You are on page 1of 9

Cardiovascular Research (2011) 91, 260268 SPOTLIGHT REVIEW

doi:10.1093/cvr/cvr035

Vascularizing the heart


Paul R. Riley * and Nicola Smart
Molecular Medicine Unit, UCL-Institute of Child Health, London WC1N 1EH, UK

Received 30 November 2010; revised 26 January 2011; accepted 27 January 2011; online publish-ahead-of-print 31 January 2011

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


Abstract As the developing heart grows and the chamber walls thicken, passive diffusion of oxygen and nutrients is replaced by
a vascular plexus which remodels and expands to form a mature coronary vascular system. The coronary arteries and
veins ensure the continued development of the heart and facilitate cardiac output with progression towards birth.
Many aspects of coronary vessel development are surprisingly not well understood and recently there has been
much debate surrounding both the developmental origin and tissue contribution of cardiovascular cells alongside
the specific signals that determine their fate and function. What is clear is that an understanding of the cellular
and molecular cues to vascularize the heart of the embryo has significant implications for adult heart disease and
regeneration, as we move towards targeted cell-based therapies for neovascularization and coronary bypass engraft-
ment. This review will focus on the proposed cellular origins for the coronary endothelium with due consideration to
the pro-epicardial organ/epicardium, sinus venosus and endocardium as potential sources, and we will explore the
outstanding questions and technical limitations with respect to accurate labelling and lineage tracing of the developing
coronaries. We will briefly document canonical vascular signalling that induces vessels in the heart alongside a focus
on the potential for developmental reprogramming and putative mechanisms underpinning venous vs. arterial cell fate.
Finally, we will extrapolate directly from development to address adult maintenance of the coronaries, vascular homeo-
stasis and remodelling in response to pathology, aligned with the potential for revascularizing the injured adult heart.
-----------------------------------------------------------------------------------------------------------------------------------------------------------
Keywords Coronary vasculogenesis Endothelial cells Proepicardial organ Sinus venosus Endocardium
Neovascularization
-----------------------------------------------------------------------------------------------------------------------------------------------------------
This article is part of the Spotlight Issue on: Cardiac Development

there is the distinct possibility of developmental studies uncovering


1. Introduction cellular and molecular cues towards neovascular repair of the dis-
The development of the coronary vessels, and subsequent attachment eased adult heart.
of the main coronaries to the closed vascular system of the embryo, is
fundamental for continued foetal growth and survival. Traditionally,
coronary vessels were thought to have a unique derivation from 2. Cellular origins of the coronary
mesothelial cells arising from the proepicardium or proepicardial
organ (PEO) and yet retain a high level of functional and molecular
vasculature
similarity to the systemic vasculature. Proepicardial cells are trans- Despite the fact that the development of the vessels of the heart has
ferred to the heart by complex signalling and epithelial mesenchymal been a subject of relatively intense study for more than a century,
transition (EMT) to form the epicardium and give rise to endothelial there is still ongoing debate regarding the cellular origin and tissue
cells (ECs), smooth muscle cells, and cardiac fibroblasts (reviewed in sources for the coronary vasculature.
Smart et al.1). Endothelial cells derived from the epicardium form a
vascular plexus by combined vasculogenesis and angiogenesis to
remodel and expand the vascular network, which subsequently 2.1 The PEO/epicardium: a historical
attaches to the aorta and recruits epicardially derived mesenchyme perspective
to become vascular smooth muscle cells (VSMCs) through arterio- Early anatomical observations documented that the coronary vessels
genesis. Interest in coronary vessel formation stems not only from simply bud off from the aorta but these were challenged by studies in
the fact that it is essential for embryonic survival, but also because which coronary arteries were observed to grow into the aorta.2 More

* Corresponding author. Tel: +44 20 7905 2345, fax: +44 20 7404 6191, Email: p.riley@ich.ucl.ac.uk
Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: journals.permissions@oup.com.
Vascularizing the heart 261

recent retroviral tagging, adenoviral cell lineage tracing, and quail in the developing heart which establishes a potential for contribution
chick chimera studies have proposed the PEO as a source of the cor- of the former to the latter population, although, to date, this has again
onary vasculature.3 5 The PEO is a transient extracardiac mesothelial proven somewhat controversial. That the primitive vascular bed might
cell population situated between the sinus venosus and the liver in arise as a continuation of the endocardium was first described in the
chick and on the surface of the septum transversum in mouse.6 developing rat heart19 and subsequently in mouse, where the first
PEO cells migrate to the developing heart and envelope the surface vessels of the heart were described as appearing by the invagination
to give rise to the epicardium. A subpopulation of epicardium-derived of the endocardium.6 In contrast, retroviral tracing in chick revealed
progenitor cells delaminate from this primitive epithelium and ECs of the coronary vessels to have a different clonal origin than
undergo EMT to generate a population of mesenchymal cells which ECs of the endocardium.7 Red-Horse et al. 13 were the first to geneti-
migrate into the underlying myocardium and give rise to VSMCs, peri- cally trace and clonally map an endocardial origin for coronary ECs,
cytes, fibroblasts, and cardiomyocytes.3,7 9 That the coronary ECs are albeit a relatively minor contribution with the tools they employed
also EPDC-derived is controversial. It is widely accepted that not all and described a process of endocardial budding to form endothelial
vascular ECs are contributed by the epicardium,10 and an increasing blood islands which they hypothesized might join the sinus venosus-
number of studies now argue that very few, if any, coronary ECs derived plexus. A more autonomous and potentially more extensive
are PEO epicardium-derived. In chick quail chimera studies with contribution of endocardial ECs to the coronary vasculature is yet

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


anti-endothelial immunohistochemistry, prospective coronary ECs to be determined, but since endocardial cells closely resemble endo-
were mapped back to the liver sinusoids extending into the sinus thelial precursor cells in terms of their molecular profile, unequivocal
venosus lumen via the PEO11 with the liver subsequently proposed lineage tracing and chimera analyses are somewhat confounded.12
to be a primary source of endothelial progenitor cells (EPCs).12 In
mouse, lineage tracing and fate mapping of epicardium derived cells
(EPDCs) during development have either failed to identify 2.3 Reconciling a common source
epicardium-derived ECs8 or observed that the epicardium gives rise for coronary ECs
to only a few isolated ECs.9 So what is the current state of play and what if any is the consensus on
the origins of the coronary vasculature? In attempting to reconcile all
of the above, it is important to take account of species differences,
2.2 A sinus venosus and endocardium with a focus on avian vs. rodent as the most studied vertebrate
contribution? models.
More recently, in mouse, coronary vessels have been shown to arise It would appear that there are two schools of thought derived from
from angiogenic sprouts of the sinus venosus, the endothelial-lined avian studies. The first advocates that the PEO/epicardium contributes
cavity at the inflow region of the embryonic heart.13 The sinus cells to the developing coronary vessels,5 although the PEO is not the
venosus was identified as a site for the earliest appearance of sole source20 and indeed it is apparent that other cell types may par-
vessels in the heart in classical embryological studies in dogfish14 ticipate in the building of the coronary tree. The second focuses on
and, in mouse, a connection of the undifferentiated coronary vascular the other cell types and in particular proposes a non-PEO source
network to the sinus venosus was also previously demonstrated.15 favouring the liver sinusoids and sinus venosus.11 In this respect, the
However, there was no historical insight into a sinus venosus origin quail-to-chick and/or retroviral techniques used to tag the develop-
for coronary artery ECs until the recent study by Red-Horse mental fate of avian PEO or sinus venosus cells cannot guarantee
et al.13 Here, the authors fate mapped lacZ+ ECs via an that all the coronary progenitors are properly labelled and, therefore,
ephrinB2-LacZ reporter to determine that coronary sprouts were not only is it difficult to encompass different contributions from differ-
continuous with the sinus venosus and did not arise from the PEO ent sources in a single experiment, but it is equally difficult to make an
or other vasculogenic sources. This was complimented by organ accurate quantitative assessment between experiments for cellular
culture and tissue recombination to demonstrate EC outgrowth contribution in coronary morphogenesis.
restricted to sinus venosus and atrium, which in turn was dependent In rodents, and in particular mice, where genetic lineage tracing and
upon unknown signals from the ventricle and epicardium, and clonal fate mapping tools are available, there has been a recent movement
analysis with an inducible VE-cadherin-CreER transgene to map the away from a single PEO/epicardium source for coronary ECs. New
origin and fate of coronary endothelial progenitors in vivo. The insight has informed on contributions from dedifferentiation of the
clonal analysis, via an early (E7.5) induction of VE-cadherin-CreER, sinus venosus endothelium at least giving rise to the coronary arteries.
suggested that coronary artery progenitors arise from differentiated This of course begs the question as to the origin of the ECs that com-
veins and this was confirmed by documenting a switch in molecular prise the early coronary veins and in particular the endothelial lining
signature from a venous (EphB4+) to arterial (ephrinB2+) identity of the sinus venosus? Moreover, the endocardium has been implicated
within the coronary sprouts as they emerged from the sinus venosus. as a source of ECs that may further contribute to the developing cor-
What the clonal analysis also revealed13 was a secondary source of onary plexus, and indeed, the precise extent of endocardial contri-
coronary ECs from the endocardium. In the mouse, endocardial cells bution remains unknown. It would appear that regardless of source,
are among the earliest ECs that differentiate from Vegfr-2-positive the coronary ECs in mammals represent a specified lineage within
(Vegfr-2+) multipotent progenitors within the heart field.16 They the vasculature arising in part from a subpopulation of venous ECs
form an endocardial tube by de novo growth or vasculogenesis and at defined stages in development. Furthermore, as far as the PEO
later become the inner epithelial lining or endocardium of the and sinus venosus are concerned, their adjacent location at the
heart,17 subsequently contributing to the formation of the cardiac inflow region of the developing mouse heart and proximal to the
valves and membranous septa by endocardial-to-mesenchymal trans- liver primordium (Figure 1A) means that it is extremely difficult with
formation.18 Relative to the endocardial ECs, coronary ECs arise later the current tools available (specific Cre-expressing lineage mouse
262 P.R. Riley and N. Smart

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


Figure 1 A schematized heart during the initial stages of coronary vasculogenesis. The relative positions of the putative tissue origins of coronary
ECs are shown in (A), with the liver sinuses (ls; blue), pro-epicardial organ/epicardium (peo; green), and sinus venosus (sv; blue) highlighted; ra, right
atrium; rv, right ventricle. Potential cellular contributions between the sources and ultimately to the developing venous and arterial sprouts are high-
lighted in (B), arising from the ls (blue), peo/epicardium (green), sv (blue), and endocardium (red); arrowheads indicate fate (blue, sv or sv venous
sprout; green, peo/epicardium; and red, arterial sprout).

lines) to unequivocally rule out common progenitor pools. Equally, contains some sort of vascular progenitor cell type or (haem-) angio-
we are unable to discern early contribution of prospective liver blast. The specific embryological origin of this angioblast population
sinusoid progenitors directly to the sinus venosus, or via the PEO and whether it contributes as a whole or in part to the sinus
which in turn contribute to the sinus venosus and ultimately the venosus ECs, or the endocardium, as an intermediate stage prior to
coronary vasculature (Figure 1B). In the principal cell lineage tracing coronary vascularization (Figure 1B), remain to be determined and
adopted by Red-Horse et al.,13 namely the crossing of are still open to speculation.
VE-Cadherin-CreER;ROSA26 animals, while they could rule in the
sinus venosus source for coronary ECs, the selective expression of 2.5 Cre-lox technology to dissect out
Cre under the control of VE cadherin and the relative timing of induc-
tion meant that they were unable to rule out the possibility that ECs,
coronary origin
either from a distinct source (PEO, endocardium) or arising as earlier When addressing the question of origin, it is imperative to consider
progenitors elsewhere and migrating to the sinus venosus, may con- the possible vagaries of fate mapping alongside the pitfalls of using
tribute to the building of the coronary tree. the favoured Cre-lox system in mouse, as weighted against species-
specific differences and possible heterogeneity in the origins of the
vascular lineages. In addition, there is a requirement to establish cri-
2.4 Confounding avian rodent teria for optimal assessment of the progressive contribution of coron-
species differences ary vascular cells from the earliest angioblast cell (if such a cell exists?)
Differences between the chick quail and mouserat studies may to the differentiated arterial EC. From the outset, we require a wider
reflect not only the outcome of different technologies applied, retro- range of markers of the prospective tissue sources for coronary cells,
viral tracing in chick vs. genetic fate mapping in mouse, but also may be both to distinguish between alternate origins (Figure 1) and to facilitate
an indictment on differences in the formation and patterning of the fate mapping at the earliest time points, thus addressing the question
vascular beds in avian compared with rodent hearts. A potentially uni- of a common progenitor pool. In this regard, the so-called natural
fying theme, however, is that most, if not all, of the reports found in history of genes we might adopt as tools for fate mapping becomes
the literature support the concept that the avian and mammalian PEO important. Within a specific source, we require finer mapping to
Vascularizing the heart 263

identify progenitors which may have overlapping and/or distinct and has been used successfully elsewhere in both chick and
expression profiles for specific markers and to identify a more com- mouse.24 Organ culture and tissue recombination, as previously
prehensive gene-set with which to speak to the entire heterogeneity described,13 is potentially informative regarding a single origin but is
of cells within a source and to resolve the complete fate map. restricted against the breadth of potential sources in vivo as it is not
In the case of the PEO, genetic markers which are expressed at an possible to ascertain whether all coronary ECs might arise from the
early stage in PEO development and in cells entirely or partially dis- specific cultured tissues. Additional insight can arise from the
tinct from previously described Tbx18-positive and Wt1-positive culture of neighbouring tissues where there may be a requirement
populations are required to trace distinct subcompartments. One for secreted signals from one tissue to induce coronary EC outgrowth
approach towards identifying novel factors around the model of a and remodelling in another and, in such cases, co-culture may facilitate
venous origin might be to build on the transcriptional code for sys- the identification of signals by mass spectrometry or via a candidate
temic vascular ECs defined by a composite cis-acting element, the approach.25
FOX:ETS motif.21 Thus, a suitable screen for coronary EC-specific Clonal analysis is a potentially powerful tool to map origin and
enhancers, which might act in concert with the FOX:ETS/ets determine proliferation, outgrowth and fate of the coronary progeni-
cluster, could be utilized to establish reporter lines and to identify tors in vivo. This analysis is, however, open to over-interpretation and
trans-acting factor(s) for use in Cre-lox lineage tracing. Subsequently, dependent upon inducible genetic labelling which can be biased from

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


selection of a specific gene locus and/or enhancer region to drive Cre the outset, so as not to be inclusive of all possible sources.13 Here, the
should be set against high-resolution in situ expression for the gene of solution might be to perform a time course of inductions to poten-
interest to ensure that there is no possibility of Cre being expressed tially encompass all clones of vascular progenitors. A further issue
within the descendant ECs or in more than one putative source for with clonal analysis is the documenting of a rare genetic recombina-
ECs to cloud interpretation on origin. Ideally, in any single study, mul- tion event and the tracking of a discrete number of clones on the
tiple Cre lines should be employed to identify potentially distinct assumption that this stochastically reflects the entire lineage. In the
sources for ECs and one must consider the efficacy of the Cre. case of venous-to-arterial reprogramming, the frequency of arterial
With variable efficacy comes differential sensitivity of individual EC clones arising from venous ECs was in fact quite low (around
alleles to Cre-mediated recombination which limits the utility of 13%)13 which could be interpreted to the effect that venous ECs
reporter alleles.22 This has important implications for interpreting are not the primary source of coronary arteries, leaving open the
Cre fate mapping experiments with the potential for alternate repor- identity of the arterial antecedents.
ters defining narrower or broader daughter cell domains. Recently, A further complementary approach is engraftment, such that
the widely used R26R reporter, based on the Rosa-26 locus, was labelled endothelial progenitors are transplanted into the embryo
shown to be less susceptible to Cre in studies tracking Isl1- and to determine lineage potential and an EC fate within the developing
Nkx2.5-expressing progenitors in cardiogenesis than an alternative heart. This is, however, compounded by technical difficulties regarding
Gata4-flap reporter, providing unique insight into the contribution the isolation and transplantation procedure, propensity for phenoty-
of Isl1 descendants to both primary and secondary heart fields and pic drift during culture, and mode of delivery. Further caveats
Nkx2.5 descendants to the coronary endothelium and smooth include subsequent graft survival alongside a reduced exposure to
muscle.23 A further generic challenge with Cre-lox technology is the appropriate in vivo signals which may be critical to determine
the issue of potential toxicology of Cre recombinase arising from EC fate.
recombination at cryptic or pseudo-loxP sites within the genome hin-
dering the cellular DNA repair machinery and ultimately resulting in 2.7 Defined sources of coronary vascular
apoptosis.22 smooth muscle
As a minimum requirement, the Cre drivers should be inducible to
In stark contrast to the coronary endothelium, the VSMCs have fairly
avoid issues with (i) spurious activation of Cre due to prior expression
well-defined origins, dependent on location within the developing
in founder populations, (ii) detecting descendants of rare cells that
heart and function (reviewed in Luttun and Carmeliet26). Simply
have activated Cre at an earlier time point, and (iii) ectopic expression
put, VSMCs of the proximal and major anterior coronary arteries
of Cre in ECs that does not reflect lineage contribution. Finally, the
arise from the neural crest and the rest derive from the PEO and
timing of induction needs to be early enough so as to trace progeni-
epicardium.27 In contrast, coronary vein VSMCs arise from an atrial
tors which may contribute transiently to intermediate sources (e.g. a
cardiomyocyte origin.28
potential PEO/epicardium contribution to the endocardium and sinus
venosus) en route to the coronaries (Figure 1).
3. Developmental programming
2.6 Classical embryology, clonal of vessels in the heart
and transplant analyses In keeping with the prevailing dogma of a PEO/epicardium origin for
Even if safeguards are imposed against a misinterpretation of the the coronary vasculature, there has been considerable focus on the
Cre-lox labelling, evidence that the coronary cells are indeed the communication of signals between the PEO/epicardium, subepicardial
linear descendents of cells that were expressing the gene (employed mesenchyme, and myocardium. These signals regulate coronary
to drive Cre) in the specific source at earlier stages must be provided development and myocardial growth and are mediated in part by
to claim lineage tracing and fate mapping. Here, Cre-lox can be com- secreted growth factors which originate from the epicardium and
plimented by other analyses. Returning to more classical embryology, EPDCs or from cardiomyocytes and which sit at the top of hierarch-
in vivo vital dye (DiI) injection and physical labelling of progenitors in ical signalling pathways to act in an autocrine or paracrine fashion.
conjunction with whole-embryo culture remains a valid approach Although some growth factors have been implicated in the formation
264 P.R. Riley and N. Smart

of vascular beds elsewhere during development, the pathways they of arterial cell fate. Delta-Notch plays a central role in vertebrate
mediate within the heart are largely distinct from those that govern organogenesis per se and regulates multiple processes during cardiac
vasculogenesis in the rest of the embryo (reviewed in Olivey and development, where it functions both cell autonomously and
Svensson29). What is evident is that reciprocal signalling between non-cell autonomously pointing to a role as a signal coordinator
lineages such as the PEO/epicardium, endocardium, and myocardium during cardiogenesis (reviewed in MacGrogan et al.33). Notch signal-
ensures developmental coupling of coronary vessels with that of myo- ling within the myocardium is known to be important for cardiomyo-
cardial growth (reviewed in Bhattacharya et al.30). Suffice to say that cyte differentiation, ventricular trabeculation and outflow tract
TGF, FGF, VEGF, PDGF, BMP, Wnt/b-catenin and retinoic acid path- development, and there is also a critical role for Notch in the endo-
ways have all been implicated in epicardialmyocardial or myocar- cardium, such that Notch1, via interplay with myocardial Bmp2,
dialepicardial signalling principally in support of PEO/epicardial EMT, restricts endocardial EMT to a valve-forming field.34 In avian coronary
outgrowth, protrusion and adhesion of EPDCs plus the induction and vasculogenesis, active Notch1 (the Notch1 intracellular domain;
remodelling of the coronary plexus. For detailed accounts of growth N1ICD) is present within the mesothelial cells of the PEO, the
factor function and downstream signalling in coronary development, mesenchymal cells of the epicardium and in the ECs within the
refer to reviews elsewhere1,26,29 31 and references therein. nascent vessel plexus at the atrioventricular junction, suggesting that
Notch1 is important in the chick for epicardial EMT and coronary

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


3.1 Signalling to specify arteriovenous progenitor cell differentiation.35 In mice, which lack Notch1 or have
a combined loss of the Notch targets, Hey1 and Hey2, the mutant
identity?
phenotype is characterized by cardiac defects and coronaries which
When considering the molecular control of vessel formation in the
fail to express arterial markers, including ephrinB2. This suggests
developing heart, a key question is what are the signals which
that Hey1 and Hey2 are essential transducers of Notch signals and
specify venous vs. arterial identity? Physical forces such as blood
may mediate arterial fate decision.36 Notch also functions at later
flow have been implicated in the regulation of EC fate. Coronary
stages in coronary maturation and is up-regulated in response to
veins develop before arteries and the latter have been considered
VEGF signalling to establish a Notch/ephrinB2 cascade and ensure
simplistically as a by-product of the recruitment of a medial layer of
coronary arteriogenesis.15
VSMCs in response to pressure increase and blood flow alteration
after connection to the aorta.26 Hence, direct stretch and sheer
forces acting on the endothelium or VSMC differentiation and sub-
sequent smooth muscle paracrine signalling may instruct coronary 3.3 Determinants of venous identity
ECs to adopt an arterial fate, or at later stages ensure appropriate and reprogramming?
arterial EC differentiation and functional maturation. A robust argu-
Although Notch signalling may define coronary arterial ECs, we have
ment against altered haemodynamics as determinants of arterial EC
no current insight into the determinants of coronary venous fate or
fate arises in the study proclaiming coronary artery formation by
the signals which instigate venous reprogramming in the process of
developmental reprogramming of venous cells.13 In the model pro-
coronary artery formation. During remodelling of the systemic
posed, sprouting sinus venosus ECs are initially differentiated with a
primary capillary plexus in the early embryo, arteriovenous differen-
venous identity characterized by Ephb4 expression and other
tiation involves suppression of Notch signalling in venous ECs by
venous markers such as Vegfr3, Np2, Aplnr and COUP-TFII but, as
the nuclear orphan receptor COUP-TFII which permits expression
angiogenesis extends beyond the sinus venosus, those vessels which
of the venous marker EphB4.37 This seems to indicate a default arter-
invade the myocardium begin to express ephrinB2 and other arterial
ial EC fate in systemic vasculogenesis which requires active suppres-
markers such as Dll4, Hey1, Notch4 and Depp. The timing of the
sion. It is not known whether this is the case in early coronary
initial down-regulation of Ephb4 as the first stage in venous repro-
vessel development but is somewhat unlikely, given the recent pre-
gramming towards arterial fate is E11.5 in the mouse which signifi-
cedent for venous EC differentiation as the earlier event within the
cantly precedes arteriogenesis and the connection of the vascular
sinus venosus, followed by subsequent reprogramming towards an
network to the aorta. This suggests that arterial specification is a pre-
arterial fate.13
programmed spatial event occurring in response to either positive
In terms of venous reprogramming, although again untested in
induction via signals from the myocardium/endocardium or reduced
terms of the coronary vasculature, during systemic vasculogenesis,
exposure to inhibitory signals from the PEO/epicardium and sinus
it is clear that the EphB4 receptor tyrosine kinase and its plasma
venosus. However, this does not rule out a role for haemodynamics
membrane-spanning ligand, ephrinB2, demarcate venous and arter-
in ensuring subsequent assembly into mature coronary arteries. The
ial domains, respectively.38 Moreover, a number of studies of
identity of flow/pressure-dependent signals is currently unknown
EphB4- and ephrinB2-null mice have revealed roles for forward
but, outside of the coronaries, molecular readouts have been ele-
and reverse EphB4 ephrinB2 signalling in vascular morphogen-
gantly defined in response to haemodynamic changes in the develop-
esis.38 40 This establishes a molecular basis for bidirectional pro-
ing branchial arch arteries (BAAs). Blood flow in the left sixth BAA is
gramming of venous arterial EC fate; however, the exact
sufficient to stimulate PDGFR and VEGFR2 signalling to maintain
function and mode of action of bidirectional EphB4 ephrinB2 sig-
arterial structure whereas, with decreased blood flow in the right
nalling remains unknown (reviewed in Adams and Alitalo41). An
sixth BAA, the consequence is arterial regression.32
understanding of the molecular signals which instruct dedifferentia-
tion and reprogramming of coronary vessels is an obvious priority
3.2 Coronary artery induction by Notch in terms of not only identifying pathways underlying coronary
The molecular profile of coronary arterial ECs, which includes Dll4, development but towards the induction of new vessels in adult
Hey1 and Notch4, implicates Notch signalling as a potential inducer cardiac repair.
Vascularizing the heart 265

4. Adult maintenance and between endothelium-derived paracrine factors appears to regulate


vascular homeostasis, and similarly, VSMCs rarely proliferate under
pathological remodelling normal physiological conditions, rather they remain in a differentiated
contractile state.
4.1 Maturation of the post-natal coronaries
The coronary vasculature continues to mature post-natally, growing 4.3 Consequences of endothelial
significantly in the first 2 weeks after birth to keep pace with 80% dysfunction in vascular disease
increase in cardiac mass.42 Indeed, a 2.8-fold increase in capillary
In disease states leading to deterioration in endothelial function, a pro-
growth occurs during the first 5 days of post-natal life, predominantly
portion of VSMCs adopt a dedifferentiated, proliferative state leading
by intussusceptive growth, i.e. splitting of pre-existing capillaries.43
to pathological changes in the vascular walls, including a loss of cyto-
FGFs play a key role in perinatal arterial development, such that pre-
skeletal markers and their ability to respond to contractile stimuli. If
cocious expression of FGF-2 following retroviral epicardial injection in
the processes of endothelial homeostasis and repair both fail, endo-
chicks causes abnormal branching of coronary vessels,44 whereas
thelial damage ultimately leads to arterial stiffness (arteriosclerosis)
anti-FGF-2-neutralizing antibodies, administered between 5 and 12
and lipid accumulation (atherosclerosis). Atherosclerosis is not
days after birth, inhibited arteriolar growth.42 Coronary artery remo-

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


simply an inevitable degenerative consequence of ageing, rather a
delling is necessary to establish an effective hierarchy within the arter-
form of chronic inflammation.52 The inflammatory cytokines
ial tree. Initiation of blood flow through the coronary system provides
interleukin-1, tumour necrosis factor-a and g-interferon transform
the stimulus for remodelling the main arteries, inducing EC prolifer-
ECs to an active state. Activated ECs express leukocyte adhesion mol-
ation and up-regulating VEGF, integrin aVb3, PECAM-1 and
ecules and produce tissue factor to create a procoagulant environ-
VE-cadherin45 to increase vessel diameter. Anti-VEGF neutralizing
ment which is augmented by accompanying vasoconstriction
antibody disturbed the normal arteriolar hierarchy, implying that
through an up-regulation of endothelin-1 in a setting of diminished
although FGF-2 appears to direct arteriolar growth, its interaction
nitric oxide (NO) production. This in turn is permissive for neointimal
with VEGF serves to establish a normal hierarchy of the coronary
proliferation, due to loss of the usual VSMC growth inhibitory factors,
tree, limiting growth of terminal arterioles and facilitating growth
and vascular occlusion.
and remodelling of upstream arterioles.42
Coronary artery occlusion may not always lead to infarction since
4.2 Homeostatic maintenance and in two-thirds of patients with CAD, collateral arteries develop from
pre-existent interconnecting arterioles, by proliferation of VSMCs
turnover of adult endothelium
and ECs, to bypass the stenosis.53 Fluid shear stress (FSS) is the
Once the mature coronary architecture is established, appropriate primary trigger for collateral growth and increased pulsatile FSS
angiogenic signalling is required thereafter for its maintenance. leads to deformation of the endothelium and activation of all NO
Renewal of vascular cells, as and when required, is facilitated by pro- synthase isoforms. NO production, followed by VEGF secretion,
genitor cell populations, derived either from bone marrow or resident induces monocyte chemoattractant protein-1 synthesis that attracts
within the vessels or myocardium46 (further reviewed in Hibbert monocytes and T cells to attach to the endothelium; these produce
et al.47). Even the healthy endothelium is turned over approximately proteases to digest the internal elastic lamina and extracellular
every 3 years48 and the cellular source for its renewal may not necess- matrix and simultaneously induce a phenotypic change of ECs and
arily derive from the same embryonic lineage as the original. The VSMCs into the synthetic and proliferative types. Following prolifer-
notion that vessels in the adult are derived only by angiogenesis, ation, vessels mature via orderly arrangement of the VSMCs in circular
and not by vasculogenesis as in the embryo, has now been superseded layers, establishment of cell-to-cell contacts and synthesis of elastin
following the acceptance that circulating EPCs, derived from bone and collagen to provide scaffold for the larger vessel.
marrow, represent at least one source of vascular progenitors that
actively contribute to new vessel growth in the adult. Analogous in
many respects to the embryonic angioblast, EPCs are mobilized, 5. Neovascularization and bypass
when required, from the bone marrow and incorporate into new
or injured vessels where they develop into mature ECs during the
engraftment
processes of re-endothelialization and neovascularization (reviewed Although the phenomenon of collateral growth has long been recog-
in Smart and Riley49). The precise molecular definition of EPCs nized, attempts to therapeutically stimulate these processes have
across animal models, and including humans, is somewhat controver- lagged behind. Capillaries that form by vasculogenesis, without
sial, although there is a consensus that their beneficial effects may also smooth muscle support, are unstable and regress over time.
be mediated, in part, through paracrine secretion of angiogenic factors Compared with vasculogenesis, arteriogenesis is more complex and
(Figure 2). has proven more recalcitrant to therapy. Arteriogenesis is not
ECs ordinarily reside in a non-proliferative state throughout their stimulated by the same signals, such as hypoxia, but rather by
average lifespan of between 1 and 3 years.50 Despite their quiescent haemodynamic forces, principally shear stress54; therefore, although
appearance, ECs are highly metabolically active, playing key roles in single genes or proteins successfully induce coronary angiogenesis,
controlling vasomotor tone, blood cell trafficking, haemostatic the simultaneous activation of multiple growth factors is seemingly
balance, permeability and immunity. The endothelium exerts effects required to stimulate arteriogenesis.55 Therapeutic methodologies
on both the surrounding VSMCs and blood cells to coordinately regu- have, therefore, evolved to concomitantly activate several growth
late tissue blood supply balancing vasodilation with vasoconstriction, factors, for example, by providing a metabolic stimulus, such as the
VSMC differentiation, inflammatory responses, maintenance of thyroxine analogue 3,5-diiodothyropropionic acid (DITPA).56 DITPA
blood fluidity and prevention of bleeding.51 A critical balance administration to rats post-MI enhanced levels of VEGF, FGF-2,
266 P.R. Riley and N. Smart

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


Figure 2 Cellular maintenance and repair of the post-natal coronary vasculature. Under conditions of vascular homeostasis (maintenance) in the
adult heart, circulating EPCs respond to a plethora of growth factor and chemokine signalling to home to the coronaries and replenish ECs. Following
arterial occlusion leading to vessel regression and MI, a subepicardial graft of c-kit+ cardiac progenitor cells (CPCs) is able to migrate to the site of
vascular injury and differentiate into de novo ECs and VSMCs to establish a bypass of the occlusion and reperfuse the injured myocardium.67 lad, left
anterior descending artery; mi, myocardial infarction; ANG1, angiopoietin 1; EPO, erythropoietin; FGF, fibroblast growth factor; HGF, hepatic growth
factor; HIF-1a, hypoxia factor-1a; SDF-1, stromal cell-derived factor-1; VEGF, vascular endothelial growth factor.

Ang-1, and Tie-2, leading to increased arteriolar density and improved embryonic programme that created the tissue in the first instance.
left ventricular function. As such, there is much to gain from understanding the embryonic
A natural consequence of arterial occlusion and ischaemia is the mechanisms of vasculogenesis (recently reviewed).1,49 The possibility
establishment of a hypoxic environment which has significant effects of a post-natal contribution from cardiac populations that play a
on the neovascular response. The regulation of angiogenesis by role during embryonic development is provided from studies in zeb-
hypoxia depends on a precise balance between positive and negative rafish which suggest that EPDCs are incorporated into the heart not
angiogenic regulatory molecules. Increasing evidence points to the only during regeneration, but also during continuous growth of the
existence of complex feedback networks such that factors generally adult heart.60 It remains to be determined whether the mammalian
considered pro-angiogenic, such as HIF-1, additionally activate angio- epicardium contributes to coronary homeostasis but, if so, the con-
genic inhibitors including Regulator of G protein signalling 5 (RGS5)57 tribution is likely to be much reduced compared with the fish for at
and Delta-like ligand 4 (Dll4)58 downstream of VEGF. The negative least two reasons. The epicardial contribution in zebrafish may be
feedback regulation of angiogenesis during hypoxia may explain not peculiar to the continued organ growth and hyperplasia which is
only the inadequate natural collateral circulation in ischaemic heart an adaptive precedent in this species and, moreover, in mammals
disease but also the disappointing and inconsistent results of clinical even in the injury setting, an exogenous stimulus, such as Thymosin
trials.59 Consequently, identifying key feedback inhibitors may reveal b4, is required to fully reinstate embryonic potential to adult
new targets for angiogenic therapy. EPDCs61 and enable neovascularization of the ischaemic adult
heart.62

5.1 Developmental programming to 5.2 Adult progenitor cell vascular repair


facilitate cell-based neovascularization Recent studies have recognized the ability of a multitude of cardiac-
An evolving paradigm in regenerative medicine is that tissue repair and vessel-resident progenitor cell populations to engraft into sites
in the adult is frequently underpinned by a re-activation of the of vascular injury or homeostatic replenishment (reviewed in
Vascularizing the heart 267

Hibbert et al.47). Along similar lines, the discovery that circulating 4. Munoz-Chapuli R, Perez-Pomares JM, Macias D, Garcia-Garrido L, Carmona R,
Gonzalez M. Differentiation of hemangioblasts from embryonic mesothelial cells? A
EPCs participate in homeostatic neovascular repair has led to many
model on the origin of the vertebrate cardiovascular system. Differentiation 1999;
animal studies (reviewed in Miller-Kasprzak and Jagodzinski63) and ulti- 64:133 141.
mately clinical trials to enhance EPC mobilization from bone marrow 5. Perez-Pomares JM, Carmona R, Gonzalez-Iriarte M, Atencia G, Wessels A,
Munoz-Chapuli R. Origin of coronary endothelial cells from epicardial mesothelium
to effect neovascularization, with promising preliminary results.64,65
in avian embryos. Int J Dev Biol 2002;46:1005 1013.
With regard to arteriogenic therapy, EPCs would appear inferior to 6. Viragh S, Challice CE. The origin of the epicardium and the embryonic myocardial cir-
those progenitor cells that possess a broader differentiation potential culation in the mouse. Anat Rec 1981;201:157 168.
that also includes pericytes or smooth muscle cells, given the need for 7. Mikawa T, Fischman DA. Retroviral analysis of cardiac morphogenesis: discontinuous
formation of coronary vessels. Proc Natl Acad Sci USA 1992;89:9504 9508.
mural cell stabilization of vessels. Smooth muscle progenitor cells 8. Cai CL, Martin JC, Sun Y, Cui L, Wang L, Ouyang K et al. A myocardial lineage derives
were more recently identified in bone marrow, circulating blood from Tbx18 epicardial cells. Nature 2008;454:104 108.
and vascular adventitia66 and may prove more successful for coronary 9. Zhou B, Ma Q, Rajagopal S, Wu SM, Domian I, Rivera-Feliciano J et al. Epicardial pro-
genitors contribute to the cardiomyocyte lineage in the developing heart. Nature
neovascularization. More recently, c-kit+ cardiac progenitor cells 2008;454:109 113.
were shown to differentiate into ECs and SMCs to form conductive 10. Morabito CJ, Kattan J, Bristow J. Mechanisms of embryonic coronary artery develop-
(up to 250 mm) and intermediate-sized coronary arteries together ment. Curr Opin Cardiol 2002;17:235 241.
11. Poelmann RE, Gittenberger-de Groot AC, Mentink MM, Bokenkamp R, Hogers B.
with resistance arterioles and capillaries. These new vessels con-

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


Development of the cardiac coronary vascular endothelium, studied with antien-
nected with the primary circulation to more than double myocardial dothelial antibodies, in chicken-quail chimeras. Circ Res 1993;73:559568.
flow following MI and thereby constitutes an example of a biological 12. Lie-Venema H, Eralp I, Maas S, Gittenberger-de Groot AC, Poelmann RE,
coronary bypass67 (Figure 2). Deruiter MC. Myocardial heterogeneity in permissiveness for epicardium-derived
cells and endothelial precursor cells along the developing heart tube at the onset
of coronary vascularization. Anat Rec A Discov Mol Cell Evol Biol 2005;282:120 129.
13. Red-Horse K, Ueno H, Weissman IL, Krasnow MA. Coronary arteries form by devel-
5.3 Tissue engineering of vascular grafts opmental reprogramming of venous cells. Nature 2010;464:549 553.
14. De Andres AV, Munoz-Chapuli R, Sans-Coma V. Development of the coronary
A natural progression from cell-based therapy has been the develop-
arteries and cardiac veins in the dogfish (Scyliorhinus canicula). Anat Rec 1993;235:
ment of tissue engineering techniques. New vessel formation and 436 442.
improvements in neovascularization have been reported in hindlimb 15. van den Akker NM, Caolo V, Wisse LJ, Peters PP, Poelmann RE, Carmeliet P et al.
Developmental coronary maturation is disturbed by aberrant cardiac vascular endo-
ischaemia using human smooth muscle cell sheets68 or Matrigel
thelial growth factor expression and Notch signalling. Cardiovasc Res 2008;78:
plugs seeded with a combination of human endothelial and cord 366 375.
blood-derived mesenchymal progenitor cells.69 For coronary angio- 16. Sugi Y, Markwald RR. Formation and early morphogenesis of endocardial endothelial
genesis, in vitro engineered three-dimensional neonatal cardiomyocyte precursor cells and the role of endoderm. Dev Biol 1996;175:6683.
17. Drake CJ, Fleming PA. Vasculogenesis in the day 6.5 9.5 mouse embryo. Blood 2000;
sheets containing preformed EC networks were transplanted onto 95:1671 1679.
infarcted rat hearts, resulting in a significantly elevated capillary 18. Eisenberg LM, Markwald RR. Molecular regulation of atrioventricular valvuloseptal
density with blood vessels originating from the cardiac patch connect- morphogenesis. Circ Res 1995;77:16.
19. Ostadal B, Schiebler TH, Rychter Z. Relations between development of the capillary
ing with host capillaries.70 wall and myoarchitecture of the rat heart. Adv Exp Med Biol 1975;53:375 388.
20. Perez-Pomares JM, Macias D, Garcia-Garrido L, Munoz-Chapuli R. The origin of the
subepicardial mesenchyme in the avian embryo: an immunohistochemical and quail-
6. Future perspectives chick chimera study. Dev Biol 1998;200:57 68.
21. De Val S, Chi NC, Meadows SM, Minovitsky S, Anderson JP, Harris IS et al. Combi-
Our understanding of embryonic and post-natal coronary vasculature natorial regulation of endothelial gene expression by ets and forkhead transcription
factors. Cell 2008;135:1053 1064.
development has brought us some way towards developing effective 22. Schmidt-Supprian M, Rajewsky K. Vagaries of conditional gene targeting. Nat Immunol
strategies to promote neovascularization in cardiovascular repair. 2007;8:665 668.
Numerous outstanding questions remain regarding the lineages that 23. Ma Q, Zhou B, Pu WT. Reassessment of Isl1 and Nkx2-5 cardiac fate maps using a
Gata4-based reporter of Cre activity. Dev Biol 2008;323:98 104.
contribute the cells of the coronary vasculature, the precise mechan- 24. Serbedzija GN, Burgan S, Fraser SE, Bronner-Fraser M. Vital dye labelling demon-
isms that govern vascular cell fate, and the regulation of arteriovenous strates a sacral neural crest contribution to the enteric nervous system of chick
identity, vessel growth, and maintenance. However, further insight and mouse embryos. Development 1991;111:857 866.
25. Ishii Y, Garriock RJ, Navetta AM, Coughlin LE, Mikawa T. BMP signals promote proe-
into vascularizing the heart during development should pave the
picardial protrusion necessary for recruitment of coronary vessel and epicardial pro-
way for significant advances in neovascular therapy. genitors to the heart. Dev Cell 2010;19:307 316.
26. Luttun A, Carmeliet P. De novo vasculogenesis in the heart. Cardiovasc Res 2003;58:
Conflict of interest: none declared. 378 389.
27. Gittenberger-de Groot AC, Deruiter MC, Bergwerff M, Poelmann RE. Smooth
muscle cell origin and its relation to heterogeneity in development and disease. Arter-
Funding ioscler Thromb Vasc Biol 1999;19:1589 1594.
P.R.R. is supported by grants from the British Heart Foundation and 28. Vrancken Peeters MP, Gittenberger-de Groot AC, Mentink MM, Poelmann RE.
Medical Research Council, and N.S. is a recipient of a British Heart Foun- Smooth muscle cells and fibroblasts of the coronary arteries derive from epithelial-
dation Intermediate Basic Science Research Fellowship. mesenchymal transformation of the epicardium. Anat Embryol (Berl) 1999;199:
367 378.
29. Olivey HE, Svensson EC. Epicardial-myocardial signaling directing coronary vasculo-
References genesis. Circ Res 2010;106:818 832.
1. Smart N, Dube KN, Riley PR. Coronary vessel development and insight towards neo- 30. Bhattacharya S, Macdonald ST, Farthing CR. Molecular mechanisms controlling the
vascular therapy. Int J Exp Pathol 2009;90:262 283. coupled development of myocardium and coronary vasculature. Clin Sci (Lond)
2. Bogers AJ, Gittenberger-de Groot AC, Poelmann RE, Peault BM, Huysmans HA. 2006;111:3546.
Development of the origin of the coronary arteries, a matter of ingrowth or out- 31. Tomanek RJ, Zheng W. Role of growth factors in coronary morphogenesis. Tex Heart
growth? Anat Embryol (Berl) 1989;180:437 441. Inst J 2002;29:250 254.
3. Mikawa T, Gourdie RG. Pericardial mesoderm generates a population of coronary 32. Yashiro K, Shiratori H, Hamada H. Haemodynamics determined by a genetic pro-
smooth muscle cells migrating into the heart along with ingrowth of the epicardial gramme govern asymmetric development of the aortic arch. Nature 2007;450:
organ. Dev Biol 1996;174:221 232. 285 288.
268 P.R. Riley and N. Smart

33. MacGrogan D, Nus M, de la Pompa JL. Notch signaling in cardiac development and 54. Yu AY, Shimoda LA, Iyer NV, Huso DL, Sun X, McWilliams R et al. Impaired physio-
disease. Curr Top Dev Biol 2010;92:333 365. logical responses to chronic hypoxia in mice partially deficient for hypoxia-inducible
34. Luna-Zurita L, Prados B, Grego-Bessa J, Luxan G, del Monte G, Bengura A et al. Inte- factor 1alpha. J Clin Invest 1999;103:691 696.
gration of a Notch-dependent mesenchymal gene program and Bmp2-driven cell 55. van Royen N, Piek JJ, Buschmann I, Hoefer I, Voskuil M, Schaper W. Stimulation of
invasiveness regulates murine cardiac valve formation. J Clin Invest 2010;120: arteriogenesis; a new concept for the treatment of arterial occlusive disease. Cardio-
3493 3507. vasc Res 2001;49:543 553.
35. Yang K, Doughman YQ, Karunamuni G, Gu S, Yang YC, Bader DM et al. Expression of 56. Zheng W, Weiss RM, Wang X, Zhou R, Arlen AM, Lei L et al. DITPA stimulates arter-
active Notch1 in avian coronary development. Dev Dyn 2009;238:162 170. iolar growth and modifies myocardial postinfarction remodeling. Am J Physiol Heart
36. Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M. The Notch target genes Circ Physiol 2004;286:H1994 H2000.
Hey1 and Hey2 are required for embryonic vascular development. Genes Dev 2004; 57. Jin Y, An X, Ye Z, Cully B, Wu J, Li J. RGS5, a hypoxia-inducible apoptotic stimulator
18:901 911. in endothelial cells. J Biol Chem 2009;284:23436 23443.
37. You LR, Lin FJ, Lee CT, DeMayo FJ, Tsai MJ, Tsai SY. Suppression of Notch signalling 58. Lobov IB, Renard RA, Papadopoulos N, Gale NW, Thurston G, Yancopoulos GD
by the COUP-TFII transcription factor regulates vein identity. Nature 2005;435: et al. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angio-
98 104. genic sprouting. Proc Natl Acad Sci USA 2007;104:3219 3224.
38. Adams RH, Wilkinson GA, Weiss C, Diella F, Gale NW, Deutsch U et al. Roles of 59. Simons M. Angiogenesis: where do we stand now? Circulation 2005;111:1556 1566.
ephrinB ligands and EphB receptors in cardiovascular development: demarcation of 60. Wills AA, Holdway JE, Major RJ, Poss KD. Regulated addition of new myocardial and
arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis. Genes epicardial cells fosters homeostatic cardiac growth and maintenance in adult zebra-
Dev 1999;13:295 306. fish. Development 2008;135:183 192.
39. Adams RH, Diella F, Hennig S, Helmbacher F, Deutsch U, Klein R. The cytoplasmic 61. Smart N, Risebro CA, Melville AA, Moses K, Schwartz RJ, Chien KR et al. Thymosin
domain of the ligand ephrinB2 is required for vascular morphogenesis but not b4 induces adult epicardial progenitor mobilization and neovascularization. Nature

Downloaded from http://cardiovascres.oxfordjournals.org/ at rouen university hospital on February 9, 2013


cranial neural crest migration. Cell 2001;104:57 69. 2007;445:177 182.
40. Foo SS, Turner CJ, Adams S, Compagni A, Aubyn D, Kogata N et al. Ephrin-B2 con- 62. Smart N, Risebro CA, Clark JE, Ehler E, Miquerol L, Rossdeutsch A et al. Thymosin
trols cell motility and adhesion during blood-vessel-wall assembly. Cell 2006;124: beta4 facilitates epicardial neovascularization of the injured adult heart. Ann N Y
161 173. Acad Sci 2010;1194:97 104.
41. Adams RH, Alitalo K. Molecular regulation of angiogenesis and lymphangiogenesis. 63. Miller-Kasprzak E, Jagodzinski PP. Endothelial progenitor cells as a new agent contri-
Nat Rev Mol Cell Biol 2007;8:464 478. buting to vascular repair. Arch Immunol Ther Exp (Warsz) 2007;55:247 259.
42. Tomanek RJ, Sandra A, Zheng W, Brock T, Bjercke RJ, Holifield JS. Vascular endo- 64. Schachinger V, Assmus B, Honold J, Lehmann R, Hofmann WK, Martin H et al. Nor-
thelial growth factor and basic fibroblast growth factor differentially modulate early malization of coronary blood flow in the infarct-related artery after intracoronary
postnatal coronary angiogenesis. Circ Res 2001;88:1135 1141. progenitor cell therapy: intracoronary Doppler substudy of the TOPCARE-AMI
43. van Groningen JP, Wenink AC, Testers LH. Myocardial capillaries: increase in number trial. Clin Res Cardiol 2006;95:13 22.
by splitting of existing vessels. Anat Embryol (Berl) 1991;184:65 70. 65. Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV et al. Repair of
44. Mikawa T. Retroviral targeting of FGF and FGFR in cardiomyocytes and coronary vas- infarcted myocardium by autologous intracoronary mononuclear bone marrow cell
cular cells during heart development. Ann N Y Acad Sci 1995;752:506 516. transplantation in humans. Circulation 2002;106:1913 1918.
45. Sho E, Komatsu M, Sho M, Nanjo H, Singh TM, Xu C et al. High flow drives vascular 66. Shimizu K, Sugiyama S, Aikawa M, Fukumoto Y, Rabkin E, Libby P et al. Host bone-
endothelial cell proliferation during flow-induced arterial remodeling associated with marrow cells are a source of donor intimal smooth-muscle-like cells in murine
the expression of vascular endothelial growth factor. Exp Mol Pathol 2003;75:111. aortic transplant arteriopathy. Nat Med 2001;7:738 741.
46. Kovacic JC, Boehm M. Resident vascular progenitor cells: an emerging role for non- 67. Tillmanns J, Rota M, Hosoda T, Misao Y, Esposito G, Gonzalez A et al. Formation of
terminally differentiated vessel-resident cells in vascular biology. Stem Cell Res 2009;2: large coronary arteries by cardiac progenitor cells. Proc Natl Acad Sci USA 2008;105:
215. 1668 1673.
47. Hibbert B, Olsen S, OBrien E. Involvement of progenitor cells in vascular repair. 68. Hobo K, Shimizu T, Sekine H, Shinoka T, Okano T, Kurosawa H. Therapeutic angio-
Trends Cardiovasc Med 2003;13:322 326. genesis using tissue engineered human smooth muscle cell sheets. Arterioscler Thromb
48. Sane DC. The endothelial life insurance plan. Blood 2004;104:2615 2616. Vasc Biol 2008;28:637 643.
49. Smart N, Riley PR. The stem cell movement. Circ Res 2008;102:1155 1168. 69. Melero-Martin JM, De Obaldia ME, Kang SY, Khan ZA, Yuan L, Oettgen P et al. Engin-
50. Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and eering robust and functional vascular networks in vivo with human adult and cord
mechanisms. Circ Res 2007;100:158 173. blood-derived progenitor cells. Circ Res 2008;103:194 202.
51. Grover-Paez F, Zavalza-Gomez AB. Endothelial dysfunction and cardiovascular risk 70. Sekine H, Shimizu T, Hobo K, Sekiya S, Yang J, Yamato M et al. Endothelial cell cocul-
factors. Diabetes Res Clin Pract 2009;84:1 10. ture within tissue-engineered cardiomyocyte sheets enhances neovascularization and
52. Ross R. Atherosclerosisan inflammatory disease. N Engl J Med 1999;340:115 126. improves cardiac function of ischemic hearts. Circulation 2008;118(Suppl. 14):
53. Schaper W. Collateral circulation: past and present. Basic Res Cardiol 2009;104:5 21. S145 S152.

You might also like