You are on page 1of 12

ATVB In Focus

Developmental Biology in the Vasculature


Series Editor: Mark Majesky, PhD

Mechanisms of Vessel Branching


Filopodia on Endothelial Tip Cells Lead the Way
Frederik De Smet, Inmaculada Segura, Katrien De Bock, Philipp J. Hohensinner, Peter Carmeliet

Abstract—Filopodia, “the fingers that do the walking,” have been identified on endothelial cells at the tip of sprouting
vessels for half a century, but the key role of the tip cell in vessel branching has been recognized only in the past few
years. A model is emerging, whereby tip cells lead the way in a branching vessel, stalk cells elongate the sprout, and
a very recently discovered phalanx cell ensures quiescence and perfusion of the newly formed branch. Recent genetic
studies have shed light on the molecular signature of these distinct endothelial phenotypes; this provides a novel
conceptual framework of how vessel morphogenesis occurs. Here, we will discuss the molecular candidates that
participate in the decision of endothelial cells to adapt these distinct fates and highlight the emerging insights on how
these cells send out filopodia while navigating. (Arterioscler Thromb Vasc Biol. 2009;29:639-649.)
Key Words: endothelial tip cells 䡲 stalk cells 䡲 phalanx cells 䡲 filopodia 䡲 sprouting angiogenesis

M uch has been learned about the importance of angio-


genesis in health and disease, but our understanding of
how vessels branch remains incomplete. An exciting break-
depends, to a substantial extent, on the organization of their
cytoskeleton; for instance, for an EC to migrate, it needs to form
spike-like filopodia, fan-like lamellipodia, and polarize its actin
through in the field has been the recognition that several types cytoskeleton in the direction of migration.
of specialized endothelial cells (ECs), each with distinct
cellular fate specifications, are required to build a functional Endothelial Tip Cells
branch.1 The first and, to a certain extent, the most undertak- Even though the existence of “filliform processes” at the tip
ing cell in a vessel branch is the “tip cell,” which leads the of endothelial sprouts was already observed in the brain
way. With their continuously searching filopodia, these tip almost half a century ago,6 the concept and importance of
cells sense and respond to guidance cues in their microenvi- vessel guidance was not fully appreciated until recently.7,8
ronment, similar to how an axonal growth cone in the nervous Classical studies have documented the existence of “seamless
system2 or an epithelial tip cell in the fruitfly airway system3 ECs” in vivo and postulated the existence of different
explores its surroundings. Even in the sea squirt, tip cells use endothelial fates (trunk cells with lumen and tip cells without
filopodia to build a primitive vascular network.4 It is therefore lumen) on growing sprout capillaries.9 Electron microscopy
not surprising that several classes of molecules and principles, studies further described the existence of filopodia at the
used by navigating axons or epithelial cells, are evolutionary leading edge of growing capillaries (Figure 1a and 1b).10
conserved and shared, and even might have been coopted by the Tip cells have now been observed in various models of
migrating endothelial tip cell.1,2 “Stalk cells” trail behind the tip sprouting angiogenesis. Key features of this cell are their
cell and elongate the stalk of the sprout; they proliferate, form location at the forefront of vessel branches, highly polarized
junctions, lay down extracellular matrix, and form a lumen. nature, and numerous filopodia probing the environment,
“Phalanx cells” are the most quiescent ECs, lining vessels once while migrating toward an angiogenic stimulus.7,11 They do
the new vessel branches have been consolidated; they form a not form a lumen and, with some exceptions, proliferate
smooth cobblestone monolayer and are aligned as in a phalanx minimally (Figure 1c and 1d).7,12,13 Tip cells have a specific
formation of the ancient Greek soldiers, are covered by peri- molecular signature, characterized by the expression of
cytes, stick to each other via tight junctions, are embedded in a VEGF receptor (VEGFR) 2, VEGFR3, platelet-derived
thick basement membrane, and stay foot. These cells are growth factor (PDGF)-BB, Unc5B, Delta-like ligand-4
engaged in optimizing blood flow, tissue perfusion, and oxygen- (Dll4), neuropilin-1 (NRP1), and others (see below).7,14 –18
ation.5 Tip, stalk, and phalanx ECs each have a specialized These cells detect gradients of navigatory cues and integrate
function in vessel branching. How these cells execute their job combinatorial molecular codes into directional migration.

Received January 23, 2009; revision accepted February 23, 2009.


From the Vesalius Research Center, VIB, K.U. Leuven, Belgium.
Correspondence to Peter Carmeliet, MD, PhD, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000, Leuven,
Belgium. E-mail peter.carmeliet@vib-kuleuven.be
© 2009 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.109.185165

639
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
640 Arterioscler Thromb Vasc Biol May 2009

lularly, generating the Notch intracellular domain (NICD)


that acts as a transcriptional regulator.21 Tip cells are exposed
to the highest levels of VEGF, which induces the expression
of Dll4 in these cells.21 Dll4 binds to Notch on neighboring
(future stalk) ECs and downregulates VEGFR2 signaling; this
dampens the VEGF-induced expression of Dll4 in these cells,
thereby establishing a self-reinforcing feedback that allows
the leading cell to gain and retain its tip position, while
preventing the follower neighbors from leaving their position
in the stalk. Genetic mosaic studies reveal that Notch regu-
lates EC specification by actively suppressing the tip cell
phenotype in stalk cells.19
A number of questions about the mechanisms of Dll4 and
Notch remain unanswered. For instance, it is unknown
whether Dll4 reverse-signaling prevents the tip cell from
becoming a stalk cell by inactivating Notch signaling in this
cell through internalization of the receptor.22 Another ques-
Figure 1. Vascular sprouts are guided by endothelial tip cells. a, tion is whether Dll4 induces cytoskeletal rearrangements in
Original picture showing an embryonic endothelial tip cell
obtained by electron microscopy (reprinted with permission the tip cell. Notch ligands such as Dll4, Dll1, and Jagged1
from10). b, Confocal micrograph showing filopodia extensions at contain a PDZ-binding motif, which facilitates interactions
the leading tip cell (reprinted with permission from7). c, Scheme with adaptor proteins, thereby mediating adhesion and migra-
from 1972 proposing several endothelial subtypes in the angio-
tion in the ligand-expressing cell.23 Such adaptor molecules
genic sprout (reprinted with permission from9). d, Schematic
representation of a tip cell (green) extending filopodia toward an include Dlg1, MAGI proteins, and syntenin, which interact
angiogenic stimulus (red gradient), followed by stalk cells (pur- with the cytoskeleton.23
ple), while phalanx cells (gray) remain quiescent.
Lamellipodia and Filopodia Lead the Way
Central to their phenotype is the formation of filopodia. We The key job of endothelial tip cells is to navigate, a process
will first describe how tip cells are selected in the nascent that relies on correct probing of microenvironmental cues,
vessel branch, and explain why ECs do not move as a sheet and translating them into a dynamic process of adhesion (at
but form a sprout in response to an angiogenic signal. the front) and deadhesion (at the rear), that ultimately leads to
cell movement. Therefore, the tip cell forms lamellipodia and
Induction of Tip Cell Formation by VEGFR2 filopodia. Surprisingly, however, relatively little is known
and Dll4 about the processes regulating the assembly of these cellular
Genetic studies show that a VEGF gradient is important in the protrusions in ECs. We will therefore briefly overview some
process of selection and induction of the endothelial tip cell.7 key insights of this process, as deduced from studying the
Via binding to VEGFR2, VEGF induces a signaling cascade, axon growth cone, and thereafter discuss our current under-
which enables one EC to take the lead and become a tip cell, standing of these processes in ECs.
while its neighbors are prevented from doing so and, instead, Lamellipodia are short veil-like structures in close prox-
are instructed to become stalk cells. Such lateral inhibition imity to the plasma membrane that contain a highly branched
relies on tip-to-stalk cell communication by Dll4/Notch actin network. Filopodia, on the other hand, consist of long
signaling (Figure 2a).18 –20 ECs express various Notch recep- spiky plasma membrane protrusions containing tight parallel
tors (Notch1, 3, 4) and ligands (Dll1, Dll4, Jagged1, bundles of filamentous actin (F-actin), which usually extend
Jagged2).21 After ligand binding, Notch is cleaved intracel- from lamellipodia (Figure 3a).24 –26 Filopodia and lamellipo-

a Role of VEGF and Dll4 b Cytoskeletal rearrangements c Inflammatory cytokines


in tip cell formation induce tip cell formation
VEGF VEGF
Figure 2. Molecular pathways of tip cell signaling.
VEGFR2 VEGFR2
FGF2 Angiostatin TNFα a, VEGF signaling via VEGFR2 enables tip cell for-
VEGFR3 FGFR mation (green), whereas Dll4 signaling induces
NRP1 Amot Netrin Bradykinin stalk cells (purple). VEGFR3 is upregulated,
Integrins TNFR
whereas Neuropilin 1 (NRP1) is maintained. b, Dif-
Unc5B ferent signaling cascades converge to small
SRF CD13
MMP GTPases activation, thereby regulating filopodia
Dll4 induction Jagged1 and lamellipodia formation in endothelial cells. c,
active GTPase induction
active Inflammatory cytokines (blue gradient), including
Filopodia, GTPase tumor necrosis factor ␣ (TNF␣) and bradykinin,
Lamellipodia, also induce a tip cell phenotype.
?
Cell migration
Dll4 Jagged1
Notch Notch

Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015


De Smet et al Mechanisms of Vessel Branching 641

a Stress fibers Lamellipodia Filopodia


active
small GTPase RhoA Rac1 Cdc42

Exta Cellular
Matrix

Focal
contact point

b Figure 3. Overview of the filopodia/la-


mellipodia machinery during cell migra-
G-actin
F-actin tion. a, Schematic representation of fila-
Negative end Positive end mentous actin in filopodia (red),
(-) Cofilin Profilin, lamellipodia (blue), and stress fibers
(+)
(green). Cellular protrusions anchor in
Formin,
the extracellular matrix by focal contact
ARP2/3 Ena/Vasp points (brown). b, Regulation of filamen-
Wasp tous (F-actin) and globular (G-actin) actin
Retraction through Elongation through
dynamics. c, Guanine nucleotide
de-polymerization and Branching of F-actin to polymerization
exchange factors (GEFs) and GTPase
retrograde flow generate meshworks for filopodia extension activating proteins (GAPs) regulate GTP-
bound (active) and GDP-bound (inactive)
RhoA, Rac1, and Cdc42 states, respec-
c Guanine nucleotide d RhoA Rac1 Cdc42 tively. d, RhoA, Rac1, and Cdc42 down-
exchange factor stream signaling regulates actin
(GEF) dynamics.
+GTP ROCK PAK WASP-
ARP2/3
small GTPase small GTPase
LIMK
GDP GTP
Pi branching
GTPase Cofilin
activating protein
(GAP)
de-polymerization
inactive active

dia are highly dynamic structures, generated within minutes ments into parallel bundles, which are cross-linked by mem-
after stimulation.27 Both structures are capable of probing the bers of the Fascin protein family.24 Coincidently, several
environment, thereby sensing the presence of attractive or other proteins, including members of the Inverse (I)-BAR
repulsive guidance cues.26 An attractive cue will induce domain such as Insulin receptors substrate p53 (IRSp53),
F-actin polymerization thereby extending filopodia, whereas prepare the plasma membrane by inducing bulging of its
a repulsive cue causes depolymerization and retrograde actin surface.24
flow, resulting in retraction.26 Filopodia and lamellipodia also The main regulators of filopodia and lamellipodia forma-
adhere and form focal contact points to connect the cytoskel- tion are members of the Rho small GTPases, of which RhoA,
eton to the extracellular matrix (ECM).27 This allows stress Rac1, and Cdc42 have been most extensively studied (Figure
fibers of actin/myosin filaments to pull the cell toward these 3c).25,26 These molecules are activated by binding of GTP
anchors, and induce forward movement.27 nucleotides, supplied by Guanine nucleotide Exchange Fac-
F-actin fibers are elongated at their ‘barbed’ or positive end tors (GEFs). Inactivation occurs via their intrinsic GTPase
by proteins of the Profilin, Ena/Vasp, and Formin families, activity, which converts GTP into GDP; this process is
which promote polymerization of G-actin into long F-actin stimulated by GTPase Activating Proteins (GAPs). RhoA,
strings in proximity of the plasma membrane (Figure 3b).24,25 Rac1, and Cdc42 are activated in response to stimulation of
Whereas Profilin and Formin directly bind to G-actin and many membrane receptors, including receptor tyrosine
present it to the extending fiber, Ena/Vasp functions as an kinases and G protein– coupled receptors.26 Cdc42 and Rac1
anticapping protein keeping the barbed end clear of inhibitory regulate filopodia and lamellipodia formation, respectively,
cap-proteins.25–27 Elongation of these filaments pushes the through activation of p21-activated kinase (PAK); RhoA is
leading edge forward and promotes cell migration. At the involved in adhesion and forward movement through regula-
“pointed” or negative end of the actin-filament (pointing tion of stress fiber formation via the Rho-associated serine-
toward the inside of the cell), Cofilin depolymerizes and threonine protein kinase (ROCK; Figure 3d).25–27 PAK and
shortens the actin string.25–27 Branching of F-actin filaments ROCK activate LIM-kinase, which, through inhibition of
is mediated by the Actin-related protein-2/3 (ARP2/3) com- Cofilin, blocks F-actin depolymerization.26 Activated Cdc42
plex and members of the Wiscott-Aldrich Syndrome protein and Rac1 also interact with the WASP:ARP2/3 complex,
(WASP) family.25,26 Myosin X groups these growing fila- thereby inducing F-actin branching.25 Filopodia extension is
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
642 Arterioscler Thromb Vasc Biol May 2009

also regulated by targeting Ena/Vasp proteins to the mem- lamellipodia. Water entry increases local hydrostatic pres-
brane at the leading edge of migrating cells.26 Once extended, sure, producing cell membrane expansion to form a protru-
filopodia make contact with ECM components such as sion, thereby enhancing lamellipodia dynamics.41 According
laminin, fibronectin, and collagen through integrins on their to this model, actin repolymerization thereafter stabilizes the
surface that initiate the formation of focal contact points.27 An protrusion.
important regulator of this process is the focal adhesion
kinase (FAK).28 Hence, signals from integrins and growth Modulation of VEGF Signaling in Tip Cells
factor receptors are transduced to the cytoskeleton in these Besides VEGFR2, tip cells also express other VEGF recep-
filopodia.28 tors, including VEGFR3 (also known as Flt4) and NRP1. In
development, VEGFR3 is present in endothelia, but becomes
Formation and Regulation of Filopodia in largely restricted to the lymphatic endothelium in adult-
Endothelial Tip Cells hood.16 However, in active vascular endothelia, VEGFR3
To navigate, tip cells become polarized, so that their leading reappears42 and its expression is mainly confined to filopodial
front extends filopodia, whereas their rear maintains contact extensions on tip cells at the sprouting front.16 Blocking
with trailing stalk cells to avoid branch desintegration. How VEGFR3 reduces the number of sprouts and branch points
VEGF induces these processes is poorly understood. Based and EC proliferation. VEGFR2 signaling induces VEGFR3
on experiments in fibroblasts and neurons, Rho small GTPase expression in tip cells, whereas Notch downregulates its
proteins have been identified as key regulators of cell expression in stalk cells.16 As is the case for VEGFR2, tip
migration and morphogenesis.25–27 Increasing evidence sug- cells express higher levels of VEGFR3 than stalk cells.
gests that these molecules also act downstream of VEGFR2 VEGFR2 and -3 are able to form heterodimers, and may
to regulate the formation of filopodia, lamellipodia, and stress transmit distinct signals than receptor homodimers.43 An
fibers, and consolidate adhesion in ECs as well. Indeed, in intriguing possibility is that such VEGFR2/3 heterodimers
response to VEGF, Cdc42 triggers the formation of filopodia may be involved in tip cell functions. VEGFR3 also regulates
and regulates cell polarization through microtubule organiza- vessel integrity, though it is unknown whether this is an effect
tion, whereas Rac1, together with PAK, controls lamellipodia mediated via tip or, more likely, via stalk cells.42
formation in response to VEGF.29,30 Loss of Rac1 results in NRPs function as endothelial receptors for members of the
early embryonic lethality attributable to defective vessel Sema3 and VEGF families.2,44 As VEGF receptors, NRPs are
branching.31 RhoA induces stress fiber formation and medi- essential for cardiovascular development and tumor angio-
ates EC permeability, migration, and stabilization of capillary genesis.44 Both NRP1 and NRP2 are detected in the devel-
tubes in response to VEGF.30,32,33 Shear stress also increases oping vasculature. The use of an anti-NRP1 antibody engi-
RhoA activity, and the formation of stress fibers, focal neered to bind solely VEGF, and not Sema3s, has revealed
adhesions, and junctional complexes.34 Inhibition of RhoA that Sema3s have little effect on VEGF/NRP1-driven vascu-
impairs EC migration and tube formation, while a dominant lar development.44 However, Sema3F is a potent inhibitor of
negative RhoA protein inhibits angiogenesis.35 As in other tumor angiogenesis, progression, and metastasis.44 NRPs
cells, Cdc42 activates Rac1 in ECs.30 either form receptor complexes with VEGFR1, -2, or -344,45
For Rac1 to promote EC migration, it needs to be activated or induce direct signaling through synectin (also known as
by the GEF Vav2.36 In vitro, silencing of Vav2 impairs NIP), a PDZ domain-containing protein that acts as a scaffold
VEGF-induced EC migration, which is in line with its for downstream signaling cascades. Genetic studies show that
function in neurite outgrowth,36 whereas Vav2 knockout mice NRP1 and synectin regulate vessel branching and morpho-
have cardiovascular defects.37 Wave2, a member of the genesis.44,46 In the absence of NRP1, sprouts in the embryonic
WASP proteins, is also required for actin reorganization and hindbrain do not branch and fuse into a vascular plexus,15
EC movement38: loss of Wave2 impairs vessel branching and while blocking the binding of VEGF to NRP1 prevents
the formation of EC lamellipodia. Single deficiency of each vascular remodeling.44 The defective vessel branching and
of the Ena/VASP proteins, which promote actin polymeriza- fusion were not caused by the absence of tip cells or filopodia
tion, results in relatively subtle phenotypes, but triple defi- but were the result of defects in lateral filopodia extension,
ciency disrupts vessel integrity and endothelial barrier func- which is critical for turning and fusion of tip cells.15 How
tion.39 Several components of the machinery responsible to NRP1 regulates tip cell filopodia and in particular their lateral
generate and consolidate focal adhesion points in other cells extensions remains to be further explored. NRP1 induces
is also conserved in ECs, and is, in part, mediated by filopodial extension and navigation of tumor cells and neu-
integrins.28,40 Moreover, FAK regulates EC migration in rons, though this activity is context-dependent. In tumor cells,
response to VEGF, whereas deficiency of FAK in ECs leads filopodia formation in response to NRP1/VEGFR2 signaling
to vessel defects and regression.40 is mediated via Cdc42 activation.47
Genetic studies reveal that Aquaporin-1 (AQP1), a water
channel protein increasing water permeability in ECs, regu- Novel Regulators of Tip Cell Formation
lates EC migration.41 As a consequence of local actin depo- Only a few other molecules have been identified to induce
lymerization and transmembrane ionic fluxes, the cytoplasm filopodia formation in ECs (Figure 2c). Inflammation is
adjacent to the leading edge of migrating cells undergoes known to activate ECs and stimulate vessel branching. Brief
rapid changes in osmolality. AQPs, polarized to lamellipodia, exposure to the inflammatory cytokine TNF␣ primes ECs for
facilitate osmotic water flow across the plasma membrane in angiogenic sprouting by inducing a tip cell phenotype and
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
De Smet et al Mechanisms of Vessel Branching 643

expression of the tip cell markers PDGF-BB and VEGFR2.48 Another novel player is the Serum Response Factor (SRF)
Furthermore, the Notch ligand Jagged1 was induced in tip transcription factor, which interacts with other transcription
cells via a NF-kB dependent mechanism, raising the question factors including members of the ETS and GATA families.54
whether Jagged1 might have a similar role in tip cell Late in development, SRF expression becomes confined to
induction in pathological (inflammatory) angiogenesis as ECs in small vessels, more precisely in tip and stalk ECs.54
Dll4 has in physiological conditions.48 Another inflammatory Conditional endothelial SRF-deficient embryos succumb be-
mediator, bradykinin, induces filopodia formation in ECs cause of reduced vessel branching. This is not attributable to
through activation of the Cdc42 pathway, a process which is a reduced number of tip cells or to abnormal expression of
modulated by the transmembrane peptidase CD13. 49 Notch1 or Dll4 but to the presence of thinner and fewer
Sphingosine-1-phosphate (S1P), an antiinflammatory phos- filopodia per tip cell, with a disorganized actin structure at the
pholipid derivative which has angiogenic and chemo- base of each filopodium. This abnormal actin organization
attractant properties,50 has also been involved in the forma- also occurs in stalk cells, suggesting a fundamental role in
tion lamellipodia on ECs. It is released from activated cytoskeletal rearrangements. In vitro, loss of SRF-function
platelets, and exerts its function on ECs via binding to its results in aberrant EC migration and tube-formation (Figure
S1P-G protein– coupled receptors (S1P1–3). S1P1 signaling 2b) attrituable to decreased F-actin formation. SRF has been
is involved in the translocation of the Arp2/3 complexes from linked to RhoA signaling, with which it cooperates in
an intracellular location to the cellular migratory front, regulating transcription of ß-actin. Moreover, loss of SRF
resulting in the formation of lamellipodia.50 This process is reduces VE-Cadherin expression, which could explain the
dependent on Cdc42 and Rac1 activation.50 loss of vessel integrity and stability.54 Another regulator of
Genetic studies in the Drosophila airway system reveal EC filopodia formation is the Melanoma-associated antigen
that fibroblast growth factors (FGFs) determine the specifi- (MAGE) D1, which was initially identified as a cell surface
cation of an epithelial tip cell.3 Indeed, the FGF homologue antigen expressed by tumor cells. Overexpression of
Branchless, which binds to its receptor Breathless on tracheal MAGE-D1 inhibits EC migration and adhesion, and disrupts
cells promotes outgrowth of terminal branches. Airway tip cytoskeletal rearrangements and lamellipodia formation.55
cells respond directly to Branchless and lead branch out- Not surprisingly, some classical axon guidance molecules
growth, whereas trailing stalk cells receive a secondary signal also regulate tip cell navigation. The Netrin guidance receptor
to follow the lead cells and form a tube. These roles are not Unc5B is also expressed by endothelial tip cells.17 Binding of
genetically prespecified; rather, there is competition between netrin-1 to Unc5B induces collapse of filopodia in the
cells such that those with the highest FGFR activity take the developing retina (Figure 2b), whereas knockdown of Unc5B
tip position, whereas those with less FGFR activity assume induces ectopic filopodial extensions.17 In Dll4 heterozygous-
subsidiary positions and form the branch stalk. Competition deficient mice, an increase in Unc5B-positive tip cells was
appears to involve Notch-mediated lateral inhibition that also observed.18 The bidirectional signaling system of trans-
prevents extra cells from assuming the lead,3 analogous to the membrane Eph receptors and ephrin ligands is involved in
endothelial system.1 Whether FGFR levels also determine the arterial/venous boundary formation in the embryo.1 Ephrin/
specification of an endothelial tip versus stalk cell remains to Eph family members are also reciprocally expressed between
be determined.1 Interestingly, in a mixed EC population, in blood vessels and their surrounding tissues,1 and complemen-
which FGF receptor-1 (FGFR1) was silenced in half of the tary ligand/receptor expression patterns provide guidance
cells, receptor-expressing cells pioneered migration in re- cues during vascular development, similar to the mechanism
sponse to FGF2, whereas silenced cells were unable to take described for neuronal development.2 In the process of
the lead but, nonetheless, were still capable of trailing behind spinogenesis, motile dendritic filopodia explore their envi-
the leader.51 These findings suggest that FGFRs may regulate ronment to contact with the appropriate presynaptic partner,
endothelial tip cell migration as well (Figure 2b). while EphB forward signaling is required for filopodia
Angiomotin (Amot), a membrane-associated protein that motility and synaptogenesis.56 All these observations raise the
binds angiostatin, is expressed by activated endothelia, and possibility that Ephrin/Eph signaling or other molecules
controls EC migration via its PDZ binding motif.52 Trans- involved in this neuronal process might also regulate endo-
genic mice expressing a PDZ-deficient Amot in ECs lose thelial filopodia formation.
their response to growth factors, which leads to vessel
defects.52 Amot deficiency in mouse and zebrafish also The Invasiveness of the Tip Cell
impairs vessel branching and integrity, with defective filop- Sprouting angiogenesis is an invasive process that requires
odia formation, and stalling of sprouting intersegmental proteolytic degradation of the extracellular matrix (ECM).
vessels (ISVs; Figure 2b).52 Although Amot does not affect Tip ECs express matrix metalloproteinases (MMPs), and their
VEGF-induced EC survival or proliferation, it renders ECs expression is regulated by the composition of the surrounding
unresponsive to chemotactic stimuli. The subcellular local- ECM they have to invade (Figure 2b). Membrane type-1
ization of Amot in lamellipodia and its ability to bind cell MMP (MT1-MMP; also known as MMP14) regulates angio-
polarity proteins indicate that it is required for cytoskeletal genesis12 and appears indispensable for ECs to form invading
reorganization during migration.52 Moreover, Syx, a RhoA channels.33,57 There is ample evidence that MT1-MMP is
GEF, was recently identified as an important PDZ interacting present at the leading tip of invading ECs,12,57 while its
partner of Amot, regulating the activity of RhoA in the expression is downregulated in stalk cells during vessel
leading front of the migrating cell.53 stabilization and maturation as a result of an endothelial-
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
644 Arterioscler Thromb Vasc Biol May 2009

a Notch signaling induces b Mechanisms of lumen formation


a stalk cell phenotype
?
Dll4 pinocytosis

Figure 4. Molecular pathways of stalk


coalescence
cell signaling. a, A stalk cell (purple) is
Dll1Jagged1 induced by lateral inhibition through Dll4/
Notch1
Notch signaling, which reduces the
NICD expression levels of VEGFRs and NRP
and, via Nrarp, stabilizes Wnt signaling.
Intra-cellular Inter-cellular Other Notch ligands (Dll1 and Jagged1)
are also present. b, Schematic repre-
c “Push” model of vessel sprouting sentation of intracellular and intercellular
Flt1 VEGFR2/3 models of lumen formation. c, Perivas-
Nrarp NRP1 EGFL7+/+ EGFL7-/- cular EGFL7 deposition (blue) in the
Wnt angiogenic sprout regulates vascular
morphogenesis.
Tip cell
Stalk cell
Phalanx cell
VEGF
Cell-cell EGFL7
contact

pericyte crosstalk.12 The role and need for MMPs in vessel which are instructed to become stalk cells (Figure 4a).1,19
branching may vary. Indeed, vessels in the adult are embed- Notch signaling in stalk cells dampens the migratory response
ded in a thick basement membrane (rich in type IV collagen to VEGF, impairs filopodia extension, and, overall, inhibits
and laminin), whereas the basement membrane is merely a vessel branching by lowering the expression of VEGFR2,
thin layer or even absent during development, which might VEGFR3, NRP1, and CXCR4.16,62 Hence, inhibition of
explain why loss of MT1-MMP does not impair vascular Notch signaling increases vessel branching, by promoting a
development in the embryo.12 These findings thus suggest switch from stalk to tip cell differentiation and stimulating
that degradation of the basement membrane is not limiting migration and proliferation of tip cells, in the mouse retina
vessel branching in the embryo, but a more critical hurdle in and hindbrain,18,19,63 zebrafish embryo,14 and tumor mod-
the adult. els.20,64,65 Notch-regulated ankyrin-repeat protein (Nrarp) is a
ECM components are often used by tip cells as scaffold to downstream target of Notch that counteracts Notch signaling
navigate.58 Also, on arrest of VEGF-targeted therapy, vessels by destabilizing NICD. Recent studies reveal that Nrarp is
regrow alongside ghost tracks of basement membrane.59 expressed in stalk cells at branch points, where it overcomes
Integrin receptors induce cytoskeletal rearrangements, each the activity of Notch to induce cell cycle arrest and quies-
with specific effects; for instance, ␣5ß1 and ␣vß3 (both cence.66 At the same time, Nrarp stimulates Wnt signaling in
receptors of fibronectin) differentially regulate activation of stalk cells, which stabilizes the stalk and prevents EC retrac-
Cofilin and thereby also EC migration (Figure 2b).60 Lami- tion in part via tightening EC junctions.66
nins are heterotrimeric protein complexes, which are pro- Specification of the tip/stalk cell phenotype by Notch is
duced by both tip and stalk cells and deposited as pericellular likely more complex. Indeed, Dll4 is not the only Notch
matrix.61 In the absence of laminin ß1 or ␥1 subunits, vessel ligand expressed in sprouting vessels. Notably, loss of Dll4,18
sprouts fail to form normally.61 When vessels branch, EC tip Jagged1, or Dll121 each results in distinct vascular defects,
cells are exposed to different ECM components than those indicating that these 3 ligands are not functionally redundant.
present in the basement membrane; some of these matrix Also, expression of these components is nonoverlapping in
molecules also stimulate sprout formation.58 postnatal retinal vessel development: Dll4 is the only
ligand expressed in tip cells, whereas Jagged1 and Dll1 are
Endothelial Stalk Cells present in stalk cells.21 Soluble Jagged1 reduces tip cell
A second endothelial subtype, called “stalk cell,” trails number, filopodia, and vessels density.19,67 An outstanding
behind the leading tip cell. Their task is to proliferate, question is whether Jagged1 instructs cells at the vascular
elongate the stalk, form a lumen, and connect to the circula- front to become or remain tip cells, possibly via regulating
tion.7,11 In contrast to tip cells, stalk cells do not extend Notch signaling in this cell. Differences in Notch signaling
filopodia.7 by distinct ligands may fine-tune vessel branching, as
occurs in other cellular systems, such as in lymphocyte
Induction of Stalk Cells by Notch and Wnt development.68
As explained above, lateral inhibition via Dll4/Notch under- Dll4/Notch signaling upregulates the expression of genes
lies the selection of a tip cell at the expense of its neighbors, that influence EC migration and adhesion, such as VCAM1,
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
De Smet et al Mechanisms of Vessel Branching 645

the RhoGTPase RND1, or the RasGEF RAPGEF5.69 Dll4/ Robo4 Is Necessary for Stalk Cell Stabilization
Notch also promotes cell adhesion by activating ß1-integrins Originally discovered in Drosophila neurons as receptors for
through NICD transcription-independent mechanisms.70 axon guidance cues, members of the Roundabout (Robo)
Further, Notch1 stimulation by Jagged1 induces microtu- receptor family are also expressed by ECs. The Slit/Robo
bule stabilization in neurons,71 while promoting cell– cell system in vertebrates consists of three Slit proteins (Slit1–3),
over cell–ECM interactions in fibroblasts72; if this would which bind to Robo receptors (Robo1– 4). Robo4 is structur-
be also the case in ECs, Notch would hereby promote stalk ally divergent from other Robo receptors and primarily
stabilization and prevent stalk cell retraction. Interestingly, expressed by ECs, in particular in microvessels.79 Robo4
the ECM protein Microfibril-Associated Glycoprotein-2 activates pathways involved in cell migration,79 including
(MAGP-2) also induces actin rearrangements and vessel WASP and Ena/Vasp family members in ECs.80 The role of
branching through antagonizing Notch signaling in ECs.73 Robo4 in EC migration is, however, debated. Some reports
describe a role in repulsion,81,82 whereas others suggest a
Fine Tuning of Stalk Morphogenesis by sFlt1 promigratory effect.83,84 Genetic studies further show that
VEGFR1 (Flt1) and its soluble form, sFlt1, act as a trap for Robo4 stabilizes nascent vessels during postnatal retinal
VEGF, VEGF-B, and PlGF.74 Both endothelial tip and stalk angiogenesis.85 Robo4 is absent from most tip cells, but
cells express VEGFR1 during retinal vascular development.7 expressed by stalk cells, where activation by Slit2 would
VEGFR1 and its soluble form are upregulated on Dll4-acti- downregulate VEGF signaling and inhibit activation of Src-
vated Notch-signaling in stalk cells,69 yet neutralization of kinase and Rac1 in this model. Also, Slit2 inhibits EC
VEGFR1 does not affect vessel branching in vivo.7 Also, migration, tube formation, and permeability, whereas vessels
␥-secretase, which proteolytically activates Notch in stalk in the ischemic retina are more leaky and unstable in Robo4
cells, cleaves membrane-anchored VEGFR1: release of its mutant mice.85 However, it remains debated whether Slit2 is
intracellular domain has been proposed to inhibit VEGFR2 the ligand of Robo4. Indeed, overexpression studies show
signaling, while shedding of its extracellular sFlt1 domain that Slit2 can be immunoprecipitated with Robo4,81 but others
may further dampen VEGF by acting as a trap.75 All these did not observe binding of Slit2 to Robo4.83 Recently, Robo4
was reported to form heterodimers with Robo1, which in-
effects may potentially reduce or spatially restrict the overall
creases the formation of filopodia in ECs.80 Thus, unraveling
response of ECs to VEGF through sequestration of extracel-
the role of Robo-signaling in tip/stalk cells requires further
lular VEGF. Although PlGF may activate ECs via signaling
investigation.
through membrane-anchored VEGFR1 in pathological con-
ditions,74 it remains to be defined whether this involves an
Stalk Cells and Lumen Formation
effect on tip and/or stalk cells.
After tip cell induction and stalk elongation, a vessel branch
needs to generate a lumen and initiate blood flow. Most of our
Stalk Cells Maintain Vessel Integrity
conceptual advances in lumen-formation have been generated
To elongate the stalk, stalk cells must divide, maintain
in vitro, by studying the behavior of ECs in two- or three-
contact with the leading tip cell, and form a lumen. Similar to
dimensional gels.33 In these models, lumenogenesis relies on
tip cells, maintenance of a stalk cell phenotype requires
the formation and coalescence of intracellular pinocytic
cytoskeletal restructuring. However, the molecular (down-
vacuoles. In zebrafish embryos, intra- and intercellular fusion
stream) mechanisms of how trailing cells remain in contact
of vacuoles generates a lumen (Figure 4b).86 However, recent
with the leading front remain mostly unexplored. VE- findings suggest that the lumen in zebrafish vessels is formed
Cadherin is important to maintain cell– cell contacts, as its by the arrangement of ECs around an intercellular lumen.13
absence induces random nondirectional migration of discon- Vacuole formation depends on an interaction of integrins
nected cells.51 Cadherins do not form rigid and fixed struc- with the ECM, whereas fusion of vacuoles requires rearrange-
tures, but rather exhibit a flow-like movement on reorgani- ment of the actin and microtubule cytoskeleton.33 EC vacu-
zation of the cytoskeleton.76 Indeed, when epithelial cells oles accumulate in a polarized pattern, adjacent to the
move, cadherin clusters rapidly form at the leading edge, centrosome.87 Both Rac1 and Cdc42, but not RhoA, localize
while they are absorbed at the rear end. As mentioned above, to vacuole membranes during formation of a lumen.33,87
Nrarp-induced Wnt signaling is also important for stabiliza- Downstream signaling includes members of the WASP,
tion of the stalk and preventing EC retraction by improving PAK, polarity proteins, and protein kinase C (PKC).33,87
intercellular junctions.66 FGF signaling has also been impli- However, an inhibitor of ROCK activity (which is down-
cated in vascular homeostasis and integrity through tighten- stream of RhoA) impairs vacuole formation,88 suggesting that
ing of EC junctions.77 Besides the above described “pull” in some conditions, RhoA may regulate lumen formation.
model, whereby the tip cell pulls stalk cells in the sprout, Additionally, activation of endothelial RhoA-GTP on loss of
recent data also provide evidence for a “push” model (Figure Cerebral Cavernous Malformations 2 (CCM2) causes cy-
4c), whereby dividing stalk cells push the sprout forward. toskeletal changes and impairs lumen formation.89 Once the
Indeed, in the absence of the vascular-specific secreted factor lumen has formed, additional signals drive EC proliferation,
EGFL7, an ECM protein mainly secreted by stalk cells, growth, or enlargement, modifying the lumen size. Such
newly formed ECs at the base of the stalk accumulate in effects have been reported for different isoforms of VEGF,11
enlarged, but nonelongating sprouts.78 Notch1,67 and laminin.61
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
646 Arterioscler Thromb Vasc Biol May 2009

a behavior.5 Hence, by lowering the activity of an oxygen


Phd2+/+ b Phd2+/-
sensor, ECs readjust their shape and phenotype to improve
oxygen delivery in case of its shortage.
The molecular mechanisms underlying the endothelial
phalanx phenotype remain to be largely explored. Unlike tip
cells, phalanx cells extend few filopodia and migrate poorly
in response to VEGF, but form a tight barrier. They resemble
stalk cells by depositing a basement membrane and establish-
c Signaling pathways to maintain ing junctions, but differ from these cells by their increased
a phalanx cell phenotype quiescence, and reduced mitogenic response to VEGF. The
tip/stalk cell model of vessel branching suggests that high
levels of VEGF stimulate the induction of a migratory EC tip
FGFR cell phenotype, while intermediate levels promote the prolif-
VE-Cadherin erative stalk cell phenotype. Genetic studies show that loss of
FGF2 VEGF in ECs causes widespread EC dysfunction and desin-
VEGFR1 tegration, indicating that low levels of VEGF are critical for
migration, quiescent ECs to survive.90 It is also known that the junctional
proliferation molecule VE-Cadherin shifts the EC response to VEGF from
VEGF
proliferation and migration to survival and quiescence.91
Interestingly, phalanx cells in PHD2 haplodeficient mice are
more quiescent by expressing elevated levels of VE-Cadherin
VEGFR2
Quiescence and sFlt1, which acts as a VEGF trap to counteract the tip
Survival VE-Cadherin cell-inducing activity of VEGF (Figure 5c).5
Blood flow

Additional molecular pathways have been impli-


PHD2+/- cated in EC quiescence and survival. Activation of
low O2 phosphatidylinositol-3-kinase (PI3K) and protein kinase B
Shear sFlt (PKB)/Akt signaling promotes EC survival in response to
stress various signals, including VEGF, FGFs, Ang1 (Figure 5c),
and insulin-like growth factor (IGF).77,92–94 Furthermore, in
PEDF
TIE2 confluent (quiescent) ECs, Ang1 induces Tie2 translocation
BMP9
Ang TSP to cell– cell contacts and the formation of homotypic Tie2–
Tie2 transassociated complexes that include the vascular
TIE2
endothelial phosphotyrosine phosphatase, leading to inhibi-
Figure 5 tion of paracellular permeability (Figure 5c).95 Moreover,
Figure 5. Molecular pathways of phalanx cell signaling. a and b,
FGF receptor inhibition disrupts endothelial integrity and
Scanning electron microscopic images of the inner wall of tumor dissolves interendothelial junctions. 77 Also, bone-
vessels grown in wild-type (a) or PHD2⫹/⫺ (b) mice show morphogenic protein (BMP)-9, and its ALK1 receptor, and
“abnormalized” versus “normalized” phalanx ECs. c, Signaling pigment epithelium derived factor (PEDF) were recently
mechanisms regulating phalanx cells quiescence.
identified to promote EC quiescence.96,97 Interaction of
thrombospondin (TSP)-1 and -2 with their CD36 receptor
Endothelial Phalanx Cells also diverge the proliferative and migratory stimuli of angio-
Once a vessel branch is formed, ECs become quiescent, from genic factors such as VEGF into survival signals (Figure
which only 0.01% are dividing in the healthy adult. The key 5c).98 Additionally, a number of molecular pathways have
function of vessels is then to supply blood and oxygen to been recently shown to induce tumor vessel normalization,
tissues. While blood flow itself is already an important factor such as the formation of perivascular nitric oxide gradients,
for keeping ECs quiescent,34 some of the molecules, deter- but whether this involves a shift to a phalanx EC phenotype,
mining this endothelial phenotype were recently identified by remains to be explored.99
genetic studies. Indeed, endothelial-specific heterozygous
deletion of prolyl-hydroxylase domain-2 (PHD2), an oxygen Conclusion
sensor, leads to “endothelial normalization” of tumor vessels. The above proposed model of vessel branching is largely
In contrast to wild-type tumor vessels (Figure 5a), where ECs based on seminal insights on tip, stalk, and phalanx cells from
are hyper-activated, extend multiple filopodia and loose tight the last 5 years. These studies have, however, also raised a
cell-cell contacts, ECs in PHD2 haplodeficient mice were number of outstanding questions. For instance, are these
aligned in a smooth tight cobblestone monolayer, resembling distinct EC phenotypes interchangeable, and what are the key
the “phalanx formation” of ancient Greek soldiers, hence molecular determinants of this combinatorial code? Are
their name “phalanx cells” (Figure 5b).5 A shift of tumor ECs relative rather than absolute expression levels, or activation
to phalanx cells improves cancer perfusion and oxygenation, status, of membrane receptors more relevant to specify EC
thereby almost completely preventing metastasis and induc- phenotypes? How do stalk cells maintain contact with the
ing a shift from a malignant to a more benign tumor leading tip cell and trail behind without dissolving the
Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015
De Smet et al Mechanisms of Vessel Branching 647

elongating stalk? What is the molecular basis of lumen 15. Gerhardt H, Ruhrberg C, Abramsson A, Fujisawa H, Shima D, Betsholtz
formation? How do tip cells recognize their counterparts and C. Neuropilin-1 is required for endothelial tip cell guidance in the
developing central nervous system. Dev Dyn. 2004;231:503–509.
fuse with each other? What is the molecular code of the 16. Tammela T, Zarkada G, Wallgard E, Murtomaki A, Suchting S,
phalanx phenotype? Is hypoxia codetermining the specifica- Wirzenius M, Waltari M, Hellstrom M, Schomber T, Peltonen R, Freitas
tion of EC fates? What is the full implication of Wnt- C, Duarte A, Isoniemi H, Laakkonen P, Christofori G, Yla-Herttuala S,
Shibuya M, Pytowski B, Eichmann A, Betsholtz C, Alitalo K. Blocking
signaling? How do endothelial filopodia sense their environ-
VEGFR-3 suppresses angiogenic sprouting and vascular network for-
ment, are guidance signals involved? Future explorations of mation. Nature. 2008;454:656 – 660.
tip, stalk, and phalanx cell behavior will benefit from im- 17. Lu X, Le Noble F, Yuan L, Jiang Q, De Lafarge B, Sugiyama D, Breant
proved in vivo imaging such as by fluorescence nanos- C, Claes F, De Smet F, Thomas JL, Autiero M, Carmeliet P, Tessier-
Lavigne M, Eichmann A. The netrin receptor UNC5B mediates
copy.100 Finally, providing an answer to those questions will guidance events controlling morphogenesis of the vascular system.
be useful to translate these concepts into the development of Nature. 2004;432:179 –186.
novel pro- and antiangiogenic therapies. 18. Suchting S, Freitas C, le Noble F, Benedito R, Breant C, Duarte A,
Eichmann A. The Notch ligand Delta-like 4 negatively regulates endo-
thelial tip cell formation and vessel branching. Proc Natl Acad Sci
Sources of Funding U S A. 2007;104:3225–3230.
P.C. is supported by long-term structural funding (Methusalem 19. Hellstrom M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, Lindblom
funding by the Flemish Government), Interuniversity Attraction Pole P, Alva J, Nilsson AK, Karlsson L, Gaiano N, Yoon K, Rossant J,
(grant P60/30, funded by Belgian Government, BELSPO), FWO Iruela-Arispe ML, Kalen M, Gerhardt H, Betsholtz C. Dll4 signalling
G.0692.09 (Flemish Government), research grant by the Belgian through Notch1 regulates formation of tip cells during angiogenesis.
“Foundation against Cancer,” GOA 2006/11 - KU Leuven. P.J.H. is Nature. 2007;445:776 –780.
recipient of a FEBS long-term fellowship. I.S. is recipient of a Marie 20. Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P, Gale
Curie Fellowship. NW, Lin HC, Yancopoulos GD, Thurston G. Blockade of Dll4 inhibits
tumour growth by promoting non-productive angiogenesis. Nature.
2006;444:1032–1037.
Disclosures 21. Roca C, Adams RH. Regulation of vascular morphogenesis by Notch
None. signaling. Genes Dev. 2007;21:2511–2524.
22. Matsuda M, Chitnis AB. Interaction with Notch determines endocytosis
References of specific Delta ligands in zebrafish neural tissue. Development (Cam-
1. Horowitz A, Simons M. Branching morphogenesis. Circ Res. 2008;103: bridge, England). 2009;136:197–206.
784 –795. 23. Estrach S, Legg J, Watt FM. Syntenin mediates Delta1-induced cohe-
2. Carmeliet P, Tessier-Lavigne M. Common mechanisms of nerve and siveness of epidermal stem cells in culture. J Cell Sci. 2007;120(Pt
blood vessel wiring. Nature. 2005;436:193–200. 16):2944 –2952.
3. Ghabrial AS, Krasnow MA. Social interactions among epithelial cells 24. Mattila PK, Lappalainen P. Filopodia: molecular architecture and
during tracheal branching morphogenesis. Nature. 2006;441:746 –749. cellular functions. Nat Rev Mol Cell Biol. 2008;9:446 – 454.
4. Gasparini F, Longo F, Manni L, Burighel P, Zaniolo G. Tubular 25. Pollard TD, Borisy GG. Cellular motility driven by assembly and dis-
sprouting as a mode of vascular formation in a colonial ascidian assembly of actin filaments. Cell. 2003;112:453– 465.
(Tunicata). Dev Dyn. 2007;236:719 –731. 26. Huber AB, Kolodkin AL, Ginty DD, Cloutier JF. Signaling at the
5. Mazzone M, Dettori D, Leite de Oliveira R, Schmidt T, Lanahan A, growth cone: ligand-receptor complexes and the control of axon growth
Jonckx B, De Smet F, Aragones A, Vinckier S, Loges S, Luttun A, and guidance. Annu Rev Neurosci. 2003;26:509 –563.
Wyns S, Jordan B, Pisacane A, Gallez A, Lampugnani MG, Dejana E 27. Defilippi P, Olivo C, Venturino M, Dolce L, Silengo L, Tarone G. Actin
RP, Simons M, Maxwell P, Carmeliet P. Heterozygous deficiency of cytoskeleton organization in response to integrin-mediated adhesion.
PHD2 restores tumor oxygenation and inhibits metastasis via endothelial Microscopy Res Tech. 1999;47:67–78.
normalization. Cell. 2009;136:839 – 851. 28. Mitra SK, Hanson DA, Schlaepfer DD. Focal adhesion kinase: in
6. Klosovskii BN, Zhukova TP. [Effect of colchicine on remote phases of command and control of cell motility. Nat Rev Mol Cell Biol. 2005;6:
growing capillaries in the brain.]. Arkh Patol. 1963;35:38 – 44. 56 – 68.
7. Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, 29. Lamalice L, Houle F, Jourdan G, Huot J. Phosphorylation of tyrosine
Abramsson A, Jeltsch M, Mitchell C, Alitalo K, Shima D, Betsholtz C. 1214 on VEGFR2 is required for VEGF-induced activation of Cdc42
VEGF guides angiogenic sprouting utilizing endothelial tip cell upstream of SAPK2/p38. Oncogene. 2004;23:434 – 445.
filopodia. J Cell Biol. 2003;161:1163–1177. 30. Ispanovic E, Serio D, Haas TL. Cdc42 and RhoA have opposing roles in
8. Dorrell MI, Aguilar E, Friedlander M. Retinal vascular development is regulating membrane type 1-matrix metalloproteinase localization and
mediated by endothelial filopodia, a preexisting astrocytic template and matrix metalloproteinase-2 activation. Am J Physiol Cell Physiol. 2008;
specific R-cadherin adhesion. Invest Ophthalmol Visual Sci. 2002;43: 295:C600 –C610.
3500 –3510. 31. Tan W, Palmby TR, Gavard J, Amornphimoltham P, Zheng Y, Gutkind
9. Wolff JR, Bar T. ‘Seamless’ endothelia in brain capillaries during JS. An essential role for Rac1 in endothelial cell function and vascular
development of the rat’s cerebral cortex. Brain Res. 1972;41:17–24. development. Faseb J. 2008;22:1829 –1838.
10. Marin-Padilla M. Early vascularization of the embryonic cerebral 32. van Nieuw Amerongen GP, Koolwijk P, Versteilen A, van Hinsbergh
cortex: Golgi and electron microscopic studies. J Comp Neurol. 1985; VW. Involvement of RhoA/Rho kinase signaling in VEGF-induced
241:237–249. endothelial cell migration and angiogenesis in vitro. Arterioscler
11. Ruhrberg C, Gerhardt H, Golding M, Watson R, Ioannidou S, Fujisawa Thromb Vasc Biol. 2003;23:211–217.
H, Betsholtz C, Shima DT. Spatially restricted patterning cues provided 33. Davis GE, Bayless KJ, Mavila A. Molecular basis of endothelial cell
by heparin-binding VEGF-A control blood vessel branching morpho- morphogenesis in three-dimensional extracellular matrices. Anatom Rec-
genesis. Genes Dev. 2002;16:2684 –2698. ord. 2002;268:252–275.
12. Yana I, Sagara H, Takaki S, Takatsu K, Nakamura K, Nakao K, Katsuki 34. Shiu YT, Li S, Marganski WA, Usami S, Schwartz MA, Wang YL,
M, Taniguchi S, Aoki T, Sato H, Weiss SJ, Seiki M. Crosstalk between Dembo M, Chien S. Rho mediates the shear-enhancement of endothelial
neovessels and mural cells directs the site-specific expression of cell migration and traction force generation. Biophys J. 2004;86:
MT1-MMP to endothelial tip cells. J Cell Sci. 2007;120(Pt 2558 –2565.
9):1607–1614. 35. Hoang MV, Whelan MC, Senger DR. Rho activity critically and selec-
13. Blum Y, Belting HG, Ellertsdottir E, Herwig L, Luders F, Affolter M. tively regulates endothelial cell organization during angiogenesis. Proc
Complex cell rearrangements during intersegmental vessel sprouting Natl Acad Sci U S A. 2004;101:1874 –1879.
and vessel fusion in the zebrafish embryo. Dev Biol. 2008;316:312–322. 36. Garrett TA, Van Buul JD, Burridge K. VEGF-induced Rac1 activation
14. Siekmann AF, Lawson ND. Notch signalling limits angiogenic cell in endothelial cells is regulated by the guanine nucleotide exchange
behaviour in developing zebrafish arteries. Nature. 2007;445:781–784. factor Vav2. Exp Cell Res. 2007;313:3285–3297.

Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015


648 Arterioscler Thromb Vasc Biol May 2009

37. Sauzeau V, Jerkic M, Lopez-Novoa JM, Bustelo XR. Loss of Vav2 58. Rhodes JM, Simons M. The extracellular matrix and blood vessel
proto-oncogene causes tachycardia and cardiovascular disease in mice. formation: not just a scaffold. J Cell Mol Med. 2007;11:176 –205.
Mol Biol Cell. 2007;18:943–952. 59. Mancuso MR, Davis R, Norberg SM, O’Brien S, Sennino B, Nakahara
38. Yamazaki D, Suetsugu S, Miki H, Kataoka Y, Nishikawa S, Fujiwara T, T, Yao VJ, Inai T, Brooks P, Freimark B, Shalinsky DR, Hu-Lowe DD,
Yoshida N, Takenawa T. WAVE2 is required for directed cell migration McDonald DM. Rapid vascular regrowth in tumors after reversal of
and cardiovascular development. Nature. 2003;424:452– 456. VEGF inhibition. J Clin Invest. 2006;116:2610 –2621.
39. Furman C, Sieminski AL, Kwiatkowski AV, Rubinson DA, Vasile E, 60. Danen EH, van Rheenen J, Franken W, Huveneers S, Sonneveld P,
Bronson RT, Fassler R, Gertler FB. Ena/VASP is required for endothe- Jalink K, Sonnenberg A. Integrins control motile strategy through a
lial barrier function in vivo. J Cell Biol. 2007;179:761–775. Rho-cofilin pathway. J Cell Biol. 2005;169:515–526.
40. Braren R, Hu H, Kim YH, Beggs HE, Reichardt LF, Wang R. Endo- 61. Jakobsson L, Domogatskaya A, Tryggvason K, Edgar D,
thelial FAK is essential for vascular network stability, cell survival, and Claesson-Welsh L. Laminin deposition is dispensable for vasculogenesis
lamellipodial formation. J Cell Biol. 2006;172:151–162. but regulates blood vessel diameter independent of flow. Faseb J.
41. Saadoun S, Papadopoulos MC, Hara-Chikuma M, Verkman AS. 2008;22:1530 –1539.
Impairment of angiogenesis and cell migration by targeted aquaporin-1 62. Williams CK, Segarra M, Sierra Mde L, Sainson RC, Tosato G, Harris
gene disruption. Nature. 2005;434:786 –792. AL. Regulation of CXCR4 by the Notch ligand delta-like 4 in endothe-
42. Kubo H, Fujiwara T, Jussila L, Hashi H, Ogawa M, Shimizu K, Awane lial cells. Cancer Res. 2008;68:1889 –1895.
M, Sakai Y, Takabayashi A, Alitalo K, Yamaoka Y, Nishikawa SI. 63. Lobov IB, Renard RA, Papadopoulos N, Gale NW, Thurston G, Yan-
Involvement of vascular endothelial growth factor receptor-3 in main- copoulos GD, Wiegand SJ. Delta-like ligand 4 (Dll4) is induced by
tenance of integrity of endothelial cell lining during tumor angiogenesis. VEGF as a negative regulator of angiogenic sprouting. Proc Natl Acad
Blood. 2000;96:546 –553. Sci U S A. 2007;104:3219 –3224.
43. Dixelius J, Makinen T, Wirzenius M, Karkkainen MJ, Wernstedt C, 64. Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y,
Alitalo K, Claesson-Welsh L. Ligand-induced vascular endothelial Kowalski J, Watts RJ, Callahan C, Kasman I, Singh M, Chien M, Tan
growth factor receptor-3 (VEGFR-3) heterodimerization with VEGFR-2 C, Hongo JA, de Sauvage F, Plowman G, Yan M. Inhibition of Dll4
in primary lymphatic endothelial cells regulates tyrosine phosphoryla- signalling inhibits tumour growth by deregulating angiogenesis. Nature.
tion sites. J Biol Chem. 2003;278:40973– 40979. 2006;444:1083–1087.
44. Geretti E, Shimizu A, Klagsbrun M. Neuropilin structure governs VEGF 65. Segarra M, Williams CK, Sierra Mde L, Bernardo M, McCormick PJ,
and semaphorin binding and regulates angiogenesis. Angiogenesis. Maric D, Regino C, Choyke P, Tosato G. Dll4 activation of Notch
2008;11:31–39. signaling reduces tumor vascularity and inhibits tumor growth. Blood.
45. Favier B, Alam A, Barron P, Bonnin J, Laboudie P, Fons P, Mandron M, 2008;112:1904 –1911.
Herault JP, Neufeld G, Savi P, Herbert JM, Bono F. Neuropilin-2 66. Phng LK, Potente M, Leslie JD, Babbage J, Nyqvist D, Lobov I, Ondr
interacts with VEGFR-2 and VEGFR-3 and promotes human endothelial JK, Rao S, Lang RA, Thurston G, Gerhardt H. Nrarp coordinates
endothelial Notch and Wnt signaling to control vessel density in angio-
cell survival and migration. Blood. 2006;108:1243–1250.
genesis. Dev Cell. 2009;16:70 – 82.
46. Chittenden TW, Claes F, Lanahan AA, Autiero M, Palac RT, Tkachenko
67. Sainson RC, Aoto J, Nakatsu MN, Holderfield M, Conn E, Koller E,
EV, Elfenbein A, Ruiz de Almodovar C, Dedkov E, Tomanek R, Li W,
Hughes CC. Cell-autonomous notch signaling regulates endothelial cell
Westmore M, Singh JP, Horowitz A, Mulligan-Kehoe MJ, Moodie KL,
branching and proliferation during vascular tubulogenesis. Faseb J.
Zhuang ZW, Carmeliet P, Simons M. Selective regulation of arterial
2005;19:1027–1029.
branching morphogenesis by synectin. Dev Cell. 2006;10:783–795.
68. Rutz S, Mordmuller B, Sakano S, Scheffold A. Notch ligands Delta-
47. Hong TM, Chen YL, Wu YY, Yuan A, Chao YC, Chung YC, Wu MH,
like1, Delta-like4 and Jagged1 differentially regulate activation of pe-
Yang SC, Pan SH, Shih JY, Chan WK, Yang PC. Targeting neuropilin
ripheral T helper cells. Eur J Immunol. 2005;35:2443–2451.
1 as an antitumor strategy in lung cancer. Clin Cancer Res. 2007;13:
69. Harrington LS, Sainson RC, Williams CK, Taylor JM, Shi W, Li JL,
4759 – 4768.
Harris AL. Regulation of multiple angiogenic pathways by Dll4 and
48. Sainson RC, Johnston DA, Chu HC, Holderfield MT, Nakatsu MN,
Notch in human umbilical vein endothelial cells. Microvascular Res.
Crampton SP, Davis J, Conn E, Hughes CC. TNF primes endothelial
2008;75:144 –154.
cells for angiogenic sprouting by inducing a tip cell phenotype. Blood. 70. Hodkinson PS, Elliott PA, Lad Y, McHugh BJ, MacKinnon AC, Haslett
2008;111:4997–5007. C, Sethi T. Mammalian NOTCH-1 activates beta1 integrins via the small
49. Petrovic N, Schacke W, Gahagan JR, O’Conor CA, Winnicka B, GTPase R-Ras. J Biol Chem. 2007;282:28991–29001.
Conway RE, Mina-Osorio P, Shapiro LH. CD13/APN regulates endo- 71. Ferrari-Toninelli G, Bonini SA, Bettinsoli P, Uberti D, Memo M. Micro-
thelial invasion and filopodia formation. Blood. 2007;110:142–150. tubule stabilizing effect of notch activation in primary cortical neurons.
50. Lee JF, Ozaki H, Zhan X, Wang E, Hla T, Lee MJ. Sphingosine-1- Neuroscience. 2008;154:946 –952.
phosphate signaling regulates lamellipodia localization of cortactin com- 72. Lindner V, Booth C, Prudovsky I, Small D, Maciag T, Liaw L. Members
plexes in endothelial cells. Histochem Cell Biol. 2006;126:297–304. of the Jagged/Notch gene families are expressed in injured arteries and
51. Vitorino P, Meyer T. Modular control of endothelial sheet migration. regulate cell phenotype via alterations in cell matrix and cell-cell inter-
Genes Dev. 2008;22:3268 –3281. action. Am J Pathol. 2001;159:875– 883.
52. Aase K, Ernkvist M, Ebarasi L, Jakobsson L, Majumdar A, Yi C, Birot 73. Albig AR, Becenti DJ, Roy TG, Schiemann WP. Microfibril-associate
O, Ming Y, Kvanta A, Edholm D, Aspenstrom P, Kissil J, glycoprotein-2 (MAGP-2) promotes angiogenic cell sprouting by
Claesson-Welsh L, Shimono A, Holmgren L. Angiomotin regulates blocking notch signaling in endothelial cells. Microvascular Res. 2008;
endothelial cell migration during embryonic angiogenesis. Genes Dev. 76:7–14.
2007;21:2055–2068. 74. Fischer C, Mazzone M, Jonckx B, Carmeliet P. FLT1 and its ligands
53. Ernkvist M, Persson NL, Audebert S, Lecine P, Sinha I, Liu M, VEGFB and PlGF: drug targets for anti-angiogenic therapy? Nat Rev.
Schlueter M, Horowitz A, Aase K, Weide T, Borg JP, Majumdar A, 2008;8:942–956.
Holmgren L. The Amot/Patj/Syx signaling complex spatially controls 75. Boulton ME, Cai J, Grant MB. gamma-Secretase: a multifaceted reg-
RhoA GTPase activity in migrating endothelial cells. Blood. 2009;113: ulator of angiogenesis. J Cell Mol Med. 2008;12:781–795.
244 –253. 76. Kametani Y, Takeichi M. Basal-to-apical cadherin flow at cell junctions.
54. Franco CA, Mericskay M, Parlakian A, Gary-Bobo G, Gao-Li J, Paulin Nat Cell Biol. 2007;9:92–98.
D, Gustafsson E, Li Z. Serum response factor is required for sprouting 77. Murakami M, Nguyen LT, Zhang ZW, Moodie KL, Carmeliet P, Stan
angiogenesis and vascular integrity. Dev Cell. 2008;15:448 – 461. RV, Simons M. The FGF system has a key role in regulating vascular
55. Shen WG, Xue QY, Zhu J, Hu BS, Zhang Y, Wu YD, Su Q. Inhibition integrity. J Clin Invest. 2008;118:3355–3366.
of adenovirus-mediated human MAGE-D1 on angiogenesis in vitro and 78. Schmidt M, Paes K, De Maziere A, Smyczek T, Yang S, Gray A, French
in vivo. Mol Cell Biochem. 300(1–2):89 –99, 2007. D, Kasman I, Klumperman J, Rice DS, Ye W. EGFL7 regulates the
56. Kayser MS, Nolt MJ, Dalva MB. EphB receptors couple dendritic collective migration of endothelial cells by restricting their spatial dis-
filopodia motility to synapse formation. Neuron. 2008;59:56 – 69. tribution. Development (Cambridge, England). 2007;134:2913–2923.
57. van Hinsbergh VW, Koolwijk P. Endothelial sprouting and angio- 79. Legg JA, Herbert JM, Clissold P, Bicknell R. Slits and Roundabouts in
genesis: matrix metalloproteinases in the lead. Cardiovasc Res. 2008; cancer, tumour angiogenesis and endothelial cell migration. Angio-
78:203–212. genesis. 2008;11:13–21.

Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015


De Smet et al Mechanisms of Vessel Branching 649

80. Sheldon H, Andre M, Legg JA, Heal P, Herbert JM, Sainson R, Sharma cavernous malformation signaling pathway promotes vascular integrity
AS, Kitajewski JK, Heath VL, Bicknell R. Active involvement of Robo1 via Rho GTPases. Nat Med. 2009.
and Robo4 in filopodia formation and endothelial cell motility mediated 90. Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, Desai S, Ferrara N,
via WASP and other actin nucleation-promoting factors. Faseb J. 2009; Nagy A, Roos KP, Iruela-Arispe ML. Autocrine VEGF signaling is
23:513–522. required for vascular homeostasis. Cell. 2007;130:691–703.
81. Park KW, Morrison CM, Sorensen LK, Jones CA, Rao Y, Chien CB, 91. Wallez Y, Vilgrain I, Huber P. Angiogenesis: the VE-cadherin switch.
Wu JY, Urness LD, Li DY. Robo4 is a vascular-specific receptor that Trends Cardiovasc Med. 2006;16:55–59.
inhibits endothelial migration. Dev Biol. 2003;261:251–267. 92. Makinde T, Agrawal DK. Intra and extravascular transmembrane sig-
82. Seth P, Lin Y, Hanai J, Shivalingappa V, Duyao MP, Sukhatme VP. nalling of angiopoietin-1-Tie2 receptor in health and disease. J Cell Mol
Magic roundabout, a tumor endothelial marker: expression and sig- Med. 2008;12:810 – 828.
naling. Biochem Biophys Res Commun. 2005;332:533–541. 93. Conti E, Carrozza C, Capoluongo E, Volpe M, Crea F, Zuppi C,
83. Suchting S, Heal P, Tahtis K, Stewart LM, Bicknell R. Soluble Robo4 Andreotti F. Insulin-like growth factor-1 as a vascular protective factor.
receptor inhibits in vivo angiogenesis and endothelial cell migration. Circulation. 2004;110:2260 –2265.
Faseb J. 2005;19:121–123. 94. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor
signalling - in control of vascular function. Nat Rev Mol Cell Biol.
84. Kaur S, Castellone MD, Bedell VM, Konar M, Gutkind JS, Ram-
2006;7:359 –371.
chandran R. Robo4 signaling in endothelial cells implies attraction
95. Saharinen P, Eklund L, Miettinen J, Wirkkala R, Anisimov A, Wind-
guidance mechanisms. J Biol Chem. 2006;281:11347–11356.
erlich M, Nottebaum A, Vestweber D, Deutsch U, Koh GY, Olsen BR,
85. Jones CA, London NR, Chen H, Park KW, Sauvaget D, Stockton RA,
Alitalo K. Angiopoietins assemble distinct Tie2 signalling complexes in
Wythe JD, Suh W, Larrieu-Lahargue F, Mukouyama YS, Lindblom P,
endothelial cell-cell and cell-matrix contacts. Nat Cell Biol. 2008;10:
Seth P, Frias A, Nishiya N, Ginsberg MH, Gerhardt H, Zhang K, Li DY. 527–537.
Robo4 stabilizes the vascular network by inhibiting pathologic angio- 96. David L, Mallet C, Keramidas M, Lamande N, Gasc JM, Dupuis-Girod
genesis and endothelial hyperpermeability. Nat Med. 2008;14:448 – 453. S, Plauchu H, Feige JJ, Bailly S. Bone morphogenetic protein-9 is a
86. Kamei M, Saunders WB, Bayless KJ, Dye L, Davis GE, Weinstein BM. circulating vascular quiescence factor. Circ Res. 2008;102:914 –922.
Endothelial tubes assemble from intracellular vacuoles in vivo. Nature. 97. Pollina EA, Legesse-Miller A, Haley EM, Goodpaster T, Randolph-
2006;442:453– 456. Habecker J, Coller HA. Regulating the angiogenic balance in tissues.
87. Koh W, Mahan RD, Davis GE. Cdc42- and Rac1-mediated endothelial Cell Cycle (Georgetown, Tex). 2008;7:2056 –2070.
lumen formation requires Pak2, Pak4 and Par3, and PKC-dependent 98. Silverstein RL, Febbraio M. CD36-TSP-HRGP interactions in the reg-
signaling. J Cell Sci. 2008;121:989 –1001. ulation of angiogenesis. Curr Pharmaceut design. 2007;13:3559 –3567.
88. Somlyo AV, Phelps C, Dipierro C, Eto M, Read P, Barrett M, Gibson JJ, 99. Kashiwagi S, Tsukada K, Xu L, Miyazaki J, Kozin SV, Tyrrell JA, Sessa
Burnitz MC, Myers C, Somlyo AP. Rho kinase and matrix metallopro- WC, Gerweck LE, Jain RK, Fukumura D. Perivascular nitric oxide
teinase inhibitors cooperate to inhibit angiogenesis and growth of human gradients normalize tumor vasculature. Nat Med. 2008;14:255–257.
prostate cancer xenotransplants. Faseb J. 2003;17:223–234. 100. Folling J, Bossi M, Bock H, Medda R, Wurm CA, Hein B, Jakobs S,
89. Whitehead KJ, Chan AC, Navankasattusas S, Koh W, London NR, Ling Eggeling C, Hell SW. Fluorescence nanoscopy by ground-state
J, Mayo AH, Drakos SG, Marchuk DA, Davis GE, Li DY. The cerebral depletion and single-molecule return. Nat Meth. 2008;5:943–945.

Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015


Mechanisms of Vessel Branching: Filopodia on Endothelial Tip Cells Lead the Way
Frederik De Smet, Inmaculada Segura, Katrien De Bock, Philipp J. Hohensinner and Peter
Carmeliet

Arterioscler Thromb Vasc Biol. 2009;29:639-649; originally published online March 5, 2009;
doi: 10.1161/ATVBAHA.109.185165
Arteriosclerosis, Thrombosis, and Vascular Biology is published by the American Heart Association, 7272
Greenville Avenue, Dallas, TX 75231
Copyright © 2009 American Heart Association, Inc. All rights reserved.
Print ISSN: 1079-5642. Online ISSN: 1524-4636

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://atvb.ahajournals.org/content/29/5/639

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Arteriosclerosis, Thrombosis, and Vascular Biology can be obtained via RightsLink, a service of the
Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for
which permission is being requested is located, click Request Permissions in the middle column of the Web
page under Services. Further information about this process is available in the Permissions and Rights
Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, and Vascular Biology is online
at:
http://atvb.ahajournals.org//subscriptions/

Downloaded from http://atvb.ahajournals.org/ by guest on January 23, 2015

You might also like