of cardiac
hypertrophy and/or cardiomyocyte apoptosis is not sufficient fibroblasts immortalized by stable transfection of telomerase
to explain the phenotypes observed.44 Therefore, the WNT- was attenuated by active WNT signaling. The study indicates
mediated activation of ␤-catenin-dependent transcription in that myofibroblast migration and differentiation, but not
the canonical pathway appears to be central to the observed proliferation, can be modulated by interventions in Wnt/Frz
phenotype. WNT leads to inhibition of GSK3␤ via intracel- signaling.49 In summary, attenuation of cardiac fibrosis after
lular binding of axin, thereby preventing ␤-catenin phosphor- infarct may also contribute to the positive effects on LV
ylation and subsequent degradation by the proteasome.32 The remodeling seen by WNT inhibition (Figure 4).
1204 Circulation Research November 12, 2010

Similarly to this heart-specific gain-of-function mutation attenuated LV remodeling and improved ventricular func-
regarding disheveled, a disheveled isoform knockout strain tion.14,31,55 Conversely, increased mortality and a phenotype
was characterized regarding an adult cardiac phenotype. No of dilated cardiomyopathy were observed in ␤-catenin gain-
baseline phenotype was observed. Because ␤-catenin knock- of-function mutations.14,31 Conditional deletion of ␤-catenin
out mutations are embryonic lethal this either indicates that was achieved by either using the ␣MHC-MerCreMer or
disheveled is predominantly involved in the noncanonical ␣MHC-CrePR1 strains. When studied in a TAC model of
WNT pathway or other isoforms compensate for the loss of pressure overload, mice with ␣MHC-MerCreMer mediated
disheveled 1. Interestingly, cardiac hypertrophy as assessed ␤-catenin deletion subjected to the same gradient as control
by heart weight, LV wall thickness in echocardiography and mice had significantly reduced cardiac hypertrophy (heart
gene expression (atrial natriuretic factor, brain natriuretic weight CT: 138⫾11 g versus TG: 111⫾4 g) with increased
peptide) on pressure overload was attenuated in disheveled 1 fractional shortening (CT: 36.2⫾2.7%; TG: 39.1⫾2.4%) and
knockout animals.50 The cellular mechanism of this observa- less LV dilation (echo LV end diastolic dimension: CT,
tion remains unclear; an association to increased GSK3␤ and 3.56⫾0.14 mm; TG, 3.5⫾0.09 mm).56
AKT phosphorylation was described. The phenotype though We studied both gain-of-function (␣MHC-CrePR1⫻⌬N-
is also similar to knockout models of the noncanonical catenin) and loss-of function (␣MHC-CrePR1⫻␤-cat⌬ex3-6) mu-
pathway, ie, modulations of CaMKII.51 tants on 2 weeks of Ang II stimulation. Mice with stabilized
The phenotype in disheveled-1 knockout mice might also ␤-catenin in ␣MHC-positive cells had severely impaired
be linked to the role of WNT signaling in cardiac develop- fractional shortening at the end of the treatment course (TG:
ment and adult cardiac remodeling already summarized 22.⫾4.5% versus CT: 36.2⫾1.6%), whereas mice with
above. Specifically, disheveled is both necessary and suffi- ␤-catenin depletion had no significant phenotype compared
cient for the fusion of early heart precursors. Diversin links to their control littermates.14 Following chronic ligation of the
the Frz/disheveled membrane complex to the downstream left anterior descending coronary artery, ␤-catenin loss-of-
noncanonical pathway with activation of Rho and Rac. function mutants had significantly decreased mortality, re-
Diversin depletion resulted in 2 independently beating hearts duced infarct size, and improved fractional shortening four
in the zebrafish. This phenotype of cardia bifida was rescued weeks after the infarct.31 No significant effect on ejection
by RhoA injection.52 The data prove the noncanonical WNT fraction was observed at 2 weeks post infarct, suggesting that
pathway to be important for cardiac development. Given the modulation of LV remodeling rather than an acute effect on
role of disheveled /diversin in WNT signaling, these signaling the infarct size is responsible for this functional effect. This
molecules may be interesting therapeutic targets because the phenotype is very similar to the observations following
Downloaded from http://ahajournals.org by on June 13, 2021

downstream effectors RhoA, JNK, and CaMKII are known to expression of soluble frizzled receptors in the border zone of
be important mediators of maladaptive cardiac remodeling. the infarct.7,8,35
Frizzled receptors couple to heterotrimeric G proteins includ- Several mechanisms whereby ␤-catenin depletion im-
ing G␣i and G␣q.53 This state of affairs links WNT signaling to proves LV function following pressure overload or ischemia
the activation of MAPK JNK. The role of G␣i and G␣q, both have been described (Figure 4). Among other targets, we
classic 7-transmembrane receptors, in cardiac hypertrophy is consistently found ␤-catenin to regulate Tbx5, a key tran-
well established.54 WNT activation also leads to recruitment of scription factor in differentiation of first heart field progeni-
factors like ␤-arrestins, which mediate signal transduction from tors toward a cardiomyocyte phenotype.31 In association with
G␣i to the MAPK signaling cascade.53 In summary, several lines the maladaptive LV remodeling observed in gain-of-function
of evidence support both the noncanonical and the canonical ␣MHC-restricted ␤-catenin mutants subjected to chronic Ang
WNT pathway as involved in WNT/Frz-mediated maladaptive II stimulation, heart Tbx5 gene expression was reduced. In
cardiac hypertrophy. contrast, Tbx5, as well as GATA4, was upregulated in heart
tissue of transgenic mice with ␤-catenin loss-of-function
Evidence for ␤-Catenin Inhibition to mutations restricted to cells with activation of the ␣MHC-Cre
Attenuate LV Remodeling construct. This phenotype was associated with increased
More than 90% of the cellular ␤-catenin protein content is differentiation of cardiac-endogenous progenitor cells toward
bound in the membrane. Loss of ␤-catenin, however, does not a cardiomyocyte cell type in cell culture experiments with the
lead to a “membrane” phenotype because plakoglobin (␥- noncardiomyocyte fraction of heart cells from such trans-
catenin) functionally substitutes for ␤-catenin in the mem- genic mice. The cells affected expressed ␣MHC and Tbx5
brane in various tissue compartments including the heart.55 protein but not troponin T or islet-1.31 This suggests first heart
Modulations of ␤-catenin levels in the heart by WNT signal- field progenitors to be a target for negative WNT signaling in
ing or genetic modifications therefore predominantly affect the adult heart. Such cells could be identified both in the
␤-catenin– dependent transcription. murine left ventricle and in specimens from human left atrial
Both gain- and loss-of-function mutations of ␤-catenin are appendage (Figure 2). The amount of cells present in the adult
embryonic lethal between E5.5 and E10.5.32 To study the role heart as quantified by FACS of the noncardiomyocyte cell
of ␤-catenin in the adult heart, several conditional mutants fraction from murine left ventricle is sufficient to link
have been generated. Such mice exhibited no phenotype differentiation of these cells to the phenotype of attenuated
under baseline conditions.14,56 On pressure overload, chronic LV remodeling in ␣MHC/␤-catenin– depleted mice. The
angiotensin (Ang) II stimulation or experimental infarct identification of specific markers for first heart field CPCs in
␣MHC-dependent ␤-catenin loss-of-function mutations led to the adult heart is required to further analyze this hypothesis;
Bergmann WNT and Cardiac Remodeling 1205

Tbx5, ␣MHC, and other currently known markers are not situation in the heart regarding angiogenesis but not myogen-
suitable because they are also expressed in adult esis. These studies support the notion that paracrine effects of
cardiomyocytes. implanted cells are sufficient and required to enhance wound
healing and neoangiogenesis. One such study evaluated the
WNT Inhibition Enhances Mobilization of healing potential of human aorta-derived CD133pos progeni-
Endothelial Lineage Progenitor Cells From tor cells and their conditioned medium in an experimental
Bone Marrow model of ischemic diabetic ulcer.39 The CD133pos cells were
As described above, the embryonic heart develops from found to initially secrete high levels of WNT molecules,
several different CPCs. Aside from first heart field CPCs which were downregulated on differentiation into CD133neg
marked by ␣MHC and Tbx5, cells from the endothelial cells. The latter instead secreted soluble frizzled-related
lineage are also affected by WNT signaling.57 WNTs play a proteins sFRP1, -3, and -4 known to downregulate WNT-
key role in embryogenic vasculogenesis by modulating ex- dependent signaling. Consistently, conditioned medium from
pansion of primitive VEGF receptor2pos and postnatal angio- CD133pos cells accelerated wound healing and reparative
genesis.40 Mobilization and homing of vasculogenic progen- angiogenesis in comparison to vehicle.39 As depicted above,
itor cells have shown potential to enhance myocardial tissue sFRP2 was identified to be the key paracrine factor secreted
repair via enhanced neovascularization in the adult.58 Modu- by mesenchymal stem cells injected into the heart to mediate
lation of the WNT/␤-catenin signaling axis in the adult might myocardial survival and repair after an ischemic event.
allow for enhanced mobilization and homing of such cells CD133pos cells have been studied in human trials on
from bone marrow to the heart.59 chronic ischemic cardiomyopathy. One of the studies ana-
The intracellular WNT signaling antagonist dickkopf lyzed the effect parallel to surgical revascularization: patients
(Dkk)-1 was studied in the context of vasculogenic progenitor were subjected to autologous CD133pos cell transplantation
cells resident in the bone marrow. Recombinant Dkk1 injected into the ischemic border zone during the operating procedure
intraperitoneally induced mobilization of Flk1pos/sca-1pos pro- after autologous bone marrow harvest the day before. The
genitor cells. Secretion of the osteoclast differentiation factor therapy was safe and resulted in increased ejection fraction
RANKL was identified as the molecular mechanism. and other parameters of LV function compared to a well-
RANKL in turn induced release of cathepsin K responsible matched control group.62
for selective mobilization of vasculogenic progenitor cells. In summary, the WNT pathway is pivotal for vascular and
The inhibitory effect of Dkk-1 on WNT signaling was angiogenic development. Yet different conclusions depend-
measured by expression analysis of axin-2. This mobilization ing on the experimental settings have been drawn whether
Downloaded from http://ahajournals.org by on June 13, 2021

was sufficient to enhance in vivo vascularization of Matrigel stimulation of WNTs ie, by paracrine activation or inhibition
plugs implanted subcutaneously. Different from other growth of this pathway by dickkopf or sFRP is sufficient to improve
factors stimulating release of bone marrow– derived vascular angiogenesis. One possible explanation is that biphasic control
CPCs like G-CSF, Dkk1 did not lead to an induction of of WNT signaling is required for a beneficial effect where WNT
inflammatory cells from the bone marrow. Even more intrigu- activation promotes proliferation of the vasculogenic precursors,
ing was that the mechanism of vasculogenic cell mobilization whereas inhibition is required for mobilization, homing, and
is quite different between the 2 stimuli: whereas G-CSF differentiation. This interpretation would be consistent with the
reduces the amount of osteoclasts, Dkk1 indirectly cleaves observation that constitutive WNT stimulation leads to stem cell
osteoclast-derived SDF1 via cathepsin K. SDF1 is required exhaustion and multi-lineage blockade.63
for homing of such progenitor cells to ischemic areas includ-
ing the heart (Figure 4).59 Nuclear Negative Interaction Partners of
Interestingly, Flk1pos cells are multipotent cardiovascular ␤-Catenin Attenuate LV Remodeling in
progenitors that also form the endocardium and directly Association With Cell Fate Determination
respond to both BMP and WNT signaling regarding differ- Krüppel-like factors (KLFs) are a large family of zinc
entiation once homed to the myocardium.60 In vitro, WNT finger– containing transcription factors involved in regulating
activation of such cells suppressed myocyte specification, cell differentiation, cardiac remodeling, hematopoiesis, an-
whereas early Noggin⫹Dkk1 exposure significantly en- giogenesis, and stem cell fate determination by interacting
hanced cardiomyocyte specification. These data suggest that with coactivators and corepressors.64 Recent studies revealed
the WNT pathway not only controls mobilization, prolifera- the important role of KLFs as regulators of cardiac biology.65
tion, and differentiation of CPCs but also determines cell fate The family of KLFs was found to control ␤-catenin– depen-
decisions from early progenitors.60 Key markers of cell fate dent transcription in several tissue compartments like the gut
decision monitored in this study were similar to other studies: and the heart. Possibly, KLF proteins allow for tissue specific
Tbx5 and GATA4 for first heart field CPCs, Isl1 for second control of ␤-catenin transcription because KLF4 regulates
heart field CPCs and Flk1, and CD31 for hematopoietic/ transcription in the small intestine whereas we found another
vascular progenitor cells. member of the same family, namely KLF15, to control
The canonical WNT pathway is activated by ischemic ␤-catenin– dependent transcription in the heart.
events. In addition to affect endothelial cells it was found to Specifically, we searched for cardiac-specific ␤-catenin inter-
regulate smooth muscle cell proliferation and apoptosis.61 action partners able to modify the Wnt/␤-catenin transcriptional
Several studies have analyzed the role of WNTs in peripheral activity specifically in the heart using a yeast 2-hybrid system
ischemia, which probably can be transferred to the ischemic with a cardiac-specific library. Our own unpublished data
1206 Circulation Research November 12, 2010

describe a novel interaction between the KLF15 and ␤-catenin functional cardiomyocytes in the adult heart. This is sufficient
resulting in an inhibition of ␤-catenin/TCF3 transcriptional for functionally relevant adult heart regeneration in the
activity via the N-terminal domain of KLF15 in cardiomyocytes. context of aging as well as following injury.
Moreover, Nemo-like kinase (NLK), a Wnt/␤-catenin signaling
inhibitor, was found to interact with KLF15. NLK is activated Clinical Implications of Available Data
downstream of the noncanonical WNT pathway via CaMKII Regarding WNT Signaling in Adult
linking the 2 pathways. Cardiac Remodeling
KLF15 is expressed at high levels only in the heart. KLF15 The data summarized above prove the WNT pathway to be
was previously reported to modulate cardiomyocyte hyper- required and sufficient for adult LV remodeling. Namely,
trophy and fibrosis under stress.66,67 We found that mice with inhibition of the WNT pathway on several levels (soluble
global KLF15 deletion develop normally with no apparent frizzled receptors, dickkopf, inhibition of disheveled, ␤-catenin
phenotype at baseline. However, on aging and after patho- depletion) has proven beneficial for cardiac remodeling (Figures
logical stress the mice exhibit progressive cardiac deteriora- 3 and 4). The noncanonical WNT pathway links WNT signaling
tion whereas no apparent defects were observed in other to well known players in adult cardiac remodeling, namely
organs. The cardiac phenotype was associated with a reduc- RhoA and CaMKII. Given the new findings of an interplay
tion of cardiogenic precursor cells (Sca1pos/␣MHCpos and between the noncanonical and canonical pathways, the latter
Tbx5pos/cTnTneg)60 and upregulation of the vascular (CD31) comes into focus.46 The role of ␤-catenin in cardiac development
and endocardial Flk1 progenitor pool: the total CD31pos and is now well established. The canonical WNT/␤-catenin pathway
CD31pos/Ki67pos, as well as the Flk1pos/Ki67pos, CPC pool has a conserved role in vertebrate heart development, regulating
was increased, as revealed by FACS analysis of the noncar- and restricting cardiac precursor cell formation and subsequent
diomyocyte fraction of the left ventricle from KLF15 knock- heart muscle differentiation.
out mice as compared to control mice. We conclude that Several lines of evidence, both in vitro and in vivo, suggest
KLF15 determines CPC cell fate via ␤-catenin in the adult cardiac CPCs and their responsiveness to WNT signaling to
heart. Moreover, these data support the concept that mainte- be a possible therapeutic target. Some authors suggest the ex
nance of CPC-driven cardiomyocyte regeneration is required vivo amplification by WNT stimulation and reinsertion of
for cell homeostasis during cardiac aging. In addition, cell amplified cells to be a favorable model.3 Given the high
fate regulation is important for adaptive remodeling on prevalence of cells resembling first heart field cardiac pro-
cardiac injury by ischemic events or increased afterload as genitors in the adult (Figure 1), boosting endogenous repair
observed in arterial hypertension. mechanisms also appears feasible. One such approach build-
Downloaded from http://ahajournals.org by on June 13, 2021

As a note of caution to this concept, one must recognize ing on the above summarized data would aim to inhibit WNT
that until the identification of an unequivocally accepted signaling temporarily in the heart. This can be achieved by
marker of adult LV cardiac progenitor cells, these data only local injection of bone marrow– derived mononuclear cells or
provide associations of heart physiology with biochemical subpopulations thereof like CD133pos cells, which were
and cellular events in the CPC pool but no causal relationship. found to be capable of secreting sFRP.39 These molecules
New concepts are warranted to decipher the role of CPCs in scavenge WNT proteins and prohibit binding of WNTs to
the adult heart. Interestingly, second heart field progenitors membrane-bound frizzled receptors.
giving rise to the right ventricle are marked by islet-1 and In summary, the WNT pathway plays a pivotal role in adult
have been characterized in detail.20 –22,68 Proliferation and cardiac remodeling and may be suitable for therapeutic
differentiation of islet-1pos cells isolated from the embryo are interventions. Among several currently discussed molecular
also controlled by WNT/␤-catenin, however only few islet- and cellular mechanisms whereby WNT inhibition attenuates
1pos cells could be isolated from the adult heart suggesting a LV remodeling (Figure 4), reactivation of the developmental
limited role of this precursor cell pool after birth. In contrast, program building the left ventricle from first heart field
we found ⬇10% of the noncardiomyocyte cell fraction of the progenitors in the embryo appears feasible. This maneuver
left ventricle to express a reporter gene controlled by the may restore functional LV myocardium from resident precur-
␣MHC-promoter with 2 different conditional mouse strains, sor cells without the need of exogenous cell therapy. To prove
namely the ␣MHC-CrePR1 and the ␣MHC-MerCreMer.31 this hypothesis, consensus must be found regarding which
This cell fraction overlaps in part with previously described markers are specific for adult heart LV CPCs. It is now clear
adult heart CPCs, namely the c-kitpos cells, as well as the that several different adult heart CPCs can be identified
sca-1pos cell population.31 However, both c-kit and sca-1 are similar to the situation in the developing heart. Tissue-
global markers of stem and progenitor cell pools rather than unspecific markers like c-kit or sca-1 are indirect markers that
being heart-specific. Future research needs to identify specif- not suitable for cell tracking. Islet-1pos cells will only be
ically the cardiac progenitor cell pool; currently available relevant for the second heart field progenitors restricted to the
data suggest several different cell pools including locally right ventricle and the left ventricle outflow tract in the
resident cardiomyogenic CPCs, as well as angiogenic cells developing heart. The population of ␣MHCpos/cTnTneg/
possibly homing from the bone marrow, to be involved in Tbx5pos/eHandpos cells similar to first heart field progenitors
adult cardiac remodeling. in the developing heart (Figure 2) is a possible candidate for
On the basis of the currently available data from our group adult LV cardiac precursor cells. These cells are responsive to
and others, we believe that negative WNT/␤-catenin signal- negative WNT signaling and appear to be able to regenerate
ing enhances first heart field CPC differentiation toward functional LV myocardium in vivo.31 Whatever the mecha-
Bergmann WNT and Cardiac Remodeling 1207

nism, inhibition of WNT in the adult heart may allow for MesP1 drives vertebrate cardiovascular differentiation through Dkk-1-
additional therapeutic interventions on top of currently avail- mediated blockade of Wnt-signalling. Nat Cell Biol. 2008;10:338 –345.
12. Blin G, Nury D, Stefanovic S, Neri T, Guillevic O, Brinon B, Bellamy V,
able therapy. Rucker-Martin C, Barbry P, Bel A, Bruneval P, Cowan C, Pouly J,
Mitalipov S, Gouadon E, Binder P, Hagege A, Desnos M, Renaud JF,
Acknowledgments Menasche P, Puceat M. A purified population of multipotent cardiovas-
M.W.B. apologizes for the many interesting articles that have not cular progenitors derived from primate pluripotent stem cells engrafts in
postmyocardial infarcted nonhuman primates. J Clin Invest. 2010;120:
been covered in this review because of space restrictions. M.W.B.
1125–1139.
thanks Claudia Noack, MSc; Maria-Patapia Zafiriou, PhD; and Laura
13. Zhu W, Shiojima I, Ito Y, Li Z, Ikeda H, Yoshida M, Naito AT, Nishi J,
Zelarayan, PhD (ECRC, Charité Campus Buch, & Max-Delbrück Ueno H, Umezawa A, Minamino T, Nagai T, Kikuchi A, Asashima M,
Center, Berlin, Germany) for expert scientific work and fruitful Komuro I. IGFBP-4 is an inhibitor of canonical Wnt signalling required
discussions. Invaluable technical assistance over many years by for cardiogenesis. Nature. 2008;454:345–349.
Bärbel Pohl (MDC Berlin) formed the solid foundation of this work. 14. Baurand A, Zelarayan L, Betney R, Gehrke C, Dunger S, Noack C,
M.W.B. thanks Kai Jaquet, PhD (Department of Cardiology, St Busjahn A, Huelsken J, Taketo MM, Birchmeier W, Dietz R, Bergmann
Georg, Hamburg, Germany) and Stephan Geidel, MD (Department MW. Beta-catenin downregulation is required for adaptive cardiac
of Cardio-thoracic surgery, St Georg, Hamburg, Germany) for remodeling. Circ Res. 2007;100:1353–1362.
fruitful cooperation regarding human cardiac progenitor cells. Inter- 15. Santini MP, Tsao L, Monassier L, Theodoropoulos C, Carter J,
esting scientific discussions with Leon de Windt (Maastricht Uni- Lara-Pezzi E, Slonimsky E, Salimova E, Delafontaine P, Song YH,
versity Medical Center, The Netherlands) and Jean-Luc Balligand Bergmann M, Freund C, Suzuki K, Rosenthal N. Enhancing repair of the
(Unit of Pharmacology and Therapeutics, University of Louvain mammalian heart. Circ Res. 2007;100:1732–1740.
Medical School, Brussels, Belgium) contributed to the opinions 16. Gessert S, Kuhl M. The multiple phases and faces of Wnt signaling
expressed in this article. during cardiac differentiation and development. Circ Res. 2010;107:
186 –199.
17. Olson EN. Development. The path to the heart and the road not taken.
Sources of Funding Science. 2001;291:2327–2328.
Supported by Deutsche Forschungsgemeinschaft grant BE 2025/8-2 18. Klaus A, Saga Y, Taketo MM, Tzahor E, Birchmeier W. Distinct roles of
and grants from the Jürgen Manchot and the Grimmke Foundation. Wnt/beta-catenin and BMP signaling during early cardiogenesis. Proc
Natl Acad Sci U S A. 2007.
Disclosures 19. Gessert S, Kuhl M. Comparative gene expression analysis and fate
None. mapping studies suggest an early segregation of cardiogenic lineages in
xenopus laevis. Dev Biol. 2009;334:395– 408.
20. Lin L, Cui L, Zhou W, Dufort D, Zhang X, Cai CL, Bu L, Yang L, Martin
References J, Kemler R, Rosenfeld MG, Chen J, Evans SM. Beta-catenin directly
1. Oka T, Xu J, Molkentin JD. Re-employment of developmental tran- regulates islet1 expression in cardiovascular progenitors and is required
scription factors in adult heart disease. Semin Cell Dev Biol. 2007;18: for multiple aspects of cardiogenesis. Proc Natl Acad Sci U S A. 2007;
117–131. 104:9313–9318.
Downloaded from http://ahajournals.org by on June 13, 2021

2. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabe-Heider F, 21. Kwon C, Qian L, Cheng P, Nigam V, Arnold J, Srivastava D. A regu-
Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisen latory pathway involving Notch1/beta-catenin/Isl1 determines cardiac
J. Evidence for cardiomyocyte renewal in humans. Science. 2009;324: progenitor cell fate. Nat Cell Biol. 2009;11:951–957.
98 –102. 22. Qyang Y, Martin-Puig S, Chiravuri M, Chen S, Xu H, Bu L, Jiang X, Lin
3. Domian IJ, Chiravuri M, van der Meer P, Feinberg AW, Shi X, Shao Y, L, Granger A, Moretti A, Caron L, Wu X, Clarke J, Taketo MM,
Wu SM, Parker KK, Chien KR. Generation of functional ventricular heart Laugwitz KL, Moon RT, Gruber P, Evans SM, Ding S, Chien KR. The
muscle from mouse ventricular progenitor cells. Science. 2009;326: renewal and differentiation of Isl1⫹ cardiovascular progenitors are con-
426 – 429. trolled by a Wnt/beta-catenin pathway. Cell Stem Cell. 2007;1:165–179.
4. Ueno S, Weidinger G, Osugi T, Kohn AD, Golob JL, Pabon L, Reinecke 23. Singh AM, Li F-Q, Hamazaki T, Kasahara H, Takemaru K-I, Terada N.
H, Moon RT, Murry CE. Biphasic role for Wnt/beta-catenin signaling in Chibby, an antagonist of the Wnt/{beta}-catenin pathway, facilitates cardio-
cardiac specification in zebrafish and embryonic stem cells. Proc Natl myocyte differentiation of murine embryonic stem cells. Circulation. 2007;
Acad Sci U S A. 2007;104:9685–9690. 115:617–626.
5. Naito AT, Shiojima I, Akazawa H, Hidaka K, Morisaki T, Kikuchi A, 24. Buckingham M, Meilhac S, Zaffran S. Building the mammalian heart
Komuro I. Developmental stage-specific biphasic roles of Wnt/beta- from two sources of myocardial cells. Nat Rev. 2005;6:826 – 835.
catenin signaling in cardiomyogenesis and hematopoiesis. Proc Natl Acad 25. Prall OW, Menon MK, Solloway MJ, Watanabe Y, Zaffran S, Bajolle F,
Sci U S A. 2006;103:19812–19817. Biben C, McBride JJ, Robertson BR, Chaulet H, Stennard FA, Wise N,
6. Malekar P, Hagenmueller M, Anyanwu A, Buss S, Streit MR, Weiss CS, Schaft D, Wolstein O, Furtado MB, Shiratori H, Chien KR, Hamada H,
Wolf D, Riffel J, Bauer A, Katus HA, Hardt SE. Wnt signaling is critical Black BL, Saga Y, Robertson EJ, Buckingham ME, Harvey RP. An
for maladaptive cardiac hypertrophy and accelerates myocardial Nkx2–5/Bmp2/Smad1 negative feedback loop controls heart progenitor
remodeling. Hypertension. 2010;55:939 –945. specification and proliferation. Cell. 2007;128:947–959.
7. Barandon L, Couffinhal T, Ezan J, Dufourcq P, Costet P, Alzieu P, 26. Zeisberg EM, Ma Q, Juraszek AL, Moses K, Schwartz RJ, Izumo S, Pu
Leroux L, Moreau C, Dare D, Duplaa C. Reduction of infarct size and WT. Morphogenesis of the right ventricle requires myocardial expression
prevention of cardiac rupture in transgenic mice overexpressing FrzA. of Gata4. J Clin Invest. 2005;115:1522–1531.
Circulation. 2003;108:2282–2289. 27. Bruneau BG, Bao ZZ, Fatkin D, Xavier-Neto J, Georgakopoulos D,
8. Alfaro MP, Pagni M, Vincent A, Atkinson J, Hill MF, Cates J, Davidson Maguire CT, Berul CI, Kass DA, Kuroski-de Bold ML, de Bold AJ,
JM, Rottman J, Lee E, Young PP. The Wnt modulator sFRP2 enhances Conner DA, Rosenthal N, Cepko CL, Seidman CE, Seidman JG. Cardio-
mesenchymal stem cell engraftment, granulation tissue formation and myopathy in Irx4-deficient mice is preceded by abnormal ventricular
myocardial repair. Proc Natl Acad Sci U S A. 2008;105:18366 –18371. gene expression. Mol Cell Biol. 2001;21:1730 –1736.
9. Lindsley RC, Gill JG, Murphy TL, Langer EM, Cai M, Mashayekhi M, 28. Sanchez A, Jones WK, Gulick J, Doetschman T, Robbins J. Myosin
Wang W, Niwa N, Nerbonne JM, Kyba M, Murphy KM. Mesp1 coor- heavy chain gene expression in mouse embryoid bodies. An in vitro
dinately regulates cardiovascular fate restriction and epithelial- developmental study. J Biol Chem. 1991;266:22419 –22426.
mesenchymal transition in differentiating ESCs. Cell Stem Cell. 2008;3: 29. Lyons GE, Schiaffino S, Sassoon D, Barton P, Buckingham M. Devel-
55– 68. opmental regulation of myosin gene expression in mouse cardiac muscle.
10. Bondue A, Lapouge G, Paulissen C, Semeraro C, Iacovino M, Kyba M, J Cell Biol. 1990;111:2427–2436.
Blanpain C. Mesp1 acts as a master regulator of multipotent cardiovas- 30. Jones WK, Grupp IL, Doetschman T, Grupp G, Osinska H, Hewett TE,
cular progenitor specification. Cell Stem Cell. 2008;3:69 – 84. Boivin G, Gulick J, Ng WA, Robbins J. Ablation of the murine alpha
11. David R, Brenner C, Stieber J, Schwarz F, Brunner S, Vollmer M, myosin heavy chain gene leads to dosage effects and functional deficits in
Mentele E, Muller-Hocker J, Kitajima S, Lickert H, Rupp R, Franz WM. the heart. J Clin Invest. 1996;98:1906 –1917.
1208 Circulation Research November 12, 2010

31. Zelarayán LC, Noack C, Sekkali B, Kmecova J, Gehrke C, Renger A, WM. Interruption of Wnt signaling attenuates the onset of pressure
Zafiriou MP, van der Nagel R, Dietz R, de Windt LJ, Balligand JL, overload-induced cardiac hypertrophy. Hypertension. 2007;49:473– 480.
Bergmann MW. Beta-catenin downregulation attenuates ischemic cardiac 51. Passier R, Zeng H, Frey N, Naya FJ, Nicol RL, McKinsey TA, Overbeek
remodeling through enhanced resident precursor cell differentiation. Proc P, Richardson JA, Grant SR, Olson EN. CaM kinase signaling induces
Natl Acad Sci U S A. 2008;105:19762–19767. cardiac hypertrophy and activates the MEF2 transcription factor in vivo.
32. Grigoryan T, Wend P, Klaus A, Birchmeier W. Deciphering the function J Clin Invest. 2000;105:1395–1406.
of canonical Wnt signals in development and disease: conditional loss- 52. Moeller H, Jenny A, Schaeffer HJ, Schwarz-Romond T, Mlodzik M,
and gain-of-function mutations of beta-catenin in mice. Genes Dev. 2008; Hammerschmidt M, Birchmeier W. Diversin regulates heart formation
22:2308 –2341. and gastrulation movements in development. Proc Natl Acad Sci U S A.
33. Schumann H, Holtz J, Zerkowski HR, Hatzfeld M. Expression of secreted 2006;103:15900 –15905.
frizzled related proteins 3 and 4 in human ventricular myocardium cor- 53. Force T, Woulfe K, Koch WJ, Kerkela R. Molecular scaffolds regulate
relates with apoptosis related gene expression. Cardiovasc Res. 2000;45: bidirectional crosstalk between Wnt and classical seven-transmembrane-
720 –728. domain receptor signaling pathways. Sci STKE. 2007;2007:pe41.
34. Dufourcq P, Couffinhal T, Ezan J, Barandon L, Moreau C, Daret D, 54. Syed F, Odley A, Hahn HS, Brunskill EW, Lynch RA, Marreez Y, Sanbe
Duplaa C. FrzA, a secreted frizzled related protein, induced angiogenic A, Robbins J, Dorn GW II. Physiological growth synergizes with patho-
response. Circulation. 2002;106:3097–3103. logical genes in experimental cardiomyopathy. Circ Res. 2004;95:
35. Mirotsou M, Zhang Z, Deb A, Zhang L, Gnecchi M, Noiseux N, Mu H, 1200 –1206.
Pachori A, Dzau V. Secreted frizzled related protein 2 (sFRP2) is the key 55. Zhou J, Qu J, Yi XP, Graber K, Huber L, Wang X, Gerdes AM, Li F.
Akt-mesenchymal stem cell-released paracrine factor mediating myo- Up-regulation of {gamma}-catenin compensates for the loss of {beta}-
cardial survival and repair. Proc Natl Acad Sci U S A. 2007;104: catenin in adult cardiac myocytes. Am J Physiol Heart Circ Physiol.
1643–1648. 2007;292:H270 –H276.
36. Zhang Z, Deb A, Pachori A, He W, Guo J, Pratt R, Dzau VJ. Secreted 56. Chen X, Shevtsov SP, Hsich E, Cui L, Haq S, Aronovitz M, Kerkela R,
frizzled related protein 2 protects cells from apoptosis by blocking the Molkentin JD, Liao R, Salomon RN, Patten R, Force T. The {beta}-
effect of canonical Wnt3a. J Mol Cell Cardiol. 2009;46:370 –377. catenin/T-cell factor/lymphocyte enhancer factor signaling pathway is
37. Kobayashi K, Luo M, Zhang Y, Wilkes DC, Ge G, Grieskamp T, Yamada required for normal and stress-induced cardiac hypertrophy. Mol Cell
C, Liu TC, Huang G, Basson CT, Kispert A, Greenspan DS, Sato TN. Biol. 2006;26:4462– 4473.
Secreted frizzled-related protein 2 is a procollagen c proteinase enhancer 57. Logan CY, Nusse R. The Wnt signaling pathway in development and
with a role in fibrosis associated with myocardial infarction. Nat Cell disease. Annu Rev Cell Dev Biol. 2004;20:781– 810.
Biol. 2009;11:46 –55.
58. Ziebart T, Yoon CH, Trepels T, Wietelmann A, Braun T, Kiessling F,
38. Barandon L, Dufourcq P, Costet P, Moreau C, Allieres C, Daret D, Dos
Stein S, Grez M, Ihling C, Muhly-Reinholz M, Carmona G, Urbich C,
Santos P, Daniel Lamaziere JM, Couffinhal T, Duplaa C. Involvement of
Zeiher AM, Dimmeler S. Sustained persistence of transplanted proan-
frza/sFRP-1 and the Wnt/frizzled pathway in ischemic preconditioning.
giogenic cells contributes to neovascularization and cardiac function after
Circ Res. 2005;96:1299 –1306.
ischemia. Circ Res. 2008;103:1327–1334.
39. Barcelos LS, Duplaa C, Krankel N, Graiani G, Invernici G, Katare R,
59. Aicher A, Kollet O, Heeschen C, Liebner S, Urbich C, Ihling C, Orlandi
Siragusa M, Meloni M, Campesi I, Monica M, Simm A, Campagnolo P,
A, Lapidot T, Zeiher AM, Dimmeler S. The Wnt antagonist dickkopf-1
Mangialardi G, Stevanato L, Alessandri G, Emanueli C, Madeddu P.
mobilizes vasculogenic progenitor cells via activation of the bone marrow
Human CD133⫹ progenitor cells promote the healing of diabetic ische-
endosteal stem cell niche. Circ Res. 2008;103:796 – 803.
Downloaded from http://ahajournals.org by on June 13, 2021

mic ulcers by paracrine stimulation of angiogenesis and activation of Wnt


60. Misfeldt AM, Boyle SC, Tompkins KL, Bautch VL, Labosky PA,
signaling. Circ Res. 2009;104:1095–1102.
Baldwin HS. Endocardial cells are a distinct endothelial lineage derived
40. Ezan J, Leroux L, Barandon L, Dufourcq P, Jaspard B, Moreau C,
Allieres C, Daret D, Couffinhal T, Duplaa C. FrzA/sFRP-1, a secreted from Flk1⫹ multipotent cardiovascular progenitors. Dev Biol. 2009;333:
antagonist of the Wnt-frizzled pathway, controls vascular cell prolif- 78 – 89.
eration in vitro and in vivo. Cardiovasc Res. 2004;63:731–738. 61. Couffinhal T, Dufourcq P, Duplaa C. Beta-catenin nuclear activation:
41. Wang X, Xiao Y, Mou Y, Zhao Y, Blankesteijn WM, Hall JL. A role for common pathway between Wnt and growth factor signaling in vascular
the {beta}-catenin/T-cell factor signaling cascade in vascular remodeling. smooth muscle cell proliferation? Circ Res. 2006;99:1287–1289.
Circ Res. 2002;90:340 –347. 62. Stamm C, Kleine H-D, Choi Y-H, Dunkelmann S, Lauffs J-A, Lorenzen
42. van de Schans VA, Smits JF, Blankesteijn WM. The Wnt/frizzled B, David A, Liebold A, Nienaber C, Zurakowski D, Freund M, Steinhoff
pathway in cardiovascular development and disease: friend or foe? Eur G. Intramyocardial delivery of CD133⫹ bone marrow cells and coronary
J Pharmacol. 2008;585:338 –345. artery bypass grafting for chronic ischemic heart disease: safety and
43. van Empel VPM, De Windt LJ. Myocyte hypertrophy and apoptosis: a efficacy studies. J Thorac Cardiovasc Surg. 2007;133:717–725.
balancing act. Cardiovasc Res. 2004;63:487– 499. 63. Scheller M, Huelsken J, Rosenbauer F, Taketo MM, Birchmeier W,
44. Zelarayan L, Gehrke C, Bergmann MW. Role of beta-catenin in adult Tenen DG, Leutz A. Hematopoietic stem cell and multilineage defects
cardiac remodeling. Cell Cycle. 2007;6:2120 –2126. generated by constitutive ␤-catenin activation. Nat Immunol. 2006;7:
45. van Gijn ME, Snel F, Cleutjens JP, Smits JF, Blankesteijn WM. Over- 1037–1047.
expression of components of the Frizzled-Dishevelled cascade results in 64. Kaczynski J, Cook T, Urrutia R. Sp1- and Kruppel-like transcription
apoptotic cell death, mediated by beta-catenin. Exp Cell Res. 2001;265: factors. Genome Biol. 2003;4:206.
46 –53. 65. Haldar SM, Ibrahim OA, Jain MK. Kruppel-like factors (KLFs) in muscle
46. Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: a biology. J Mol Cell Cardiol. 2007;43:1–10.
prelude for more. Circ Res. 2010;106:1798 –1806. 66. Fisch S, Gray S, Heymans S, Haldar SM, Wang B, Pfister O, Cui L,
47. Backs J, Song K, Bezprozvannaya S, Chang S, Olson EN. CaM kinase II Kumar A, Lin Z, Sen-Banerjee S, Das H, Petersen CA, Mende U,
selectively signals to histone deacetylase 4 during cardiomyocyte hyper- Burleigh BA, Zhu Y, Pinto YM, Liao R, Jain MK. Kruppel-like factor 15
trophy. J Clin Invest. 2006;116:1853–1864. is a regulator of cardiomyocyte hypertrophy. Proc Natl Acad Sci U S A.
48. Zelarayan LC, Noack C, Zafiriou MP, Renger A, Bergmann MW. Wnt 2007;104:7074 –7079.
signaling molecules in left ventricular remodeling: focus on dishevelled 1. 67. Wang B, Haldar SM, Lu Y, Ibrahim OA, Fisch S, Gray S, Leask A, Jain
Hypertension. 2010;55:852– 854. MK. The Kruppel-like factor KLF15 inhibits connective tissue growth
49. Laeremans H, Rensen SS, Ottenheijm HC, Smits JF, Blankesteijn WM. factor (CTGF) expression in cardiac fibroblasts. J Mol Cell Cardiol.
Wnt/frizzled signalling modulates the migration and differentiation of 2008;45:193–197.
immortalized cardiac fibroblasts. Cardiovasc Res. 2010;87:514 –523. 68. Bu L, Jiang X, Martin-Puig S, Caron L, Zhu S, Shao Y, Roberts DJ, Huang
50. van de Schans VA, van den Borne SW, Strzelecka AE, Janssen BJ, van PL, Domian IJ, Chien KR. Human Isl1 heart progenitors generate diverse
der Velden JL, Langen RC, Wynshaw-Boris A, Smits JF, Blankesteijn multipotent cardiovascular cell lineages. Nature. 2009;460:113–117.

You might also like