You are on page 1of 11

Am J Physiol Cell Physiol 293: C1–C11, 2007.

First published March 21, 2007; doi:10.1152/ajpcell.00415.2006. Invited Review

Notch signaling in the developing cardiovascular system


Kyle Niessen1,3 and Aly Karsan1,2,3,4
1
Departments of Medical Biophysics and 2Pathology and Laboratory Medicine, British Columbia
Cancer Agency, Vancouver; and the 3Experimental Medicine Program and 4Department of Pathology
and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada

Niessen K, Karsan A. Notch signaling in the developing cardiovascular system.


Am J Physiol Cell Physiol 293: C1–C11, 2007. First published March 21, 2007;
doi:10.1152/ajpcell.00415.2006.—The Notch proteins encompass a family of trans-
membrane receptors that have been highly conserved through evolution as medi-
ators of cell fate. Recent findings have demonstrated a critical role of Notch in the
developing cardiovascular system. Notch signaling has been implicated in the
endothelial-to-mesenchymal transition during development of the heart valves, in
arterial-venous differentiation, and in remodeling of the primitive vascular plexus.
Mutations of Notch pathway components in humans are associated with congenital
defects of the cardiovascular system such as Alagille syndrome, cerebral autoso-
mal dominant arteriopathy with subcortical infarcts and leukoencephalopathy
(CADASIL), and bicuspid aortic valves. This article focuses on the role of the
Notch pathway in the developing cardiovascular system and congenital human
cardiovascular diseases.
cardiac development; endothelial-mesenchymal transformation; vasculogenesis;
angiogenesis

NOTCH PATHWAY (also known as RBP-J␬ and CBF1) (123). The ankyrin repeats
are involved in protein-protein interactions including interac-
In mammals, the Notch receptor family consists of four type
tion with CSL; however, the seventh ankyrin repeat in coop-
I transmembrane receptors (Notch1 to Notch4) that regulate
eration with the TAD domain recruits transcriptional activators
cell fate decisions through cell-cell interaction (27). Notch
such as mastermind-like (MAML) and the histone acetyltrans-
receptors are translated as a large (⬃300 kDa) precursor
ferase (HAT) complex (69, 124). The PEST domain is in-
protein comprising an extracellular, a transmembrane, and an
volved in regulating protein half-life of the Notch receptors
intracellular domain that undergoes processing in the trans-
(31, 92).
Golgi network by Furin (76). Furin cleavage occurs at the
In mammals, there are five Notch ligands, Jagged1, Jagged2
S1 cleavage site and generates an extracellular fragment
(also called Serrate1 and Serrate2), Delta-like1 (Dll), Dll3, and
(NotchEC) and an extracellular-transmembrane-intracellular
Dll4, collectively referred to as the DSL (Delta/Serrate/Lag-2)
fragment (NotchTM) that is expressed on the cell surface as a
family (Fig. 1). DSL proteins are themselves type I transmem-
noncovalently linked heterodimer stabilized by a Ca2⫹ ion (5) brane proteins, with an extracellular domain comprised of 7–16
(Fig. 1). EGF-like repeats and a DSL domain, which is unique to Notch
The extracellular domain of Notch receptors is comprised of ligands. Jagged1 and Jagged2 have an additional cysteine-rich
29 –36 multiple epidermal growth factor (EGF)-like repeats, domain and a von Willebrand factor type C domain in the
depending on the specific Notch receptor, and 3 lin-12/Notch extracellular region (27).
(LNR) motifs. The EGF-like motifs are responsible for ligand The EGF-like repeats are thought to stabilize receptor-ligand
interaction, while LNR motifs are responsible for preventing interaction, while the DSL domain is responsible for Notch
receptor activation in the absence of receptor-ligand engage- receptor activation through interaction with EGF-like repeats
ment (35, 101, 104, 113). The intracellular domain of Notch 11 and 12 of the Notch receptors (104). The cysteine-rich
receptors is comprised of a recombination signal binding pro- domain of Jagged ligands are thought to control Notch receptor
tein-1 for J␬ (RBP-J␬)-associated molecule (RAM) domain, binding specificity, while the von Willebrand factor type C
seven cdc10/ankyrin repeats, of which only the COOH-termi- domain is thought to be involved in ligand dimerization (27).
nal six assume the proper ankyrin fold, and a transactivation The intracellular regions of the DSL ligands are relatively short
domain (TAD) present in Notch1, Notch2, and Notch3. In and contain a PDZ domain in Jagged1 and Dll1 thought to be
addition, there are two nuclear localization signals, a glu- involved in activating downstream signaling through mecha-
tamine-rich stretch, and a PEST domain (2, 21, 69, 95, 139). nisms that are not well understood currently (1, 118).
The RAM domain is involved in potentiating Notch signaling
through interaction with the transcription factor CSL [C pro-
NOTCH RECEPTOR-LIGAND ACTIVATION
moter binding factor-1 (CBF1), suppressor of hairless, Lag-1]
Notch receptor-ligand interaction is thought to result in a
Address for reprint requests and other correspondence: A. Karsan, Dept. of
conformational change of the Notch extracellular domain ex-
Medical Biophysics, British Columbia Cancer Research Centre, 675 W. 10th posing a motif that is recognized and cleaved by the metallo-
Ave., Vancouver, BC, Canada V5Z 1L3 (e-mail: akarsan@bccrc.ca). protease tumor necrosis factor-␣ converting enzyme TACE/
http://www.ajpcell.org 0363-6143/07 $8.00 Copyright © 2007 the American Physiological Society C1
Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
C2 NOTCH AND CARDIAC DEVELOPMENT

Fig. 1. Notch receptors and ligands. There are 4 Notch receptors (Notch1–
Notch4) and 5 ligands [Jagged1/2, Delta-like (Dll)-1/3/4] in mammals. Notch
receptors are expressed on the cell surface as heterodimers stabilized through
calcium-dependent interactions. The extracellular domain contains 29 –36
epidermal growth factor (EGF)-like repeats (human Notch receptors), 3 Lin-
12/Notch (LNR) repeats, and a heterodimerization domain. The intracellular
domain contains an RBP-J␬-associated molecule (RAM) domain, 7 ankyrin
(ANK) repeats, 2 nuclear localization signals (NLS), a transactivation (TAD)
domain, and a PEST domain. Notch ligands are also expressed on the cell
surface. The extracellular domains contain a Delta/Serrate/Lag2 (DSL) domain
unique to Notch ligands and also contain multiple EGF repeats. Jagged1/2 also
contains a cysteine-rich domain and a von Willebrand factor type C domain.
The intracellular domains of Jagged1 and Dll1 have been shown to contain
PDZ domains, which may interact with downstream signaling components to
activate transcription. (For additional definitions of abbreviations in Figs. 1–3,
see text.)

ADAM17 at the S2 cleavage site (7). S2 cleavage occurs just


extracellular to the plasma membrane releasing the extracellu-
lar Notch fragment. S2 cleavage is followed by two intramem-
branous cleavage steps mediated by the ␥-secretase complex
comprising presenilin1, presenilin2, Pen-2, Aph-1, and
nicastrin (15, 29, 46, 121). The ␥-secretase complex has been
shown to cleave Notch receptors at two distinct sites, the S4
site between Ala1731-Ala1732 and the S3 site at the conserved
Val1744 in the mouse Notch1 protein (94, 116). ␥-Secretase
cleavage ultimately releases the intracellular domain of Notch
(NotchICD), which subsequently translocates to the nucleus to
effect gene transcription (15, 94) (Fig. 2).
The most thoroughly investigated downstream target of
Notch signaling is the CSL transcription factor. In the absence
of NotchICD, CSL recruits either the silencing mediator of
retinoid and thyroid hormone receptors (SMRT)/nuclear recep-
tor corepressor (NcoR)/histone deacetylase (HDAC)-1 or the
CBF1 interacting corepressor (CIR)/HDAC2/Sin3A-associated
protein, 30-kDa (SAP30) complex to negatively regulate target

Fig. 2. The Notch signaling pathway. Notch receptors undergo processing in


the trans-Golgi network by Furin and are expressed on the cell surface as a
noncovalently linked heterodimer stabilized by a calcium ion. Receptor-ligand
interaction results in 3 additional cleavage events that result in the release of
the intracellular region of the Notch receptor (NotchICD). The ectodomain of
the Notch receptor and the ligand are thought to be endocytosed by the
signaling cell. NotchICD then translocates to the nucleus, where it binds and
converts CSL from a transcriptional repressor to a transcriptional activator of
the Hes and Hey family of genes. Notch signaling is negatively regulated by
hyperphosphorylation of nuclear NotchICD by the nuclear kinase CycC:
CDK8, which is recruited to NotchICD by the Notch pathway coactivator
MAML.

AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
NOTCH AND CARDIAC DEVELOPMENT C3
gene expression (45, 59). NotchICD binding to CSL in the
nucleus converts CSL from a transcriptional repressor to a
transcriptional activator. CSL has been shown to bind prefer-
entially to a consensus sequence defined by (C/T)(A/G)TG(A/
G/T)GA(A/G/T) (98). However, most of the identified direct
CSL target genes have a less permissive (C/T)(A/G)TGGGAA
binding sequence. The hairy enhancer of split (HES) and
hairy/enhancer of split-related with YRPW motif (Hey, also
called HESR, CHF, Hrt) families of basic helix-loop-helix
transcription factors are the most thoroughly investigated direct
Notch-CSL targets (13, 48). Other direct targets include Cyclin
D1, p21, glial fibrillary acidic protein (GFAP), Nodal, Myc,
PTEN, Ephrin B2, and smooth muscle ␣-actin (SMA) (34, 36,
50, 62, 65, 90, 102, 106, 132).
The exact composition of transcriptional activators and the
temporal sequence of recruitment to the CSL-NotchICD com-
plex remain to be elucidated. However, it has been shown that
the NotchICD complex includes Ski interacting protein
(SKIP), a protein that can interact with CSL, Notch, or SMRT
but promotes NotchICD-CSL interactions over CSL-SMRT
interaction (74, 138). Additionally, the coactivator MAML
binds the NotchICD-CSL complex but does not bind either
NotchICD or CSL independently (86, 87, 96). MAML itself
has transactivation potential and therefore, in combination with
the NotchICD TAD domain, activates gene transcription (30).
The p300-HAT complex is also recruited to NotchICD, which
in cooperation with MAML is necessary for activation of gene
transcription (30) (Fig. 2).
To negatively regulate NotchICD activity, MAML also
recruits the nuclear kinase CycC:CDK8, which hyperphospho-
rylates the TAD and PEST domain of NotchICD (31). Phos-
phorylation of NotchICD at conserved serine residues in the
PEST domain by CycC:CDK8 recruits the ubiquitin ligase
Fbw7/Sel7, resulting in ubiquitin-mediated proteasome degra-
dation (31). Negative regulation of Notch signaling also occurs Fig. 3. Cardiac development. Heart precursors are identifiable in the anterior
by GSK-3␤ via phosphorylation of NotchICD, and ubiquitina- third of the primitive streak (blue and yellow) but not in the node (dark grey).
tion of NotchICD by the E3-ubiquitin ligases Itch and c-Cbl As embryonic development progresses, the heart precursors undergo epithelial-
to-mesenchymal transition (EMT) and migrate bilaterally from the primitive
targets NotchICD for degradation (24, 28, 52, 99). streak forming the left and right primary heart fields (embryonic day 7.5; E7.5).
In addition to the Notch interacting proteins discussed The primary and secondary (green) heart fields undergo anterior-medial mi-
above, Deltex binds the ankyrin repeats of NotchICD and gration and fusion forming the cardiac crescent (E8.0). The primitive heart
positively regulates Notch signaling, independent of CSL. The tube is formed by fusion of the cardiac crescent at the embryonic midline at
E8.5. The primary heart field gives rise to the left ventricle (blue), right
exact mechanism is unclear but involves targeting NotchICD to ventricle (purple), and atria (yellow), while the secondary heart field gives rise
the late endosome where it accumulates (44, 51, 79). Other to cells of the outflow tract (green). The heart tube undergoes elongation at the
posttranslational events also affect various aspects of Notch atrial and ventricular ends, and looping to form a C-shaped structure is initiated
signaling, including receptor and ligand stability (99) and shortly after formation of the heart tube at E8.5. At E9.5, EMT is initiated in
receptor-ligand specificity (42), and have been extensively the atrioventricular (AV) canal (red) and outflow tract (OFT) (green), and the
cardiac cushions become cellularized by EMT. EMT-derived cells from the
reviewed previously (9, 37, 39, 57, 88). AV canal contribute to the membranous atrial septum and mitral and tricuspid
valves, while OFT EMT-derived cells combined with neural crest-derived cells
CARDIAC DEVELOPMENT contribute to the aortic and pulmonary valves. E9.5 and E11.5 represent images
of mouse embryonic hearts at the respective stages. Original images can be
Early Cardiac Progenitors found at (http://www.mouseatlas.org/morgen/content).
The heart is the first functional organ to form during verte-
brate development. Cells fated to form the heart are first primitive streak forming the left and right heart fields, also
identifiable based on expression of cardiac markers in the referred to as the primary heart field. Formation of the primary
anterior third of the primitive streak (not including the node) heart field is dependent on expression of the basic helix-loop-
during gastrulation (embryonic day 7.0; E7.0) (Fig. 3). The helix (bHLH) transcription factors Mesp1 and Mesp2 (61).
transcriptional coactivator CITED2 is expressed by primitive Mesp1 and Mesp2 double-deficient embryos have a specific
streak cells that will go on to become heart, and represents one defect in the development of the cardiac and anterior-cephalic
of the earliest known markers of cardiac progenitors (114). mesoderm (61). With the use of Mesp1- and Mesp2-deficient
The prospective heart precursors undergo epithelial-to-mesen- embryonic stem cells in chimera embryo studies, it was shown
chymal transition (EMT) and migrate bilaterally from the that Mesp1 and Mesp2 are redundantly required for EMT and
AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org
Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
C4 NOTCH AND CARDIAC DEVELOPMENT

migration of cardiac and anterior-cephalic mesoderm from the the embryo, the cardiac crescent fuses into a linear tube-like
primitive streak (61). Lineage analysis of the Mesp1-derived structure that starts beating at E8 in the mouse and at ⬃3 wk
cells demonstrated that the majority of cells in the myocardium of gestation in humans (117) (Fig. 3).
and endocardium are derived from Mesp1 expressing meso- In addition to the primary heart field, the existence of a
derm (111, 112). secondary heart field has been identified in both the developing
The endoderm underling the primary heart field plays an chick and mouse heart. The secondary heart field is located in
important role in specifying the cardiogenic phenotype. Using the splanchnic mesoderm that underlies the floor of the caudal
transplantation studies of mesoderm from a noncardiogenic pharynx and expresses many of the same markers as the
source transplanted into the cardiogenic field, Inagaki et al. primary heart field such as Nkx2.5 and GATA4, but also
(47) demonstrated that noncardiogenic mesoderm can be re- expresses unique makers such as FGF-10 and Nkx3.1 (60,
programmed to the cardiac fate. This reprogramming was 115). The extent of the contribution of cells from the secondary
further shown to be in part due to secreted factors from the heart field to the adult heart is not fully understood. In the
endoderm, such as members of the bone morphogenetic protein chick, the secondary heart field generates the smooth muscle
(BMP) family, sonic hedgehog, fibroblast growth factor cells of the conus and truncus only (14, 127). In the mouse,
(FGF)-8, and Crescent. In addition to the endoderm, the ecto- cells from the secondary heart field migrate into the arterial
derm secretes Wnt inhibitors that are required for induction of pole between E8.25 and E10.5, and, in addition to the smooth
a cardiogenic fate (reviewed in Ref. 6). muscle cells of the conus and truncus, a population of the
Even at this early stage in heart development, cell fate myocardial cells of the right ventricle are also generated from
analysis reveals that compartmentalization of heart chambers the secondary heart field (60) (Fig. 3, green).
has occurred. Myocardial atrial precursors are present in the
posterior region and ventricular progenitors in the anterior Heart Looping
region of the heart field (105). The most frequently used
Fusion of the cardiac crescent along the midline results in
primary heart field markers are Nkx2.5 (homolog of Drosoph-
the formation of the primitive heart tube. The primitive heart
ila Tinman), BMP2, Tbx20, GATA4, GATA5, and GATA6.
tube is composed of an outer myocardial cell layer and an inner
Although these markers are expressed in the primary heart
endocardial cell layer separated by a layer of extracellular
field, the exact boundaries of the primary heart field are not
matrix called the cardiac jelly. As heart development progresses,
precisely delineated by the expression of these genes. The heart
the linear heart undergoes several morphological changes that
field extends both laterally and medially of Nkx2.5 and BMP2
align and fuse the chambers of the heart. Mouse embryos that
expression, while BMP2 expression extends posterior and
are null for CSL, or Notch1-deficient embryos which also
anterior to the heart field (105).
express a Notch2-hypomorphic mutation, show randomized
The cardiac crescent is formed when the right and left heart
heart looping and axial rotation defects (65, 103). Additionally,
fields undergo anterior-medial migration and subsequently fuse
embryos deficient for the Notch ligand Dll1 display random-
at the anterior end (Fig. 3). Initially, all cells of the cardiac
ized heart looping and axial rotation (65, 97). However, this
crescent have cardiomyogenic potential, but signals from the
phenotype is not reproduced in Notch1-deficient or Notch2-
prospective myocardium and neurogenic tissue subdivide the
deficient embryos (38, 65). Defects in heart looping in Dll1-
cardiac crescent into ventral myogenic and dorsolateral non-
deficient embryos were found to be due to loss or misexpres-
myogenic domains (100). The ventral myogenic domain of the
sion of Nodal, Lefty2, and Pitx2, which are part of the evolu-
cardiac crescent gives rise to the myocardium of the heart tube,
tionarily conserved signaling cascade that controls left-right
while the dorsolateral nonmyogenic domain gives rise to the
morphogenesis. Krebs et al. (65) found that, in Dll1-deficient
mesocardial and pericardial roof cells (100). Studies in Xeno-
embryos, Nodal and Lefty2 expression was absent from the left
pus laevis have demonstrated that the Notch ligand Serrate1
lateral plate mesoderm (LPM), and Pitx2 expression was ran-
(Jagged1) but not Delta1 or Delta2 is expressed in the dorso-
domized in the LPM or not expressed at all (65). However,
lateral nonmyogenic domain, and the dorsal-most region of the
other investigators found that Nodal and Lefty2 expression was
myogenic domain expresses both Serrate1 and Notch1 in an
absent from only 50 and 25% of Dll1-deficient embryos,
overlapping pattern (107). Activation of the Notch pathway by
respectively. In the remaining embryos, Nodal and Lefty2 were
Serrate1 reinforces Serrate1 expression in the dorsolateral
expressed in the left, right, or both LPM (97). The specific
region and suppresses the myogenic potential of cells residing
reasons for the discrepancy between these two groups of
in this region (107). Although Notch activation suppresses
investigators remain to be elucidated, but is likely due to the
myogenic potential, expression of primary heart field markers
difference in genetic backgrounds of the two Dll1-deficient
Nkx2.5 and GATA4 is unaffected (107). In addition, CSL-
strains. Nodal expression was further shown to be dependent
deficient mouse embryos develop a primitive heart tube, and
on two CSL binding sites in a node-specific enhancer located
heart field specification appears normal (93, 119). These find-
⫺9.5 to ⫺8.7 kb 5⬘ of the nodal gene (65). In addition, ectopic
ings suggest that Notch signaling controls cardiac cell fate
activation of Notch signaling in early zebrafish embryos results
downstream of heart field specification.
in Nodal and Pitx2 expression in the right LPM and ultimately
Endocardial precursors are identifiable at the cardiac cres-
left-right patterning defects (103).
cent stage as a population of Flk1⫹/TAL1⫹-positive cells
distributed throughout the cardiac crescent (18). Lineage anal- Endothelial-to-Mesenchymal Transformation
ysis of Mesp1-derived mesoderm reveals that the endocardium
is derived from the same mesoderm as the myocardium, Beginning at E9.0 in the mouse, localized swellings of
thereby demonstrating that the endocardium and myocardium the cardiac jelly appear in the atrioventricular (AV) canal and
are of the same origin (111, 112). During subsequent folding of cardiac outflow tract (OFT), forming the superior and inferior
AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org
Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
NOTCH AND CARDIAC DEVELOPMENT C5
cardiac cushions. Cardiac cushions are acellular swellings of during early development (36). Notch1 activity is found to be
extracellular matrix protein secreted by the myocardium (22). highest in the trabecular endocardium, where it controls the
At E9.5, endocardial cells of the AV canal and OFT are expression of NRG1 and BMP10, which are responsible for
activated by signals emanating from the myocardium and by myocardial differentiation and proliferation, respectively (36).
interendocardial signaling pathways to undergo endothelial-to- In addition, constitutive expression of Notch1ICD in Mesp1-
mesenchymal transition. Endothelial-to-mesenchymal transi- derived cells results in abnormal heart morphogenesis (131). In
tion is a specific form of EMT required for mesenchymal cell Mesp1-derived cells, the Notch pathway is activated in the
formation from endocardial cells during cardiac cushion de- cardiogenic mesoderm and all subsequent cells of the endocar-
velopment. It is a critical process whereby endocardial cells dial and myocardial lineage (111, 112, 131). The most severe
undergo phenotypic and morphological alterations, resulting in defects induced by ectopic Notch activation in this model were
loss of apical-basolateral polarity and disruption of intercellu- defects in myocardial trabecular development, the appearance
lar junctions, and acquire the ability to degrade the basement of a cell mass in the AV canal, and right-shifted interventric-
membrane and migrate away from the confines of the endo- ular septum (131). Myocardial trabeculae were observed in the
thelial sheet to invade the underlying cardiac jelly. As devel-
compact myocardial layer and the AV canal region, two
opment progresses, the mesenchymal cells undergo prolifera-
regions where trabecular myocardium is usually absent (131).
tion, resulting in fusion of the cardiac cushions within the
In addition, in vitro experiments have clearly demonstrated that
lumen of the heart tube forming the initial septa. Further
remodeling of the cardiac cushion results in the formation of activation of the Notch pathway in endothelial cells results in
thin protruding leaflets comprised of endocardial cells and EMT (91). This study also showed that Notch-mediated EMT
extracellular matrix protein (ECM) that go on to develop into was cell autonomous and independent of TGF-␤ signaling,
the heart valves. In the AV canal, EMT-derived cells are the supporting the importance of the role of Notch signaling during
sole contributor to the mitral and tricuspid valves, while in the EMT in heart development (91).
OFT, EMT- and neural crest-derived cells contribute to the The Notch target gene Hey2 is initially expressed in
aortic and pulmonary valves. ventricle precursors of the heart tube and later becomes
There are ongoing efforts to elucidate the transcriptional restricted to the compact myocardial layer of the ventricles.
networks operating during cardiac EMT (http://www. Hey2 is also highly expressed at the onset of EMT in the AV
mouseatlas.org/morgen/content). However, both loss- and canal endocardium (128). However, expression of Hey2 is
gain-of-function studies have revealed a critical role of the not sustained in the mesenchymal cells of the AV canal,
Notch pathway in regulating EMT during heart develop- suggesting Notch activity is required for initiation of EMT
ment. In the mouse, Notch1, Notch4, and Dll4 are expressed but not for maintaining a mesenchymal phenotype. Early in
in the AV canal and OFT endocardium at the onset of EMT. heart development, Hey1 is expressed in the lateral aspect of
Furthermore, both Notch1-deficient and CSL-deficient em- the cardiac crescent, which gives rise to the sinus venosus
bryos have significantly reduced EMT in the AV canal, as and atria. Later, Hey1 becomes restricted to the atrial
determined using an ex vivo AV canal explant assay, which myocardium. HeyL is not expressed during heart develop-
provides a measure of the degree of EMT taking place (125). ment, and levels of Hey1 and Hey2 in the heart gradually
Analysis of Notch1-deficient and CSL-deficient embryonic decline after birth. The mechanism by which Hey1 and
hearts reveals reduced expression of TGF-␤2 and its recep- Hey2 become differentially expressed remains to be re-
tors (125), which are critical initiators of EMT during heart solved, since both genes are direct targets of Notch activa-
development. However, by use of the AV canal explant tion. In the atrium, there is high expression of Jagged1,
assay, exogenous TGF-␤2 or TGF-␤3 was not sufficient to which could potentially regulate Hey1 expression (78, 126).
rescue the defect in EMT in Notch1-deficient and CSL- However, Notch receptors or ligands are not observed in
deficient embryos (125). These results suggest that, al- the compact myocardial layer, suggesting that, in this con-
though Notch signaling is important for expression of text, Hey2 may be regulated by a pathway other than Notch
TGF-␤2 pathway components, it is not clear whether Notch (126).
is downstream of, upstream of, or synergistically required
Several studies have also revealed a critical role of Hey2
for TGF-␤-dependent EMT during heart development A
during heart development. The phenotypes of Hey2-null
caveat of interpreting data from the Notch1 and CSL mutant
embryos is the significant delay in embryonic development mice are variable, but these mice have high mortality in the
observed in these mutants. The delay is particularly apparent first weeks after birth because of cardiovascular defects,
in the heart, and therefore EMT might be significantly including ventricular septal defects, pulmonic stenosis, AV
delayed but not impaired. In fact, in AV canal explant canal valve irregularities, and cardiac hypertrophy (17, 25,
assays, a very small population of ␣-smooth muscle actin 63). Gene targeting studies have revealed that Hey1 or HeyL
(SMA)-positive cells is detectable, suggesting that Notch1- alone is not required for heart development. However, Hey1
deficient and CSL-deficient embryos are capable of initiat- and Hey2 double-deficient embryos die at E9.5 because of
ing cardiac EMT at a reduced level (125). Supporting a role severe heart defects, including missing ventricular trabecu-
for the Notch pathway in EMT of the AV canal, it was lae and lack of arterial differentiation (25). In addition,
shown that zebrafish injected with constitutively active combined loss of both Hey1 and HeyL results in a signifi-
Notch1 developed hypercellular AV canals and enlarged AV cant reduction in endocardial EMT, similar to the Notch1-
valves as a result of increased EMT (125). or CSL-deficient embryos (26). Known expression patterns
Notch1-deficient embryos also display defects in ventricular of Notch pathway component have been summarized in
trabeculation because of a decrease in myocardial proliferation Table 1.
AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org
Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
C6 NOTCH AND CARDIAC DEVELOPMENT

Table 1. Expression of Notch pathway components during murine heart development


Notch Pathway Component Expression Pattern

Notch1 Expressed in the cardiac crescent (E7.5). At E8.0 to E11.5 expression is limited to the entire endocardium and highly expressed
in the AV canal and outflow tract endocardium (124, 132).
Notch2 Expressed in the AV canal endocardium (E12.5) and the outflow tract (E11.5 and 14.5). Expressed in atrial and ventricular
myocardium (E13.5) (76, 80, 82).
Notch3 Expressed in the cardiac crescent (E7.5) but not detected after heart tube formation (E8.0) (132).
Notch4 Expressed in the endocardium (E10.5) (90).
Jagged1 Expressed in the AV canal and outflow tract endocardium and atrial myocardium (E10.5–E12.5) (77).
Jagged2 Expression has not been analyzed
DI11 Not expressed in the heart (4)
DI13 Expression has not been analyzed
DI14 Expressed in the cardiac crescent (E8.0) and the endocardium from E8.5 onward. Expression is further restricted to the
ventricular endocardium after E11.5 (3, 19).
Hey1 Expressed in the lateral portion of the heart tube (E8.5) and the endocardium and septum transversum (E9.5). Expressed
exclusively in the atrial myocardium at E10.5 (71, 124).
Hey2 Expressed in the anterior portion of the heart tube (E8.5) and the AV canal and OFT endocardium (E11.0). Highly expressed in
the subcompact ventricular myocardium (E10.5) (12, 71).
HeyL It has been reported that HeyL is not expressed in the embryonic heart (72), but combined targeting of Hey1 and HeyL results
in cardiovascular defects related to EMT (26).
AV, atrioventricular; E, embryonic day; OFT, outflow track; EMT, epithelial-to-mesenchymal transition.

ARTERIAL VENOUS SPECIFICATION disorganized, and the dorsal aorta and cardinal vein are formed
but are significantly reduced in size (67). These results suggest
Early Vascular Progenitors
that Notch1 is not required for angioblast differentiation but is
Vasculogenesis is the de novo development of endothelial required for remodeling of the vascular plexus. Notch4-defi-
cells from mesodermal progenitors called angioblasts, while cient mice are viable and fertile; however, Notch1/Notch4
angiogenesis is the formation of new vessels from preexisting double-deficient mice have vascular defects similar to but more
vessels. All embryonic mesoderm, with the exception of the severe than Notch1-deficient animals (67). This suggests a
prechordal plate, has angioblast potential (89). Angioblast partial redundancy between Notch1 and Notch4 in the devel-
differentiation in situ forms a primary vascular plexus that oping vasculature. Notch2-deficient mice die at E11.5 because
undergoes extensive remodeling into arteries, veins, and cap- of a variety of defects in the cardiovascular and renal systems
illaries. The three types of vascular structures are morpholog- (38, 82). Notch3-deficient mice, in contrast, are viable without
ically, functionally, and molecularly distinct. At the molecular any obvious defects in cardiovascular development (68). Vas-
level, artery-specific factors include Ephrin-B2, CD44, Neuro- cular remodeling defects are also seen in Dll4- and Jagged1-
pilin-1, Hey1, and Hey2, while vein-specific factors include deficient mice, resulting in death at E9.5 and E11.5, respec-
EphB4, COUP-TFII, and Lefty1 (11, 25, 135). tively (32, 66, 134). Expression of constitutively active Notch4
driven by the flk1 promoter in endothelial cells during devel-
Expression of Notch Pathway Components opment results in yolk sac remodeling defects and disorganized
Notch pathway components are initially expressed through- embryonic vessel development.
out the developing cardiovascular system but become mainly
restricted to cells that will acquire an arterial fate later in Molecular Patterning of Arterial Venous Specification
development. At E9.5, Notch1, Notch4, and Dll4 are expressed Arterial and venous endothelial progenitors have been
throughout the primitive vascular plexus. By E13.5, Notch1, shown to be specified before vessel assembly or blood flow
Notch2, Notch4, Dll4, Jagged1, and Jagged2 expression has (41). Recent studies have revealed a signaling cascade for
become mainly restricted to the arterial endothelium (126). arterial specification that involves sequential activation of
Notch1 and Dll4 are also expressed in the capillary endothe- Sonic Hedgehog (Shh), vascular endothelial growth factor
lium. In contrast, artery smooth muscle cells express low levels (VEGF), and Notch (71). In zebrafish, Shh deficiency results in
of Notch1 and high levels of Notch3 and Jagged1 (126). a loss of arterial specification and decreased VEGF expression
Notch4 is also expressed in the cardinal vein, human umbilical (71). In contrast, Shh activation in zebrafish results in upregu-
endothelial cells, and capillary endothelium. Expression pat- lation of VEGF and ectopic arterial specification (as deter-
terns of Notch receptors and ligands in the vasculature have mined by expression of EphrinB2) of vessels otherwise des-
been extensively reviewed in Ref. 49. When injured, arteries tined to become veins (71). Activation of the Notch pathway in
induce expression of various Notch receptors and ligands (75). mature vessel endothelium, via Tie2-driven inducible expres-
Defects in Vascular Remodeling sion of Notch4ICD, results in expression of EphrinB2 and
increased smooth muscle layers resulting in arterialization
Notch1-deficient mice die before E10.5 because of severe of the venous vessels (8). In this model, overexpression of
defects in cardiovascular development (122). In Notch1-defi- Notch4ICD results in death of adult animals within weeks of
cient embryos, the yolk sac and cerebral primary vascular expression. Interestingly, the effects of Notch4ICD expression
plexus are formed, but subsequent remodeling of the vascular are reversible on reversal of Notch4ICD expression (8). In
plexus is defective. In addition, intersomitic vessels are highly mice, Hey1/ Hey2 or Notch1/Notch4 double-deficient and
AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org
Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
NOTCH AND CARDIAC DEVELOPMENT C7
Dll4-deficient embryos display defects in arterial specification above, Notch pathway components are highly expressed in
to various degrees (25, 67). Studies in zebrafish have provided arterial endothelial cells, which correlates with the importance
significant insight into the role of the Notch pathway in of the Notch pathway in regulating endothelial function in the
arterial-venous specification. Enforced expression of Notch5, aortic valve. With the use of an in vitro system, it was
which is similar to mammalian Notch3 by sequence, in the demonstrated that Notch1, Hey1, and Hey2 repress the func-
endothelium results in reduced venous expression of flt-4, a tion of the transcription factor RUNX2 (33). RUNX2 has been
venous marker (70). Conversely, blocking Notch signaling in linked to valvular calcification in both rabbit and mouse, where
the dorsal aorta, using dominant-negative CSL, results in loss it regulates expression of several osteogenic genes, such as
of arterial expression of EphrinB2 (70). osteopontin and osteocalcin (20). It was further suggested that
Notch target genes have also been shown to play a role in Notch signaling via upregulation of Hey1 and Hey2 results in
arterial-venous specification. A mutant of gridlock, the only Hey1 or Hey2 physically interacting with RUNX2, thereby
Hey family member expressed in the zebrafish vasculature, inhibiting RUNX2 function (33). It is not known whether
shows defects in aortic assembly resembling coarctation of the Notch1 mutations found in humans result in lower Hey1 and
aorta (137). Antisense-directed knockdown of gridlock sup- Hey2 expression in the aortic valve, thereby allowing higher
presses arterial marker expression and expands contiguous RUNX2 activity, subsequent expression of osteogenic genes,
regions of the vein during embryonic vascular development and calcification of the aortic valve.
(136). However, overexpression of gridlock acts to repress
venous fate rather than promote arterial specification (136). Alagille Syndrome
The role of gridlock is controversial, as others have suggested
Mutations in the Jagged1 locus are associated with 94% of
that Notch mediates arterial specification independently of
patients with Alagille syndrome (AGS) (130). In addition, muta-
gridlock (70). In addition, VEGF stimulation was shown to
tions in the Notch2 locus have been identified in patients with
upregulate Notch1 and Dll4 expression exclusively in arterial
Jagged1-independent AGS (84). AGS is an autosomal dominant
endothelial cells. VEGF treatment of Shh-deficient but not
disorder most commonly associated with neonatal jaundice and
Notch-deficient embryos is capable of rescuing arterial
impaired development of intrahepatic bile ducts, with additional
EphrinB2 expression.
abnormalities of the eye, heart, kidney, and skeleton with variable
In venous vessels, expression of the orphan nuclear receptor
penetrance. The most common cardiovascular defect is peripheral
COUP-TFII was shown to regulate the expression of Notch1
pulmonic stenosis, and 13% of AGS patients have Tetralogy of
and Jagged1 in arterial-venous specification (135). Loss of
Fallot (58, 64, 85). Tetralogy of Fallot is a condition characterized
COUP-TFII resulted in ectopic venous expression of Notch1
by ventricular septal defect, overriding aorta, infundibular pulmo-
and Jagged1, resulting in arterialization of the venous vessels
nary stenosis, and often right ventricular hypertrophy. In less
(135). In contrast, ectopic expression of COUP-TFII in the
frequent cases, AGS has been associated with other defects of the
endothelium, via the Tie2 promoter, resulted in reduced
cardiac cushion (23, 85). The cardiac AGS phenotype is consis-
Jagged1 expression and arterial expression of EphB4, a venous
tent with the expression pattern of Jagged1 and Notch receptors in
marker. Together these findings suggest that COUP-TFII is
the cardiovascular system. Analyses of mutations in the Jagged1
upstream of Notch signaling in arterial-venous specification,
locus have revealed in some cases complete loss of the Jagged1
and that Notch signaling represses venous differentiation. Thus
locus and in other cases inactivating mutations that lead to a
the current paradigm depicts a signaling cascade for arterial
misexpressed or truncated Jagged1 protein (120). However, het-
differentiation where Shh is upstream of VEGF and VEGF is
erozygous mutations in Jagged1 or Notch2 do not reproduce the
upstream of Notch signaling.
AGS phenotype in mice (38, 134). Jagged1 heterozygote mice
MUTATIONS OF NOTCH PATHWAY COMPONENTS IN display eye defects, while Jagged1-null mice die at E10 because
HUMAN DISEASE of vascular defects (134). Notch2 haploinsufficiency in mice
results in kidney defects and myocardial hypoplasia, but the
Aortic Valve Disease phenotype does not resemble findings of cardiovascular and
kidney defects in patients with AGS (38, 81). However, mice
In humans, mutations in the Notch1 locus result in a spec-
doubly heterozygous for a Jagged1-null and Notch2-hypomorphic
trum of heart defects (33). The most prevalent malformations
allele develop jaundice and impaired development of intrahepatic
are bicuspid aortic valve disease and calcification of the aortic
bile ducts with associated abnormalities of the eye, heart, and
valve (33). Calcification of the aortic valve is the third leading
kidney, reproducing an AGS phenotype (83). The reason that
cause of heart disease in adults, while the presence of the
human Jagged1 haploinsufficiency results in AGS while in mice
bicuspid aortic valve is present in 1–2% of the population (43).
there is an additional requirement for Notch2 insufficiency is
Mutations in the Notch1 locus result in a premature stop codon
unknown. Possible explanations include a higher basal expression
in the extracellular domain of Notch1, likely resulting in rapid
of Jagged1 in mice, compensating expression of a Notch ligand,
degradation of mRNA by the nonsense-mediated mRNA decay
or increased avidity of Notch receptor-ligand interaction in the
pathway (33). The resulting defects may be due to halploin-
mouse cardiovascular system.
sufficiency, or alternatively, the premature stop codon could
result in the expression of a truncated Notch1 protein that could Cerebral Autosomal Dominant Arteriopathy with Subcortical
function as a dominant-negative mutant. These hypotheses Infarcts and Leukoencephalopathy
have yet to be tested experimentally. The mechanism by which
human Notch1 mutations effect aortic valve calcification is Cerebral autosomal dominant arteriopathy with subcortical
poorly understood. However, calcification of the aortic valve is infarcts and leukoencephalopathy (CADASIL) is an autosomal
thought to be due to endothelial dysfunction. As discussed dominant disorder associated with defects in arterial vascular
AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org
Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
C8 NOTCH AND CARDIAC DEVELOPMENT

homeostasis, resulting in increased incidence of stroke, mi- signaling in the vasculature. However, a thorough understand-
graine headaches, and mood disturbances (10, 55). CADASIL ing of the downstream targets of Hey proteins and the function
results from a progressive loss of the arterial vascular smooth of these downstream targets remains to be elucidated. Early
muscle cells and increased vascular fibrosis, leading to narrow- studies on functional changes triggered by Notch in vascular
ing of the lumen of small and medium arteries (10). CADASIL cells have been performed, but much remains to be investi-
is histologically characterized by accumulation of granular gated in this area also. Notch signaling appears to integrate
osmophilic deposits (GOMs) in the vessel media (80, 108). with feedback loops of the VEGF, platelet-derived growth
Arterial vascular defects seen in CADASIL patients can be factor, Wnt, and BMP pathways, but the molecular mecha-
systemically found; however, for unknown reasons, vascular nisms of how this occurs are still under investigation. Undoubt-
complications are only found in the brain (110). edly, there will be further examples of vascular diseases that
Missense point mutations in the first five EGF repeats in the are influenced or perhaps even caused by aberrant signaling in
Notch3 locus resulting in an odd number of cysteine residues Notch pathway genes as investigators place greater emphasis in
in the EGF repeat regions are found in 95% of CADASIL cases these unmined areas of research.
(16, 54, 56). The change in the number of cysteine residues
from six to five or seven in the EGF repeats is thought to result GRANTS
in a conformational change of the EGF repeats, leading to Work in the laboratory of A. Karsan is supported by grants from the Heart
accumulation of the ectodomain of the Notch3 receptor. As and Stroke Foundation of British Columbia and the Yukon, the Canadian
discussed above, Notch receptors are expressed as a het- Institutes of Health Research, the Canadian Cancer Society, the Stem Cell
Network, Genome Canada, and Genome British Columbia. K. Niessen was
erodimer on the plasma membrane. In patients with CADASIL, supported by a Doctoral Research Award from the Michael Smith Foundation
there is a specific accumulation of the 210-kDa ectodomain of for Health Research. A. Karsan is a Senior Scholar of the Michael Smith
Notch3 in the vascular smooth muscle plasma membrane in Foundation for Health Research.
close association with but not within the GOMs (53).
The expression of Notch3 is restricted to the vascular REFERENCES
smooth muscle cells, which correlates with the smooth muscle 1. Ascano JM, Beverly LJ, Capobianco AJ. The C-terminal PDZ-ligand
defects seen in CADASIL. Mutant mice selectively expressing of JAGGED1 is essential for cellular transformation. J Biol Chem 278:
a similar mutation in the EGF repeats of Notch3 (Arg90Cys) in 8771– 8779, 2003.
2. Beatus P, Lundkvist J, Oberg C, Pedersen K, Lendahl U. The origin
smooth muscle cells, using the SM22␣ promoter, recapitulate of the ankyrin repeat region in Notch intracellular domains is critical for
the CADASIL phenotype (109). In Notch3-Arg90Cys trans- regulation of HES promoter activity. Mech Dev 104: 3–20, 2001.
genic mice, the CADASIL phenotype takes 10 –12 mo to 3. Benedito R, Duarte A. Expression of Dll4 during mouse embryogenesis
develop (109). Mutated Notch3 receptors associated with suggests multiple developmental roles. Gene Expr Patterns 5: 750 –755,
CADASIL are expressed on the cell surface and capable of 2005.
4. Bettenhausen B, Hrabe de Angelis M, Simon D, Guenet JL, Gossler
binding Dll1, suggesting that the phenotype of CADASIL A. Transient and restricted expression during mouse embryogenesis of
patients is downstream of Notch3 ligand binding (40). One Dll1, a murine gene closely related to Drosophila Delta. Development
possible reason for the defect seen in CADASIL is that the 121: 2407–2418, 1995.
impaired clearance of the Notch3 ectodomain results in a 5. Blaumueller CM, Qi H, Zagouras P, Artavanis-Tsakonas S. Intracel-
lular cleavage of Notch leads to a heterodimeric receptor on the plasma
dominant-negative effect by sequestering ligands from the membrane. Cell 90: 281–291, 1997.
functional Notch3 receptors. In smooth muscle cells, Notch3 6. Brand T. Heart development: molecular insights into cardiac specifica-
has been shown to activate survival pathways which, if blocked tion and early morphogenesis. Dev Biol 258: 1–19, 2003.
by accumulation of the Notch3 ectodomain, could result in loss 7. Brou C, Logeat F, Gupta N, Bessia C, LeBail O, Doedens JR,
of smooth muscle cells observed in CADASIL patients (129). Cumano A, Roux P, Black RA, Israel A. A novel proteolytic cleavage
involved in Notch signaling: the role of the disintegrin-metalloprotease
However, as discussed above, Notch3-deficient mice are viable TACE. Mol Cell 5: 207–216, 2000.
and fertile with no observed defects in cardiovascular devel- 8. Carlson TR, Yan Y, Wu X, Lam MT, Tang GL, Beverly LJ, Messina
opment (68). Thus, if the mutated Notch3 acts as a dominant- LM, Capobianco AJ, Werb Z, Wang R. Endothelial expression of
negative, it suggests that ligand sequestration may also be constitutively active Notch4 elicits reversible arteriovenous malforma-
tions in adult mice. Proc Natl Acad Sci USA 102: 9884 –9889, 2005.
blocking activation of other Notch receptors expressed on 9. Cayouette M, Raff M. Asymmetric segregation of Numb: a mechanism
vascular smooth muscle cells, which may also be required for neural specification from Drosophila to mammals. Nat Neurosci 5:
for viability. A recent manuscript by Monet et al. (85a) de- 1265–1269, 2002.
monstrated that the Notch3 (Arg90Cys) mutation found in 10. Chabriat H, Vahedi K, Iba-Zizen MT, Joutel A, Nibbio A, Nagy TG,
CADASIL patients is still capable of activating the Notch Krebs MO, Julien J, Dubois B, Ducrocq X, et al. Clinical spectrum of
CADASIL: a study of 7 families. Cerebral autosomal dominant arteri-
pathway. In addition this manuscript demonstrates that even opathy with subcortical infarcts and leukoencephalopathy. Lancet 346:
when the Notch3-extracellular domain accumulates on the 934 –939, 1995.
SMC plasma membrane activation of the Notch pathway is 11. Chi JT, Chang HY, Haraldsen G, Jahnsen FL, Troyanskaya OG,
unaffected. These results suggest that accumulation of the Chang DS, Wang Z, Rockson SG, van de Rijn M, Botstein D, Brown
PO. Endothelial cell diversity revealed by global expression profiling.
Notch3-extracellular domain and the pathogenesis of CADA- Proc Natl Acad Sci USA 100: 10623–10628, 2003.
SIL are not a result of the Notch3-extracellular domain acting 12. Chin MT, Maemura K, Fukumoto S, Jain MK, Layne MD, Wa-
as a dominant-negative for the Notch pathway. tanabe M, Hsieh CM, Lee ME. Cardiovascular basic helix loop helix
In conclusion, it has become clear over the past few years factor 1, a novel transcriptional repressor expressed preferentially in the
that Notch signaling plays a crucial role in cardiovascular developing and adult cardiovascular system. J Biol Chem 275: 6381–
6387, 2000.
development and in human vascular disease. In addition, 13. Davis RL, Turner DL. Vertebrate hairy and Enhancer of split related
emerging evidence indicates that members of the Hey family of proteins: transcriptional repressors regulating cellular differentiation and
bHLH proteins are critical downstream mediators of Notch embryonic patterning. Oncogene 20: 8342– 8357, 2001.

AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
NOTCH AND CARDIAC DEVELOPMENT C9
14. de la Cruz MV, Sanchez Gomez C, Arteaga MM, Arguello C. JL. Notch signaling is essential for ventricular chamber development.
Experimental study of the development of the truncus and the conus in Dev Cell 12: 415– 429, 2007.
the chick embryo. J Anat 123: 661– 686, 1977. 37. Haltiwanger RS. Regulation of signal transduction pathways in devel-
15. De Strooper B, Annaert W, Cupers P, Saftig P, Craessaerts K, opment by glycosylation. Curr Opin Struct Biol 12: 593–598, 2002.
Mumm JS, Schroeter EH, Schrijvers V, Wolfe MS, Ray WJ, Goate 38. Hamada Y, Kadokawa Y, Okabe M, Ikawa M, Coleman JR, Tsuji-
A, Kopan R. A presenilin-1-dependent gamma-secretase-like protease moto Y. Mutation in ankyrin repeats of the mouse Notch2 gene induces
mediates release of Notch intracellular domain. Nature 398: 518 –522, early embryonic lethality. Development 126: 3415–3424, 1999.
1999. 39. Hansson EM, Lendahl U, Chapman G. Notch signaling in develop-
16. Dichgans M, Ludwig H, Muller-Hocker J, Messerschmidt A, Gasser ment and disease. Semin Cancer Biol 14: 320 –328, 2004.
T. Small in-frame deletions and missense mutations in CADASIL: 3D 40. Haritunians T, Boulter J, Hicks C, Buhrman J, DiSibio G, Shawber
models predict misfolding of Notch3 EGF-like repeat domains. Eur J C, Weinmaster G, Nofziger D, Schanen C. CADASIL Notch3 mutant
Hum Genet 8: 280 –285, 2000. proteins localize to the cell surface and bind ligand. Circ Res 90:
17. Donovan J, Kordylewska A, Jan YN, Utset MF. Tetralogy of fallot and 506 –508, 2002.
other congenital heart defects in Hey2 mutant mice. Curr Biol 12: 41. Herzog Y, Guttmann-Raviv N, Neufeld G. Segregation of arterial and
1605–1610, 2002. venous markers in subpopulations of blood islands before vessel forma-
18. Drake CJ, Fleming PA. Vasculogenesis in the day 6.5 to 9.5 mouse tion. Dev Dyn 232: 1047–1055, 2005.
embryo. Blood 95: 1671–1679, 2000. 42. Hicks C, Johnston SH, diSibio G, Collazo A, Vogt TF, Weinmaster
19. Duarte A, Hirashima M, Benedito R, Trindade A, Diniz P, Bekman G. Fringe differentially modulates Jagged1 and Delta1 signalling through
E, Costa L, Henrique D, Rossant J. Dosage-sensitive requirement for Notch1 and Notch2. Nat Cell Biol 2: 515–520, 2000.
mouse Dll4 in artery development. Genes Dev 18: 2474 –2478, 2004. 43. Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am
20. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: a Coll Cardiol 39: 1890 –1900, 2002.
transcriptional activator of osteoblast differentiation. Cell 89: 747–754, 44. Hori K, Fostier M, Ito M, Fuwa TJ, Go MJ, Okano H, Baron M,
1997. Matsuno K. Drosophila deltex mediates suppressor of Hairless-indepen-
21. Ehebauer MT, Chirgadze DY, Hayward P, Martinez Arias A, Blun- dent and late-endosomal activation of Notch signaling. Development 131:
dell TL. High-resolution crystal structure of the human Notch 1 ankyrin 5527–5537, 2004.
domain. Biochem J 392: 13–20, 2005. 45. Hsieh JJ, Zhou S, Chen L, Young DB, Hayward SD. CIR, a corepres-
22. Eisenberg LM, Markwald RR. Molecular regulation of atrioventricular sor linking the DNA binding factor CBF1 to the histone deacetylase
valvuloseptal morphogenesis. Circ Res 77: 1– 6, 1995. complex. Proc Natl Acad Sci USA 96: 23–28, 1999.
23. Eldadah ZA, Hamosh A, Biery NJ, Montgomery RA, Duke M, Elkins 46. Hu Y, Ye Y, Fortini ME. Nicastrin is required for gamma-secretase
R, Dietz HC. Familial Tetralogy of Fallot caused by mutation in the cleavage of the Drosophila Notch receptor. Dev Cell 2: 69 –78, 2002.
jagged1 gene. Hum Mol Genet 10: 163–169, 2001. 47. Inagaki T, Garcia-Martinez V, Schoenwolf GC. Regulative ability of
24. Espinosa L, Ingles-Esteve J, Aguilera C, Bigas A. Phosphorylation by the prospective cardiogenic and vasculogenic areas of the primitive
glycogen synthase kinase-3 beta down-regulates Notch activity, a link for
streak during avian gastrulation. Dev Dyn 197: 57– 68, 1993.
Notch and Wnt pathways. J Biol Chem 278: 32227–32235, 2003.
48. Iso T, Chung G, Hamamori Y, Kedes L. HERP1 is a cell type-specific
25. Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M. The
primary target of Notch. J Biol Chem 277: 6598 – 6607, 2002.
Notch target genes Hey1 and Hey2 are required for embryonic vascular
49. Iso T, Hamamori Y, Kedes L. Notch signaling in vascular development.
development. Genes Dev 18: 901–911, 2004.
Arterioscler Thromb Vasc Biol 23: 543–553, 2003.
26. Fischer A, Steidl C, Wagner TU, Lang E, Jakob PM, Friedl P,
50. Iso T, Maeno T, Oike Y, Yamazaki M, Doi H, Arai M, Kurabayashi
Knobeloch KP, Gessler M. Combined Loss of Hey1 and HeyL Causes
M. Dll4-selective Notch signaling induces ephrinB2 gene expression in
Congenital Heart Defects Because of Impaired Epithelial to Mesenchy-
endothelial cells. Biochem Biophys Res Commun 341: 708 –714, 2006.
mal Transition. Circ Res 2007.
51. Izon DJ, Aster JC, He Y, Weng A, Karnell FG, Patriub V, Xu L,
27. Fleming RJ. Structural conservation of Notch receptors and ligands.
Semin Cell Dev Biol 9: 599 – 607, 1998. Bakkour S, Rodriguez C, Allman D, Pear WS. Deltex1 redirects
28. Foltz DR, Santiago MC, Berechid BE, Nye JS. Glycogen synthase lymphoid progenitors to the B cell lineage by antagonizing Notch1.
kinase-3beta modulates notch signaling and stability. Curr Biol 12: Immunity 16: 231–243, 2002.
1006 –1011, 2002. 52. Jehn BM, Dittert I, Beyer S, von der Mark K, Bielke W. c-Cbl
29. Francis R, McGrath G, Zhang J, Ruddy DA, Sym M, Apfeld J, Nicoll binding and ubiquitin-dependent lysosomal degradation of membrane-
M, Maxwell M, Hai B, Ellis MC, Parks AL, Xu W, Li J, Gurney M, associated Notch1. J Biol Chem 277: 8033– 8040, 2002.
Myers RL, Himes CS, Hiebsch R, Ruble C, Nye JS, Curtis D. aph-1 53. Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N,
and pen-2 are required for Notch pathway signaling, gamma-secretase Piga N, Chapon F, Godfrain C, Tournier-Lasserve E. The ectodomain
cleavage of betaAPP, and presenilin protein accumulation. Dev Cell 3: of the Notch3 receptor accumulates within the cerebrovasculature of
85–97, 2002. CADASIL patients. J Clin Invest 105: 597– 605, 2000.
30. Fryer CJ, Lamar E, Turbachova I, Kintner C, Jones KA. Mastermind 54. Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P,
mediates chromatin-specific transcription and turnover of the Notch Alamowitch S, Domenga V, Cecillion M, Marechal E, Maciazek J,
enhancer complex. Genes Dev 16: 1397–1411, 2002. Vayssiere C, Cruaud C, Cabanis EA, Ruchoux MM, Weissenbach J,
31. Fryer CJ, White JB, Jones KA. Mastermind recruits CycC:CDK8 to Bach JF, Bousser MG, Tournier-Lasserve E. Notch3 mutations in
phosphorylate the Notch ICD and coordinate activation with turnover. CADASIL, a hereditary adult-onset condition causing stroke and demen-
Mol Cell 16: 509 –520, 2004. tia. Nature 383: 707–710, 1996.
32. Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, Valenzuela 55. Joutel A, Tournier-Lasserve E. Notch signalling pathway and human
DM, Murphy AJ, Adams NC, Lin HC, Holash J, Thurston G, diseases. Semin Cell Dev Biol 9: 619 – 625, 1998.
Yancopoulos GD. Haploinsufficiency of delta-like 4 ligand results in 56. Joutel A, Vahedi K, Corpechot C, Troesch A, Chabriat H, Vayssiere
embryonic lethality due to major defects in arterial and vascular devel- C, Cruaud C, Maciazek J, Weissenbach J, Bousser MG, Bach JF,
opment. Proc Natl Acad Sci USA 101: 15949 –15954, 2004. Tournier-Lasserve E. Strong clustering and stereotyped nature of
33. Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King Notch3 mutations in CADASIL patients. Lancet 350: 1511–1515, 1997.
IN, Grossfeld PD, Srivastava D. Mutations in NOTCH1 cause aortic 57. Kadesch T. Notch signaling: a dance of proteins changing partners. Exp
valve disease. Nature 437: 270 –274, 2005. Cell Res 260: 1– 8, 2000.
34. Ge W, Martinowich K, Wu X, He F, Miyamoto A, Fan G, Weinmas- 58. Kamath BM, Spinner NB, Emerick KM, Chudley AE, Booth C,
ter G, Sun YE. Notch signaling promotes astrogliogenesis via direct Piccoli DA, Krantz ID. Vascular anomalies in Alagille syndrome: a
CSL-mediated glial gene activation. J Neurosci Res 69: 848 – 860, 2002. significant cause of morbidity and mortality. Circulation 109: 1354 –
35. Greenwald I, Seydoux G. Analysis of gain-of-function mutations of the 1358, 2004.
lin-12 gene of Caenorhabditis elegans. Nature 346: 197–199, 1990. 59. Kao HY, Ordentlich P, Koyano-Nakagawa N, Tang Z, Downes M,
36. Grego-Bessa J, Luna-Zurita L, Del Monte G, Bolos V, Melgar P, Kintner CR, Evans RM, Kadesch T. A histone deacetylase corepressor
Arandilla A, Garratt AN, Zang H, Mukouyama YS, Chen H, Shou complex regulates the Notch signal transduction pathway. Genes Dev 12:
W, Ballestar E, Esteller M, Rojas A, Perez-Pomares JM, de la Pompa 2269 –2277, 1998.

AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
C10 NOTCH AND CARDIAC DEVELOPMENT

60. Kelly RG, Buckingham ME. The anterior heart-forming field: voyage to 81. McCright B, Gao X, Shen L, Lozier J, Lan Y, Maguire M, Herzlinger
the arterial pole of the heart. Trends Genet 18: 210 –216, 2002. D, Weinmaster G, Jiang R, Gridley T. Defects in development of the
61. Kitajima S, Takagi A, Inoue T, Saga Y. MesP1 and MesP2 are kidney, heart and eye vasculature in mice homozygous for a hypomor-
essential for the development of cardiac mesoderm. Development 127: phic Notch2 mutation. Development 128: 491–502, 2001.
3215–3226, 2000. 82. McCright B, Lozier J, Gridley T. Generation of new Notch2 mutant
62. Klinakis A, Szabolcs M, Politi K, Kiaris H, Artavanis-Tsakonas S, alleles. Genesis 44: 29 –33, 2006.
Efstratiadis A. Myc is a Notch1 transcriptional target and a requisite for 83. McCright B, Lozier J, Gridley T. A mouse model of Alagille syn-
Notch1-induced mammary tumorigenesis in mice. Proc Natl Acad Sci drome: Notch2 as a genetic modifier of Jag1 haploinsufficiency. Devel-
USA 103: 9262–9267, 2006. opment 129: 1075–1082, 2002.
63. Kokubo H, Miyagawa-Tomita S, Tomimatsu H, Nakashima Y, Na- 84. McDaniell R, Warthen DM, Sanchez-Lara PA, Pai A, Krantz ID,
kazawa M, Saga Y, Johnson RL. Targeted disruption of hesr2 results in Piccoli DA, Spinner NB. NOTCH2 mutations cause Alagille syndrome,
atrioventricular valve anomalies that lead to heart dysfunction. Circ Res a heterogeneous disorder of the Notch signaling pathway. Am J Hum
95: 540 –547, 2004. Genet 79: 169 –173, 2006.
64. Krantz ID, Smith R, Colliton RP, Tinkel H, Zackai EH, Piccoli DA, 85. McElhinney DB, Krantz ID, Bason L, Piccoli DA, Emerick KM,
Goldmuntz E, Spinner NB. Jagged1 mutations in patients ascertained Spinner NB, Goldmuntz E. Analysis of cardiovascular phenotype and
with isolated congenital heart defects. Am J Med Genet 84: 56 – 60, 1999. genotype-phenotype correlation in individuals with a JAG1 mutation
65. Krebs LT, Iwai N, Nonaka S, Welsh IC, Lan Y, Jiang R, Saijoh Y, and/or Alagille syndrome. Circulation 106: 2567–2574, 2002.
O’Brien TP, Hamada H, Gridley T. Notch signaling regulates left-right 85a.Monet M, Domenga V, Lemaire B, Souilhol C, Langa F, Babinet C,
asymmetry determination by inducing Nodal expression. Genes Dev 17: Gridley T, Tournier-Lasserve, Cohen-Tannoudji, Joutel A. The ar-
1207–1212, 2003. chetypal R90C CADASIL-NOTCH3 mutation retains Notch3 function in
66. Krebs LT, Shutter JR, Tanigaki K, Honjo T, Stark KL, Gridley T. vivo. Hum Mol Genet 16: 982–992, 2007.
Haploinsufficient lethality and formation of arteriovenous malformations 86. Nam Y, Sliz P, Song L, Aster JC, Blacklow SC. Structural basis for
in Notch pathway mutants. Genes Dev 18: 2469 –2473, 2004. cooperativity in recruitment of MAML coactivators to Notch transcrip-
67. Krebs LT, Xue Y, Norton CR, Shutter JR, Maguire M, Sundberg JP, tion complexes. Cell 124: 973–983, 2006.
Gallahan D, Closson V, Kitajewski J, Callahan R, Smith GH, Stark 87. Nam Y, Weng AP, Aster JC, Blacklow SC. Structural requirements for
KL, Gridley T. Notch signaling is essential for vascular morphogenesis assembly of the CSL.intracellular Notch1.Mastermind-like 1 transcrip-
in mice. Genes Dev 14: 1343–1352, 2000. tional activation complex. J Biol Chem 278: 21232–21239, 2003.
68. Krebs LT, Xue Y, Norton CR, Sundberg JP, Beatus P, Lendahl U, 88. Nickoloff BJ, Osborne BA, Miele L. Notch signaling as a therapeutic
Joutel A, Gridley T. Characterization of Notch3-deficient mice: normal target in cancer: a new approach to the development of cell fate modi-
embryonic development and absence of genetic interactions with a fying agents. Oncogene 22: 6598 – 6608, 2003.
Notch1 mutation. Genesis 37: 139 –143, 2003. 89. Noden DM. Embryonic origins and assembly of blood vessels. Am Rev
69. Kurooka H, Kuroda K, Honjo T. Roles of the ankyrin repeats and Respir Dis 140: 1097–1103, 1989.
C-terminal region of the mouse notch1 intracellular region. Nucleic Acids 90. Noseda M, Fu Y, Niessen K, Wong F, Chang L, McLean G, Karsan
Res 26: 5448 –5455, 1998. A. Smooth Muscle (alpha)-actin is a direct target of Notch/CSL. Circ Res
70. Lawson ND, Scheer N, Pham VN, Kim CH, Chitnis AB, Campos- 98: 1468 –1470, 2006.
Ortega JA, Weinstein BM. Notch signaling is required for arterial- 91. Noseda M, McLean G, Niessen K, Chang L, Pollet I, Montpetit R,
venous differentiation during embryonic vascular development. Devel- Shahidi R, Dorovini-Zis K, Li L, Beckstead B, Durand RE, Hoodless
opment 128: 3675–3683, 2001. PA, Karsan A. Notch activation results in phenotypic and functional
71. Lawson ND, Vogel AM, Weinstein BM. sonic hedgehog and vascular changes consistent with endothelial-to-mesenchymal transformation.
endothelial growth factor act upstream of the Notch pathway during Circ Res 94: 910 –917, 2004.
arterial endothelial differentiation. Dev Cell 3: 127–136, 2002. 92. Oberg C, Li J, Pauley A, Wolf E, Gurney M, Lendahl U. The Notch
72. Leimeister C, Externbrink A, Klamt B, Gessler M. Hey genes: a novel intracellular domain is ubiquitinated and negatively regulated by the
subfamily of hairy- and Enhancer of split related genes specifically mammalian Sel-10 homolog. J Biol Chem 276: 35847–35853, 2001.
expressed during mouse embryogenesis. Mech Dev 85: 173–177, 1999. 93. Oka C, Nakano T, Wakeham A, de la Pompa JL, Mori C, Sakai T,
73. Leimeister C, Schumacher N, Steidl C, Gessler M. Analysis of HeyL Okazaki S, Kawaichi M, Shiota K, Mak TW, Honjo T. Disruption of
expression in wild-type and Notch pathway mutant mouse embryos. the mouse RBP-J kappa gene results in early embryonic death. Devel-
Mech Dev 98: 175–178, 2000. opment 121: 3291–3301, 1995.
74. Leong GM, Subramaniam N, Issa LL, Barry JB, Kino T, Driggers 94. Okochi M, Steiner H, Fukumori A, Tanii H, Tomita T, Tanaka T,
PH, Hayman MJ, Eisman JA, Gardiner EM. Ski-interacting protein, a Iwatsubo T, Kudo T, Takeda M, Haass C. Presenilins mediate a dual
bifunctional nuclear receptor coregulator that interacts with N-CoR/ intramembranous gamma-secretase cleavage of Notch-1. EMBO J 21:
SMRT and p300. Biochem Biophys Res Commun 315: 1070 –1076, 2004. 5408 –5416, 2002.
75. Lindner V, Booth C, Prudovsky I, Small D, Maciag T, Liaw L. 95. Ong CT, Cheng HT, Chang LW, Ohtsuka T, Kageyama R, Stormo
Members of the Jagged/Notch gene families are expressed in injured GD, Kopan R. Target selectivity of vertebrate notch proteins. Collabo-
arteries and regulate cell phenotype via alterations in cell matrix and ration between discrete domains and CSL-binding site architecture de-
cell-cell interaction. Am J Pathol 159: 875– 883, 2001. termines activation probability. J Biol Chem 281: 5106 –5119, 2006.
76. Logeat F, Bessia C, Brou C, LeBail O, Jarriault S, Seidah NG, Israel 96. Petcherski AG, Kimble J. Mastermind is a putative activator for Notch.
A. The Notch1 receptor is cleaved constitutively by a furin-like conver- Curr Biol 10: R471–R473, 2000.
tase. Proc Natl Acad Sci USA 95: 8108 – 8112, 1998. 97. Przemeck GK, Heinzmann U, Beckers J, Hrabe de Angelis M. Node
77. Loomes KM, Taichman DB, Glover CL, Williams PT, Markowitz and midline defects are associated with left-right development in Delta1
JE, Piccoli DA, Baldwin HS, Oakey RJ. Characterization of Notch mutant embryos. Development 130: 3–13, 2003.
receptor expression in the developing mammalian heart and liver. Am J 98. Pursglove SE, Mackay JP. CSL: a notch above the rest. Int J Biochem
Med Genet 112: 181–189, 2002. Cell Biol 37: 2472–2477, 2005.
78. Loomes KM, Underkoffler LA, Morabito J, Gottlieb S, Piccoli DA, 99. Qiu L, Joazeiro C, Fang N, Wang HY, Elly C, Altman Y, Fang D,
Spinner NB, Baldwin HS, Oakey RJ. The expression of Jagged1 in the Hunter T, Liu YC. Recognition and ubiquitination of Notch by Itch, a
developing mammalian heart correlates with cardiovascular disease in hect-type E3 ubiquitin ligase. J Biol Chem 275: 35734 –35737, 2000.
Alagille syndrome. Hum Mol Genet 8: 2443–2449, 1999. 100. Raffin M, Leong LM, Rones MS, Sparrow D, Mohun T, Mercola M.
79. Matsuno K, Ito M, Hori K, Miyashita F, Suzuki S, Kishi N, Artava- Subdivision of the cardiac Nkx2.5 expression domain into myogenic and
nis-Tsakonas S, Okano H. Involvement of a proline-rich motif and nonmyogenic compartments. Dev Biol 218: 326 –340, 2000.
RING-H2 finger of Deltex in the regulation of Notch signaling. Devel- 101. Rand MD, Grimm LM, Artavanis-Tsakonas S, Patriub V, Blacklow
opment 129: 1049 –1059, 2002. SC, Sklar J, Aster JC. Calcium depletion dissociates and activates
80. Mayer M, Straube A, Bruening R, Uttner I, Pongratz D, Gasser T, heterodimeric notch receptors. Mol Cell Biol 20: 1825–1835, 2000.
Dichgans M, Muller-Hocker J. Muscle and skin biopsies are a sensitive 102. Rangarajan A, Talora C, Okuyama R, Nicolas M, Mammucari C,
diagnostic tool in the diagnosis of CADASIL. J Neurol 246: 526 –532, Oh H, Aster JC, Krishna S, Metzger D, Chambon P, Miele L, Aguet
1999. M, Radtke F, Dotto GP. Notch signaling is a direct determinant of

AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.
Invited Review
NOTCH AND CARDIAC DEVELOPMENT C11
keratinocyte growth arrest and entry into differentiation. EMBO J 20: 122. Swiatek PJ, Lindsell CE, del Amo FF, Weinmaster G, Gridley T.
3427–3436, 2001. Notch1 is essential for postimplantation development in mice. Genes Dev
103. Raya A, Kawakami Y, Rodriguez-Esteban C, Buscher D, Koth CM, 8: 707–719, 1994.
Itoh T, Morita M, Raya RM, Dubova I, Bessa JG, de la Pompa JL, 123. Tamura K, Taniguchi Y, Minoguchi S, Sakai T, Tun T, Furukawa T,
Belmonte JC. Notch activity induces Nodal expression and mediates the Honjo T. Physical interaction between a novel domain of the receptor
establishment of left-right asymmetry in vertebrate embryos. Genes Dev Notch and the transcription factor RBP-J kappa/Su(H). Curr Biol 5:
17: 1213–1218, 2003. 1416 –1423, 1995.
104. Rebay I, Fleming RJ, Fehon RG, Cherbas L, Cherbas P, Artavanis- 124. Tani S, Kurooka H, Aoki T, Hashimoto N, Honjo T. The N- and
Tsakonas S. Specific EGF repeats of Notch mediate interactions with C-terminal regions of RBP-J interact with the ankyrin repeats of Notch1
Delta and Serrate: implications for Notch as a multifunctional receptor. RAMIC to activate transcription. Nucleic Acids Res 29: 1373–1380,
Cell 67: 687– 699, 1991.
2001.
105. Redkar A, Montgomery M, Litvin J. Fate map of early avian cardiac
125. Timmerman LA, Grego-Bessa J, Raya A, Bertran E, Perez-Pomares
progenitor cells. Development 128: 2269 –2279, 2001.
106. Ronchini C, Capobianco AJ. Induction of cyclin D1 transcription and JM, Diez J, Aranda S, Palomo S, McCormick F, Izpisua-Belmonte
CDK2 activity by Notch(ic): implication for cell cycle disruption in JC, de la Pompa JL. Notch promotes epithelial-mesenchymal transition
transformation by Notch(ic). Mol Cell Biol 21: 5925–5934, 2001. during cardiac development and oncogenic transformation. Genes Dev
107. Rones MS, McLaughlin KA, Raffin M, Mercola M. Serrate and Notch 18: 99 –115, 2004.
specify cell fates in the heart field by suppressing cardiomyogenesis. 126. Villa N, Walker L, Lindsell CE, Gasson J, Iruela-Arispe ML, Wein-
Development 127: 3865–3876, 2000. master G. Vascular expression of Notch pathway receptors and ligands
108. Ruchoux MM, Chabriat H, Bousser MG, Baudrimont M, Tournier- is restricted to arterial vessels. Mech Dev 108: 161–164, 2001.
Lasserve E. Presence of ultrastructural arterial lesions in muscle and skin 127. Waldo KL, Kumiski DH, Wallis KT, Stadt HA, Hutson MR, Platt
vessels of patients with CADASIL. Stroke 25: 2291–2292, 1994. DH, Kirby ML. Conotruncal myocardium arises from a secondary heart
109. Ruchoux MM, Domenga V, Brulin P, Maciazek J, Limol S, Tour- field. Development 128: 3179 –3188, 2001.
nier-Lasserve E, Joutel A. Transgenic mice expressing mutant Notch3 128. Wang J, Sridurongrit S, Dudas M, Thomas P, Nagy A, Schneider
develop vascular alterations characteristic of cerebral autosomal domi- MD, Epstein JA, Kaartinen V. Atrioventricular cushion transformation
nant arteriopathy with subcortical infarcts and leukoencephalopathy. is mediated by ALK2 in the developing mouse heart. Dev Biol 286:
Am J Pathol 162: 329 –342, 2003. 299 –310, 2005.
110. Ruchoux MM, Guerouaou D, Vandenhaute B, Pruvo JP, Vermersch 129. Wang W, Prince CZ, Mou Y, Pollman MJ. Notch3 signaling in
P, Leys D. Systemic vascular smooth muscle cell impairment in cerebral vascular smooth muscle cells induces c-FLIP expression via ERK/MAPK
autosomal dominant arteriopathy with subcortical infarcts and leukoen- activation. Resistance to Fas ligand-induced apoptosis. J Biol Chem 277:
cephalopathy. Acta Neuropathol (Berl) 89: 500 –512, 1995. 21723–21729, 2002.
111. Saga Y, Kitajima S, Miyagawa-Tomita S. Mesp1 expression is the
130. Warthen DM, Moore EC, Kamath BM, Morrissette JJ, Sanchez P,
earliest sign of cardiovascular development. Trends Cardiovasc Med 10:
Piccoli DA, Krantz ID, Spinner NB. Jagged1 (JAG1) mutations in
345–352, 2000.
112. Saga Y, Miyagawa-Tomita S, Takagi A, Kitajima S, Miyazaki J, Alagille syndrome: increasing the mutation detection rate. Hum Mutat
Inoue T. MesP1 is expressed in the heart precursor cells and required for 27: 436 – 443, 2006.
the formation of a single heart tube. Development 126: 3437–3447, 1999. 131. Watanabe Y, Kokubo H, Miyagawa-Tomita S, Endo M, Igarashi K,
113. Sanchez-Irizarry C, Carpenter AC, Weng AP, Pear WS, Aster JC, Aisaki KI, Kanno J, Saga Y. Activation of Notch1 signaling in cardio-
Blacklow SC. Notch subunit heterodimerization and prevention of li- genic mesoderm induces abnormal heart morphogenesis in mouse.
gand-independent proteolytic activation depend, respectively, on a novel Development 133: 1625–1634, 2006.
domain and the LNR repeats. Mol Cell Biol 24: 9265–9273, 2004. 132. Whelan JT, Forbes SL, Bertrand FE. CBF-1 (RBP-Jkappa) binds to
114. Schlange T, Andree B, Arnold H, Brand T. Expression analysis of the the PTEN promoter and regulates PTEN gene expression. Cell Cycle 6:
chicken homologue of CITED2 during early stages of embryonic devel- 80 – 84, 2007.
opment. Mech Dev 98: 157–160, 2000. 133. Williams R, Lendahl U, Lardelli M. Complementary and combinatorial
115. Schneider A, Brand T, Zweigerdt R, Arnold H. Targeted disruption of patterns of Notch gene family expression during early mouse develop-
the Nkx3.1 gene in mice results in morphogenetic defects of minor ment. Mech Dev 53: 357–368, 1995.
salivary glands: parallels to glandular duct morphogenesis in prostate. 134. Xue Y, Gao X, Lindsell CE, Norton CR, Chang B, Hicks C, Gendron-
Mech Dev 95: 163–174, 2000. Maguire M, Rand EB, Weinmaster G, Gridley T. Embryonic lethality
116. Schroeter EH, Kisslinger JA, Kopan R. Notch-1 signalling requires and vascular defects in mice lacking the Notch ligand Jagged1. Hum Mol
ligand-induced proteolytic release of intracellular domain. Nature 393: Genet 8: 723–730, 1999.
382–386, 1998. 135. You LR, Lin FJ, Lee CT, DeMayo FJ, Tsai MJ, Tsai SY. Suppression
117. Sissman NJ. Developmental landmarks in cardiac morphogenesis: com- of Notch signalling by the COUP-TFII transcription factor regulates vein
parative chronology. Am J Cardiol 25: 141–148, 1970. identity. Nature 435: 98 –104, 2005.
118. Six E, Ndiaye D, Laabi Y, Brou C, Gupta-Rossi N, Israel A, Logeat
136. Zhong TP, Childs S, Leu JP, Fishman MC. Gridlock signalling
F. The Notch ligand Delta1 is sequentially cleaved by an ADAM
pathway fashions the first embryonic artery. Nature 414: 216 –220, 2001.
protease and gamma-secretase. Proc Natl Acad Sci USA 100: 7638 –
7643, 2003. 137. Zhong TP, Rosenberg M, Mohideen MA, Weinstein B, Fishman MC.
119. Souilhol C, Cormier S, Tanigaki K, Babinet C, Cohen-Tannoudji M. gridlock, an HLH gene required for assembly of the aorta in zebrafish.
RBP-Jkappa-dependent notch signaling is dispensable for mouse early Science 287: 1820 –1824, 2000.
embryonic development. Mol Cell Biol 26: 4769 – 4774, 2006. 138. Zhou S, Fujimuro M, Hsieh JJ, Chen L, Miyamoto A, Weinmaster
120. Spinner NB, Colliton RP, Crosnier C, Krantz ID, Hadchouel M, G, Hayward SD. SKIP, a CBF1-associated protein, interacts with the
Meunier-Rotival M. Jagged1 mutations in alagille syndrome. Hum ankyrin repeat domain of NotchIC To facilitate NotchIC function. Mol
Mutat 17: 18 –33, 2001. Cell Biol 20: 2400 –2410, 2000.
121. Struhl G, Greenwald I. Presenilin-mediated transmembrane cleavage is 139. Zweifel ME, Leahy DJ, Hughson FM, Barrick D. Structure and
required for Notch signal transduction in Drosophila. Proc Natl Acad Sci stability of the ankyrin domain of the Drosophila Notch receptor. Protein
USA 98: 229 –234, 2001. Sci 12: 2622–2632, 2003.

AJP-Cell Physiol • VOL 293 • JULY 2007 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (001.158.080.078) on November 25, 2023.

You might also like