You are on page 1of 11

Angiogenesis

DOI 10.1007/s10456-014-9428-3

REVIEW PAPER

Intussusceptive angiogenesis: expansion and remodeling


of microvascular networks
Steven J. Mentzer • Moritz A. Konerding

Received: 16 November 2013 / Accepted: 20 March 2014


Ó Springer Science+Business Media Dordrecht 2014

Abstract Intussusceptive angiogenesis is a dynamic Keywords Intussusceptive angiogenesis 


intravascular process capable of dramatically modifying Microcirculation  Pillars  Microscopy  Blood flow
the structure of the microcirculation. The distinctive
structural feature of intussusceptive angiogenesis is the Abbreviations
intussusceptive pillar—a cylindrical microstructure that CAM Chick chorioallantoic membrane
spans the lumen of small vessels and capillaries. The EPC Endothelial progenitor cell
extension of the intussusceptive pillar appears to be a GFP Green fluorescent protein
mechanism for pruning redundant or inefficient vessels, SEM Scanning electron microscopy
modifying the branch angle of bifurcating vessels and TEM Transmission electron microscopy
duplicating existing vessels. Despite the biological impor-
tance and therapeutic potential, intussusceptive angiogen-
esis remains a mystery, in part, because it is an
intravascular process that is unseen by conventional light
microscopy. Here, we review several fundamental ques- Introduction
tions in the context of our current understanding of both
intussusceptive and sprouting angiogenesis. (1) What are The growth of new blood vessels from existing vessels—a
the physiologic signals that trigger pillar formation? (2) process known as angiogenesis—occurs in normal devel-
What endothelial and blood flow conditions specify pillar opment as well as in pathologic conditions involving tissue
location? (3) How do pillars respond to the mechanical repair [1], organ regeneration [2] and tumorigenesis [3]. In
influence of blood flow? (4) What biological influences adult animals, early intravital microscopy observations in
contribute to pillar extension? The answers to these ques- living tissue demonstrated that new vessels formed by ‘‘the
tions are likely to provide important insights into the sending out of sprouts from the vessel already present’’ as
structure and function of microvascular networks. in early growth in an embryo [4, 5]. In other cases,
‘‘numerous new branches and short connections’’ rapidly
formed without obvious sprouts [6]. These intravital
Presented, in part, at the Trans-NIH Angiogenesis Conference, May
21, 2014. observations are now considered to represent the two
fundamental processes of new vessel growth: sprouting and
S. J. Mentzer (&) nonsprouting angiogenesis.
Room 259, Laboratory of Adaptive and Regenerative Biology,
The process of nonsprouting or ‘‘intussusceptive’’
Harvard Medical School, Brigham & Women’s Hospital,
75 Francis Street, Boston, MA 02115, USA angiogenesis was formally identified in 1986 [7], although
e-mail: smentzer@partners.org earlier reports described a similar process [8, 9]. To visu-
alize blood vessel structure, Caduff and colleagues studied
M. A. Konerding
the developing rat lung using corrosion casting and scan-
Institute of Functional and Clinical Anatomy, Johannes
Gutenberg-University, Mainz, Germany ning electron microscopy (SEM). During the phase of rapid
e-mail: konerdin@uni-mainz.de alveolarization and capillary growth (7–13 days), they

123
Angiogenesis

Fig. 1 Intussusceptive pillars in the CAM. a Corriosion casting of the view of the vessel (i) was analyzed in orthogonal planes (ii)
CAM microcirculation was imaged with SEM. b Because the casting demonstrating a typical appearance of an intussusceptive pillar in
media fills the intraluminal space, the intussusceptive pillar is seen as cross-section (iii). Unpublished figures courtesy of Drs. Maximilian’
a hole in the vessel. c Confocal microscopy of fluorescent casts Ackermann and Grace Lee
demonstrates the transluminal orientation of the pillar. An en face

observed no capillary sprouts, but small holes in the sheet- bridge provides an opportunity for local exposure to a
like alveolar microvasculature [7]. These regular and variety of blood-borne elements including soluble factors
nonrandom holes were temporally and spatially associated and progenitor cells.
with rapid expansion of the microcirculation. Importantly, The process of sprouting capillaries can be quantita-
the diameter of the alveolar capillaries was smaller after, tively studied because individual sprouts can be counted
rather than prior to, expansion suggesting that the holes and the rate of growth assessed by light microscopy. In
were involved in not only capillary replication, but also contrast, nonsprouting angiogenesis is an intravascular
capillary remodeling [7]. The authors concluded that the process. Unseen by standard light microscopy, nonsprout-
small holes reflected a mechanism of ‘‘in-itself or intus- ing or intussusceptive angiogenesis is an underappreciated
susceptional growth’’—a process that made sprouting of process of multiplicative blood vessel growth and network
individual capillary segments unnecessary [7]. remodeling.
Because the holes were seen in casts of the vessel
lumen, the holes reflected a ‘‘pillar’’ or ‘‘post’’ spanning the
lumen of the blood vessel (Fig. 1). Pillar-like microstruc- Intussusceptive pillars
tures spanning a conduit are unique in mammalian anat-
omy; however, a similar structure exists in the gills of fish, The pillars identified by Caduff et al. during rat lung
molluscs and crustaceans [10, 11]. In these organisms, development were intravascular microstructures character-
blood flows between two thin epithelial plates separated by ized by their intraluminal location. In a variety of experi-
a series of pillars or trabeculae composed of characteristic mental models, intravascular pillars have been identified in
‘‘pillar cells’’ [12]. In both mammalian blood vessels and capillaries and small vessels. For example, intravascular
fish gills, pillars are a highly adaptive design feature for pillars have been identified in the developing chick cho-
optimizing bulk fluid transport. In mammalian vessels, the rioallantoic membrane (CAM) [13], murine chemically-
selective growth or extension of intravascular pillars can be induced colitis [14], a variety of tumors [15] and the
used to efficiently modify vessel structure. Depending upon physiologic angiogenesis associated with skeletal muscle
several influences, including the intravascular flow field, training [16]. Despite the many different model systems,
pillar extension can (1) modify the branching angle of a there are currently no reliable histologic markers or con-
bifurcating vessel, (2) duplicate an existing vessel, or (3) vincing in vitro models of intussusceptive pillars. Because
prune a redundant or energetically inefficient vessel of their intraluminal location, intussusceptive pillars have
(Fig. 2). In addition, the presence of an intraluminal tissue been largely defined by corrosion casting and SEM.

123
Angiogenesis

positive pressure instillation of the resin, or is an accurate


reflection of luminal morphology, is unknown. Of note,
technical artifacts such as extravasation, resin shrinkage
and residual intravascular particulates would not account
for the observations attributable to tissue pillars [17, 24].

Pillar ultrastructure

A disadvantage of corrosion casting is that the surrounding


tissue is digested away—casts provide little information
regarding the cellular or extracellular composition of the
pillar. To image the ultrastructural features of the pillar, the
most commonly used approach is serial transmission
electron microscopy (TEM) [25]. TEM has the advantage
of ultrastructural detail of the endothelial cells and tissue
comprising the pillar. Identifying a tissue pillar in a
2-dimensional analysis, however, requires hundreds of
serial TEM sections. In most cases, reliable TEM analysis
requires a tissue with a high density of intussusceptive
pillars, such as the developing rat lung during alveolo-
genesis [7].
Without a sufficiently high frequency of intussusceptive
pillars with a predictable orientation, pillars can be difficult
Fig. 2 Schematic representation of pillar extension with three to reliably identify in 2 dimensions. For example, when
different results. Pillar growth toward the vessel angle results in the viewed in tangential TEM sections, a looping vessel may
remodeling of vessel bifurcation. Pillar extension down the axis of the give the impression of a pillar. Because of the potential for
vessel results in vessel duplication. Asymmetric pillar growth can
result in pruning of a redundant or energetically inefficient vessel
misattribution, some investigators have limited the diam-
eter range of a potential pillar from 1 to 2.5 lm [26]. In
Corrosion casting is an investigative method that per- practice, TEM is time-consuming and economically
fuses the vascular system with a low viscosity resin that infeasible for most laboratories. Nonetheless, TEM has
polymerizes within the microvasculature. To visualize the identified the composition of the pillars in several tissues
intraluminal polymer, the surrounding tissue is digested including the developing rodent lung [26], skeletal muscle
and the remaining cast of the lumen is examined by SEM [16, 27] and murine colitis [14]. These studies suggest that
[17, 18]. SEM is a high resolution imaging technique that is a typical intussusceptive pillar is composed of endothelial
sufficiently scalable to characterize the structure of vas- membranes, occasional myofibroblasts and rare pericytes.
cular networks as well as individual vessels. Using SEM, The endothelial cell origin and the tubular configuration
the corrosion casts can provide useful data about vascular of the intussusceptive pillar is reminiscent of capillary
density, the presence of intussusceptive pillars and the sprouts. An appealing idea is that the intussusceptive pillar
orientation of vascular lining cells. High-quality casts can reflects a mirror image of abluminal sprouting (Fig. 3)
reveal endothelial nuclear imprints. Because endothelial [14, 28]. In sprouting angiogenesis, the basement mem-
nuclei align with flow [19–22], these corrosion casts can brane is degraded and endothelial ‘‘tip cells’’ project into
provide a measure of cell density and the direction of blood the extracellular matrix [29]. At least in the murine colitis
flow. In several model systems, including the CAM [23], [14] and rat skeletal muscle [8, 30] models of intussus-
nuclear imprints suggest that multiple endothelial cells ceptive angiogenesis, the basement membrane is not
contribute to the initial pillar structure. degraded. In addition, the endothelial cell projections are
An interesting observation is that corrosion casts of the not oriented into the extracellular matrix, but across the
vessel lining cells neighboring the intussusceptive pillars vessel lumen. In both sprouting and intussusceptive angi-
are typically unremarkable. Membrane irregularities, ogenesis, the endothelial cell projections are believed to
micro-spikes or distinctive mural contours are not associ- contain contractile elements and a shape consistent with
ated with the presence of intraluminal pillars. There are no filopodia (Table 1).
intraluminal features that would indicate, or specify, the Endothelial cell filopodia are finger-like projections fil-
location of the pillar. Whether this observation reflects the led with tight parallel bundles of filamentous (F)-actin. In

123
Angiogenesis

Fig. 3 Speculative comparison


of the early processes of
sprouting and intussusceptive
angiogenesis. In sprouting
angiogenesis, tip cells project
into the extracellular matrix
through a disrupted basement
membrane (dotted lines). In
contrast, intussusceptive pillars
may project across the vessel
lumen with an intact basement
membrane

Table 1 Comparison of sprouting and intussusceptive angiogenesis there is no evidence that the lumen-spanning endothelial
Common features of sprouting and nonsprouting angiogenesis cells reflect tip cell characteristics; that is, dynamic filo-
Both are triggered by extraluminal processes (e.g. inflammation, podia, migratory behavior, and growth factor responsive-
metabolic, growth factors) ness [31]. Also, there is inconclusive data to support ‘‘stalk
Both sprouts and pillars reflect localized processes [initiated by cell’’ function as there is little endothelial cell proliferation
endothelial cell(s)] in the early stages of intussusceptive angiogenesis despite
Both sprouts and pillars reflect cylindrical structures the formation of a nascent pillar lumen [33, 34]. Another
Unique features of intussusceptive angiogenesis (IA) important distinction with sprouting angiogenesis is that
IA can be triggered by extraluminal or intraluminal stimulation the projections extend into the lumen of a functioning
IA does not disrupt basement membrane capillary; that is, pillar formation occurs without an obvi-
IA does not require endothelial cell proliferation ous growth factor diffusion gradient to guide endothelial
IA does not require endothelial cell abluminal migration cell projections. Since pillars develop with an intact base-
Pillar morphology adapts to intraluminal flow fields ment membrane and without obvious extraluminal growth
Pillar is exposed to, and may interact with, blood-borne elements factors, the potential role for morphogen gradients in reg-
ulating endothelial cell protrusion and endothelial cell–cell
fusion is unclear.
Regardless of the mechanism of transluminal fusion, the
sprouting angiogenesis, the elongation of these filaments capillary endothelial cells involved in both sprouting and
projects the leading edge of the endothelial cell into the intussusceptive angiogenesis necessarily remodel their
extracellular matrix. Growth factor receptors, such as local endothelial cell–cell attachments [35]. Perhaps a
VEGF receptors (VEGFR2) on the filopodia likely provide reflection of this flexible polarity, endothelial cells are
guidance cues for endothelial cell migration [31]. Although recognized to have less apical and basal lateral sorting of
the model of inward sprouting provides an appealing proteins than epithelial cells [36, 37]. Another potential
symmetry for intussusceptive angiogenesis, most endothe- distinction is the interaction of endothelial cells with the
lial cell membranous projections visualized by TEM do not extracellular matrix. In sprouting angiogenesis, the endo-
contain contractile elements [9, 14] and have been char- thelial cell–extracellular matrix interaction appears to play
acterized by the more general term ‘‘pseudopodial’’ pro- a functional role during lumenogenesis [38]; however, it is
cesses [32]. Furthermore, it is unclear whether the actin unclear how endothelial interactions with the extracellular
network in the endothelial cell cytoskeleton is sufficiently matrix are potentially involved in pillar formation.
rigid to resist blood flow in a distended capillary or have
sufficient protrusive force to project across a lumen filled
with blood. Pillars and blood flow
In sprouting angiogenesis, sprout fusion appears to
occur as a result of filopodial interactions between two The development of intravascular pillars produces a curi-
approaching tip cells. In intussusceptive angiogenesis, ous ‘‘symmetry’’ with fused sprouts (Fig. 4). An important

123
Angiogenesis

Fig. 4 Schematic comparison


of the later stages of sprouting
and intussusceptive
angiogenesis. The fusion of
abluminal sprouts may or may
not be influenced by dominant
flow fields within the
microcirculation. In contrast,
intraluminal pillars are
necessarily exposed to blood
flow within the vessel
undergoing intussusceptive
remodeling (arrow denotes
blood flowing around the
intraluminal pillar)

distinction is that intussusceptive pillars remain within the orthogonal to the bifurcation plane. Confirmed by corro-
original flow stream and are potentially responsive to sion casting and SEM as well as confocal microscopy, the
intravascular flow fields. Although the early pillars iden- orientation of the pillar relative to the bifurcation plane—as
tified by TEM and SEM are typically cylindrical structures well as its typical location through the central axis of the
\5 lm in diameter, later imaging indicates that intussus- vessel (e.g. Fig. 1c)–suggests that pillar orientation is
ceptive pillars can grow and extend to create duplicated determined, in part, by blood flow from both converging
and remodeled blood vessels. Pillar extension down the vessels [40].
axis of the vessel appears to be the mechanism responsible To investigate the influence of blood flow on pillar
for vessel duplication without the need for endothelial cell extension, we have developed 3-D finite element models of
proliferation [16]. Similarly, pillar extension in non-axial intussusceptive angiogenesis in murine colitis [41] and the
directions appears to be the mechanism responsible for chick CAM [23]. Based on measurable flow dynamics and
remodeling the branch angles of bifurcating vessels [39] or essential structural features of the blood vessels identified
pruning redundant or energetically inefficient vessel bran- by intravital microscopy, these computational models have
ches [23]. allowed us to map the distribution of mechanical forces
To identify intussusceptive pillars and provide a within the vessels containing an intussusceptive pillar. An
simultaneous assessment of blood flow, we have adapted assumption of this work is that the blood is treated as a
an intravital video microscopy system to the planar net- Newtonian fluid—effectively discounting mechanical
works in the mouse colon and the CAM [23, 40]. Using effects of blood cells. Justifying this assumption, the chick
fluorescent plasma markers, intravascular pillars can be and murine computational models have strikingly similar
identified by persistent flow voids and confirmed by time- results despite a wide variation in blood cell concentra-
lapse digital recombination. This analysis focused on the tions. Based on the comparisons of these two models
larger conducting vessels, rather than the capillary mesh- [23, 41], we suspect that our computational models closely
work (Fig. 1a, white arrows) in the chick CAM because of approximate the distribution of forces in vivo.
the complex network flow interactions in the meshwork as Using finite element flow models, we can perform
well as the difficulty in distinguishing tissue islands inci- computational ‘‘pillar deletion’’ experiments; that is, we
dentally entrapped by sprout fusion from active intussus- can calculate the distribution of forces in the same vessel
ceptive pillars. with and without intraluminal pillars (Fig. 5). These stud-
Analysis of the CAM conducting vessels has demon- ies suggest that individual pillars, or the first pillar in a
strated that approximately 1 % of the non-mesh vascular series of pillars, form in regions of the vessel with minimal
network is comprised of flow voids consistent with intus- shear stress. Pillars are primarily located in micro-hemo-
susceptive pillars [40]. Most of intussusceptive pillars were dynamic ‘‘dead zones’’ within the vessel [23, 41]; in most
identified at vessel bifurcations; 80 % of those bifurcations cases, pillars form in flow regions with shear stress below
reflected convergent flow. In most cases, the single pillar 1 dyn/cm2 [23] (Fig. 5b, white arrow). Because regions of
located at the site of convergent flow was oriented low wall shear stress are not always associated with pillars,

123
Angiogenesis

Fig. 5 Finite element models of bifurcating vessels in the chick without pillars demonstrate low shear stress in the region of the first
CAM (after [23]). a An example of a vessel with multiple or ‘‘serial’’ pillar (white arrow). d Similar flow conditions with the sequential
pillars (blood flow indicated by black arrows). b A finite element addition of the pillars (1 through 4) suggest that pillars in series form
model of the vessel without the pillars demonstrating the flow ‘‘downstream’’ from the initial pillar
velocity streamlines. c Wall shear stress calculations of the vessel

we have postulated a permissive role of wall shear stress in intravascular pillars to blood flow features suggest that
pillar development. We suspect that regions of low wall intravascular pillars are a fundamental mechanism for
shear stress are necessary, but not sufficient, for the optimizing the structure of the microcirculation.
development of intussusceptive pillars. In addition to low In addition to modifying and optimizing the structure of
shear stress, endothelial cell activation from extravascular the microcirculation, an intriguing therapeutic possibility is
or intraluminal signals may be required for pillar formation the potential for intussusceptive angiogenesis to expand
[23]. vascular networks. For example, pillars in a high flow
Once the pillar is formed, pillar extension appears to be setting could serially divide the vessel and rapidly expand
limited by wall shear stress. Multiple, or serial, pillars in the existing microcirculatory network. Multiplicative
the chick CAM provide a useful model for examining both expansion of the network would be particularly useful in
pillar formation and pillar extension. In most cases, new tissues with ischemia or high metabolic demands. The
pillars in a pillar sequence form sequentially; in other existing data suggest two intriguing mechanisms for initi-
words, the next pillar in a sequence forms ‘‘downstream’’ ating intussusceptive angiogenesis: one mechanism is
from the pre-existing pillars (e.g. Fig. 5). Although pillar triggered by intraluminal events and the second is triggered
extension occurs within hours, it is generally too slow for by extravascular events.
real-time video microscopy. Nonetheless, the shape and
extension of existing pillars suggests these intravascular
structures are limited by wall shear stress [40]. Intraluminal stimulus

Vessel growth in skeletal muscle occurs under physiologic


Network structure circumstances. Skeletal muscles demonstrate an adaptive
response to changing conditions including physiologic
The performance of vascular networks is highly dependent stress in the form of exercise training. By increasing the
on their architecture. Tissues undergoing regeneration or blood flow to muscles threefold to sixfold, endurance
repair often demonstrate heterogeneous vascular microar- training can result in a 15–20 % increase in both capillary
chitecture because angiogenesis is driven by stochastic density as well as capillary–fiber ratio [42–44]. In perhaps
processes. Initiated by metabolic demands and guided by the earliest electron microscopic description of intussus-
morphogen gradients, the mechanisms of capillary ceptive angiogenesis, Ogawa studied rats subjected to 1 h
sprouting and sprout fusion create heterogeneity that has of daily exercise training [8]. After 60 days, he demon-
implications for both blood flow distribution and the strated a significant increase in vascularity characterized by
energy cost of blood transport. intussusceptive luminal division [8].
An underappreciated role of intussusceptive angiogen- Although these results suggested that the capillary
esis is that it provides a mechanism for modifying the growth was triggered by increased blood flow associated
structure of microvascular networks. The responsiveness of with endurance training-stimulated capillary growth, the

123
Angiogenesis

interpretation of these experiments was complicated by the more protracted and spatially more distributed than is
many systemic changes associated with exercise training observed in models of sprouting angiogenesis.
[45]. Activation of specific muscle groups with electrical Third, how does the increased wall shear stress trigger
stimulation provides a more controlled experimental the development of intussusceptive pillars? To date,
alternative. When compared to endurance training, elec- in vitro studies of endothelial cell responses to variable
trical stimulation produced a similar pattern of capillary levels of shear stress have not identified pillar-like endo-
growth, but without the confounding systemic effects of thelial cell structures [22]. The absence of shear stress-
exhaustive training [46]. The results of both exercise induced in vitro pillars may reflect insufficiently sensitive
training and electrical stimulation suggested that increased assessments of endothelial morphology. Alternatively,
blood flow alone could induce intussusceptive angiogenesis shear stress-induced endothelial cell changes may require
[30, 44, 47]. in vivo elements—such as an intact capillary basement
To examine the influence of enhanced muscle flow membrane—that are necessary for the initiation of intus-
without training-associated muscular contraction, several susceptive angiogenesis. Of note, measures of endothelial
studies have used chronic peripheral arteriolar dilatation cell transcriptional responses to shear stress have demon-
[48, 49]. Using alpha-1 receptor antagonist prazosin, cap- strated the potential involvement of many genes and sig-
illary shear stress was threefold (4–15 dyn/cm2) greater naling pathways [53, 54].
than in control animals or animals treated by chronic Finally, the elevated shear stress associated with muscle
electrical stimulation alone [27, 50]. Although prazosin training in intussusceptive angiogenesis appears to be in
was associated with little demonstrable endothelial cell conflict with our blood flow simulations in the chick CAM
proliferation by PCNA staining [16, 51], there were ele- and mouse colitis models; namely, the computational
vated levels of VEGF in both the prazosin and chronic observations that pillars appear to form in regions of low
electrical stimulation conditions [50]. Most importantly, shear stress and are limited by high shear stress. A potential
histologic assessment of the prazosin-treated muscle explanation for this apparent paradox is the temporal pat-
(2 weeks) demonstrated a 40 % increase in capillary–fiber tern of blood flow in skeletal muscle. It is possible that it is
ratio [50]. TEM of the muscle in similar treatment condi- the sequential exposure of the vessel lining cells to high
tions has shown intussusceptive pillars and an intact cap- and low shear stress leads to pillar formation. The capillary
illary basement membrane [50]. leak associated with muscle training [8] may also facilitate
These results suggest that shear stress alone, in the endothelial contact reminiscent of the Baxter–Jain model
absence of obvious extraluminal growth factor or meta- of blood flow heterogeneity in tumors [55].
bolic stimulation, can trigger intussusceptive angiogenesis.
The preservation of intact basement membranes [8, 14, 30]
suggests that the process of intussusceptive angiogenesis Extraluminal stimulus
has important distinctions from typical sprouting angio-
genesis; namely, endothelial cell proliferation, basement A complementary model of adult intussusceptive angio-
membrane degradation and abluminal endothelial cell genesis is murine colitis. In chemically-induced murine
migration appear to be unnecessary for intussusceptive colitis, the administration of chemicals such as dextran
capillary replication. These observations, however, raise sodium sulfate (oral) or trinitrobenzene sulfonic acid
several intriguing questions. First, how are the divided (rectal instillation) results in both acute and chronic colitis.
capillaries redistributed within the muscle fibers? After In acute colitis, murine video endoscopy shows inflam-
2 weeks, the capillaries appear to be uniformly distributed mation of the colonic mucosa [56]. Histology of the colon
throughout the skeletal muscle [52]. The process of capil- demonstrates an infiltration of the superficial mucosa with
lary redistribution is likely associated with gradual base- an inflammatory mononuclear infiltrate [57]. In chronic
ment membrane remodeling and perhaps distraction forces colitis, the mucosa demonstrates increased vascularity.
unique to skeletal muscle. Corrosion casting and SEM of the chronically inflamed
Second, how does this process occur with minimal mucosal microcirculation has demonstrated duplicated and
endothelial cell proliferation? Capillary luminal division triplicated mucosal vessels [14].
alone—without a concomitant increase in endothelial cell The blood flow to the colonic mucosa is supplied by a
circumference—results in a 50 % decrease in cross-sec- polygonal mucosal network surrounding the colonic crypts
tional area. Although post-intussusceptive endothelial cell [58]. Within the first 4 days of inflammation, corrosion
enlargement has been observed [9], it is likely that the casting and SEM demonstrate a significant increase in the
growth phase of intussusceptive angiogenesis in skeletal diameter of mucosal plexus vessels. The diameter of the
muscle is also associated with endothelial cell division; mucosal vessels nearly doubles (8–15 lm) in many regions
however, we speculate that the proliferation is temporally of the mucosal plexus within 96 h [57]. Associated with

123
Angiogenesis

this dilatation is a 50 % reduction in flow velocity of significant distraction suggests that the arrangement of
(1,500–700 lm/s) [57]. By 7 days after the onset of acute duplicated capillaries depends upon the anatomical con-
colitis, intravital microscopy of the mucosal plexus dem- straints (crypts) and mechanical forces (skeletal muscle) in
onstrates complex flow patterns [58, 59]. In 15–25 % of the surrounding tissue.
vessel segments, tracer flow is excluded. Flow variability Third, what are the inflammatory signals that trigger
appears to be spatially related to occlusive platelet aggre- intussusceptive angiogenesis? An interesting observation in
gates within the mucosal plexus [60]. Coincident with the murine colitis is the relative imbalance of blood vessel
decrease in blood flow and increase in excluded segments, growth toward intussusceptive angiogenesis; little sprout-
corrosion casting and SEM demonstrates filling defects in ing angiogenesis was observed after chemical stimulation.
the casts consistent with intussusceptive pillars [14]. The Whether the differential angiogenic response was a
pillars are consistently identified at vessel bifurcations and reflection of the inflammatory cell composition, growth
are oriented orthogonal to the bifurcation plane. TEM factor milieu or tissue response is unknown.
demonstrates that the pillar is composed of endothelial
cells dividing the original mucosal plexus capillary lumen
into 2 vessels [14]. Pillar extension
Corrosion casting and SEM 28–31 days after the onset
of inflammation demonstrates regions of the mucosa with Although observations in the CAM indicates that pillar
significant vascular replication [14]. Some regions of the extension often occurs ‘‘downstream’’ to the initial pillar,
mucosal plexus demonstrated multiple new layers of the the mechanism of pillar extension is unknown. The mor-
vascular plexus. In both models of chemically-induced phologic evidence, based on corrosion casting and SEM,
colitis, replication of the vessels in the mucosal plexus is suggests that endothelial nuclear impressions in the
heterogeneous with ‘‘patchy’’ areas demonstrating little or expanding pillar are indistinguishable from other mural
no angiogenesis. lining cells. The endothelial cell nuclear impressions
In a time course similar to vessel duplication, a subset of reflect comparable size and orientation relative to the
vessel bifurcations in murine colitis demonstrate significant presumed flow stream [23]. There are no morphologic
remodeling of the vessel branch angle [39]. The morpho- clues suggesting an explanation for the selective down-
logic features of the remodeled vessel angles are reminis- stream proliferation of pillar endothelial cells. We have
cent of the intussusceptive process resulting in luminal also been unable to identify focal areas of endothelial
duplication. In this case, however, pillar extension appears proliferation.
to extend toward the apex of the vessel angle. Also similar Although undetected endothelial proliferation is a pos-
to other intussusceptive processes, the spatial orientation sible explanation, it is also possible that the intussusceptive
and the periodicity of the remodeled angles has suggested pillar functions as a ‘‘trap’’ for blood borne endothelial
that remodeling was a response to regional flow patterns progenitor cells (EPCs). EPCs in the blood has been sus-
[39]. pected since DeBakey and colleagues suspended a small
These results indicate that that pillar formation in two piece of crumpled Dacron— called a ‘‘hub’’—with
models of chemically-induced colitis leads to both intus- monofilament sutures in the aorta of small pigs [61].
susceptive angiogenesis and branch angle remodeling. Within a month, the piece of Dacron was coated with
Similar to skeletal muscle models, these models of extra- endothelial cells as documented by electron microscopy
luminal stimulation of intussusceptive angiogenesis sug- [62]. Although the report was underappreciated at the time,
gest several outstanding questions. First, what is the it is now clear that Stump, O’Neal, DeBakey and col-
influence of blood flow heterogeneities in intussusceptive leagues introduced the field of EPC biology. These studies
angiogenesis? Platelet aggregates are likely a common also identified an essential feature of the EPC ‘‘trap’’; that
source of heterogeneity in many models of tissue inflam- is, the efficient delivery of EPCs by placing the ‘‘hub’’ or
mation. In tumor biology, blood flow heterogeneities have ‘‘trap’’ within the flow stream.
been associated with leaky capillaries and interstitial Our group has developed a parabiotic cross-circulation
hypertension [55]. It is possible that there are features model to investigate the possibility of an EPC trap [63].
common to inflammation and tumorigenesis that contribute Parabiosis establishes a common blood circulation by
to intussusceptive angiogenesis. surgical pairing; that is, creating a surgical union of twin
Second, how are the duplicated capillaries redistributed animals or parabionts [64]. The development of genetically
in the tissue? The colitis model provides an example of engineered mice expressing green fluorescent protein
vascular duplication without the contractions associated (GFP) have provided an opportunity to track blood borne
with skeletal muscle angiogenesis. In chronic colitis, the cells with a persistent cytoplasmic marker [65]. Most
vessels were often distributed close together. The absence importantly, parabiosis provides a fate map of cell

123
Angiogenesis

migration without bone marrow transplantation or ex vivo References


labeling. Using the parabiosis model we have demonstrated
dramatic numbers of EPCs migrating to, and integrating 1. Eming SA, Brachvogel B, Odorisio T, Koch M (2007) Regulation
into, the vascular endothelium of the growing mouse lung of angiogenesis: wound healing as a model. Prog Histochem
Cytochem 42:115–170
[66].
2. Song JJ, Ott HC (2011) Organ engineering based on decellular-
We have also used the parabiosis model to investigate ized matrix scaffolds. Trends Mol Med 17:424–432
EPC migration in inflammatory colitis [14]. In the colon, 3. Zhao Y, Bao Q, Renner A, Camaj P, Eichhorn M, Ischenko I,
we tracked GFP? cells into the colonic vasculature (inte- Angele M, Kleespies A, Jauch KW, Bruns C (2011) Cancer stem
cells and angiogenesis. Int J Dev Biol 55:477–482
gration was confirmed by confocal co-localization). An
4. Clark ER (1918) Studies on the growth of blood-vessels in the tail
intriguing finding was the pattern of GFP? migration. of the frog larva—by observation and experiment on the living
Readily analyzed because of the planar vascular network in animal. Am J Anat 23:37–88
the colon, concentrations of GFP? cells were identified in 5. Schulte HW (1914) Early stages of vasculogenesis in the cat
(Felis domestica) with especial reference to the mesenchymal
regions of known intussusceptive angiogenesis; that is, at
origin of endothelium. The Wistar Institute of Anatomy and
vessel bifurcations within the mucosal plexus [14]. Further, Biology
the frequency of these aggregated GFP? cells was statis- 6. Clark ER, Clark EL (1940) Microscopic observations on the
tically identical to the frequency of intussusceptive pillars extra-endothelial cells of living mammalian blood vessels. Am J
Anat 66:1–49
[14]. Immunostaining demonstrated that these cells at the
7. Caduff JH, Fischer LC, Burri PH (1986) Scanning electron
vessel bifurcations expressed the EPC marker CD34. Also, microscope study of the developing microvasculature in the
vessels already having undergone intussusceptive duplica- postnatal rat lung. Anat Rec 216:154–164
tion demonstrate diffusely positive, but lower levels of 8. Ogawa Y (1977) On the fine structural changes of the micro-
vascular beds in skeletal muscle. J Yokohama City Univ Sec
GFP fluorescence—a characteristic of EPC that have
Sport Sci Med 6:1–19
transitioned into mature vascular lining (endothelial) cells 9. Appell H-J (1980) Morphological studies on skeletal muscle
[67]. Together, these findings suggest that blood-borne under conditions of high altitude training. Int J Sports Med
EPC may co-localize at the site of intussusceptive 1:103–109
10. Hughes GM, Morgan M (1973) Structure of fish gills in relation
angiogenesis.
to their respiratory function. Biol Rev Camb Philos Soc
48:419–475
11. Olson KR, Dewar H, Graham JB, Brill RW (2003) Vascular
anatomy of the gills in a high energy demand teleost, the skipjack
Summary tuna (Katsuwonus pelamis). J Exp Zool 297A:17–31
12. Mongera A, Singh AP, Levesque MP, Chen YY, Konstantinidis
P, Nusslein-Volhard C (2013) Genetic lineage labeling in zeb-
In this review, we discussed our current understanding of rafish uncovers novel neural crest contributions to the head,
the structure and function of the intussusceptive pillar in including gill pillar cells. Development 140:916–925
the process of intussusceptive angiogenesis. (1) Observa- 13. Makanya AN, Hlushchuk R, Djonov VG (2009) Intussusceptive
tions in the chick CAM suggest that pillars form in areas of angiogenesis and its role in vascular morphogenesis, patterning,
and remodeling. Angiogenesis 12:113–123
low shear stress. An unresolved paradox is that skeletal 14. Konerding MA, Turhan A, Ravnic DJ, Lin M, Fuchs C, Secomb
muscle intussusceptive angiogenesis appears to be stimu- TW, Tsuda A, Mentzer SJ (2010) Inflammation-induced intus-
lated by high blood flow and elevated wall shear stress. (2) susceptive angiogenesis in murine colitis. Anat Rec 293:849–857
The nonrandom orientation of pillars orthogonal to the 15. Ribatti D, Djonov V (2012) Intussusceptive microvascular
growth in tumors. Cancer Lett 316:126–131
bifurcation plane of branching vessels in most models 16. Egginton S, Zhou AL, Brown MD, Hudlicka O (2001) Unorthodox
suggests that pillar location is dependent upon blood flow, angiogenesis in skeletal muscle. Cardiovasc Res 49:634–646
but the requisite conditions for pillar formation remain 17. Konerding MA (1991) Scanning electron microscopy of corro-
unclear. (3) Finite element models of the chick CAM and sion casting in medicine. Scanning Microsc 5:851–865
18. Lametschwandtner A, Aharinejad S (1997) Scanning electron
murine colitis models suggest that shear stress has both a microscopy/corrosion casting technique in biological and medical
permissive and limiting influence on intussusceptive pillar; research. State of the art and perspectives. In: Recent advances in
however, these observations are unresolved with the high microscopy of cells, tissues and organs. Antonio Delfino, Rome.
blood flow conditions associated with skeletal muscle pp 51–58
19. Gottlieb AI, Langille BL, Wong MK, Kim DW (1991) Structure
intussusceptive angiogenesis. (4) The intravascular loca- and function of the endothelial cytoskeleton. Lab Invest
tion of the intussusceptive pillar suggests its potential role 65:123–137
in localizing blood-borne elements, such as EPCs, to areas 20. Malek AM, Izumo S (1996) Mechanism of endothelial cell shape
of the microcirculation undergoing active angiogenesis. change and cytoskeletal remodeling in response to fluid shear
stress. J Cell Sci 109(Pt 4):713–726
We anticipate that further studies in these areas will pro- 21. Galbraith CG, Skalak R, Chien S (1998) Shear stress induces
vide fundamental insights into the remodeling and expan- spatial reorganization of the endothelial cell cytoskeleton. Cell
sion of microvascular networks. Motil Cytoskelet 40:317–330

123
Angiogenesis

22. Barbee KA, Davies PF, Lal R (1994) Shear stress-induced reor- 43. Egginton S, Hudlicka O, Brown MD, Walter H, Weiss JB, Bate A
ganization of the surface topography of living endothelial cells (1998) Capillary growth in relation to blood flow and perfor-
imaged by atomic force microscopy. Circ Res 74:163–171 mance in overloaded rat skeletal muscle. J Appl Physiol 85:
23. Lee GS, Filipovic N, Lin M, Gibney BC, Simpson DC, Koner- 2025–2032
ding MA, Tsuda A, Mentzer SJ (2011) Intravascular pillars and 44. Hudlicka O, Brown M, Egginton S (1992) Angiogenesis in
pruning in the extraembryonic vessels of chick embryos. Dev skeletal and cardiac-muscle. Physiol Rev 72:369–417
Dyn 240:1335–1343 45. Green DJ, Maiorana AJ, Cable NT (2008) Point: counterpoint:
24. Schraufnagel DE, Schmid A (1988) Microvascular casting of the exercise training does/does not induce vascular adaptations
lung: effects of various fixation protocols. J Electron Microsc beyond the active muscle beds. J Appl Physiol 105:1002–1004
Tech 8:185–191 46. Milkiewicz M, Hudlicka O, Brown MD, Silgram H (2005) Nitric
25. Nico B, Crivellato E, Ribatti D (2007) The importance of electron oxide, VEGF, and VEGFR-2: interactions in activity-induced
microscopy in the study of capillary endothelial cells: an histor- angiogenesis in rat skeletal muscle. Am J Physiol Heart Circ
ical review. Endothelium 14:257–264 Physiol 289:H336–H343
26. Burri PH, Tarek MR (1990) A novel mechanism of capillary 47. Hudlicka O, Brown MD (2009) Adaptation of skeletal muscle
growth in the rat pulmonary microcirculation. Anat Rec 228: microvasculature to increased or decreased blood flow: role of
35–45 shear stress, nitric oxide and vascular endothelial growth factor.
27. Williams JL, Cartland D, Rudge JS, Egginton S (2006) VEGF J Vasc Res 46:504–512
trap abolishes shear stress- and overload-dependent angiogenesis 48. Tornling G, Adolfsson J, Unge G, Ljungqvist A (1980) Capillary
in skeletal muscle. Microcirculation 13:499–509 neoformation in skeletal-muscle of dipyridamole-treated rats.
28. Paku S, Dezso K, Bugyik E, Tovari J, Timar J, Nagy P, Laszlo V, Arzneimittel-Forschung 30–1:791–792
Klepetko W, Dome B (2011) A new mechanism for pillar for- 49. Hudlicka O, Komarek J, Wright AJA (1981) The effect of a
mation during tumor-induced intussusceptive angiogenesis. Am J xanthine derivative, 1-(50 -oxohexyl)-3-methyl-7-propylxanthine
Pathol 179:1573–1585 (hwa-285), on heart performance and regional blood-flow in dogs
29. Eming SA, Hubbell JA (2011) Extracellular matrix in angio- and rabbits. Br J Pharmacol 72:723–730
genesis: dynamic structures with translational potential. Exp 50. Milkiewicz M, Brown MD, Egginton S, Hudlicka O (2001)
Dermatol 20:605–613 Association between shear stress, angiogenesis, and VEGF in
30. Brown MD, Hudlicka O (2003) Modulation of physiological skeletal muscles in vivo. Microcirculation 8:229–241
angiogenesis in skeletal muscle by mechanical forces: involve- 51. Hudlicka O, Graciotti L, Fulgenzi G, Brown MD, Egginton S,
ment of VEGF and metalloproteinases. Angiogenesis 6:1–14 Milkiewicz M, Granata AL (2003) The effect of chronic skeletal
31. Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, muscle stimulation on capillary growth in the rat: are sensory
Abramsson A, Jeltsch M, Mitchell C, Alitalo K, Shima D, nerve fibres involved? J Physiol Lond 546:813–822
Betsholtz C (2003) VEGF guides angiogenic sprouting utilizing 52. Rivilis I, Milkiewicz M, Boyd P, Goldstein J, Brown MD, Egg-
endothelial tip cell filopodia. J Cell Biol 161:1163–1177 inton S, Hansen FM, Hudlicka O, Haas TL (2002) Differential
32. Hansen-Smith FM, Hudlicka O, Egginton S (1996) In vivo involvement of MMP-2 and VEGF during muscle stretch-versus
angiogenesis in adult rat skeletal muscle: early changes in cap- shear stress-induced angiogenesis. Am J Physiol Heart Circ
illary network architecture and ultrastructure. Cell Tissue Res Physiol 283:H1430–H1438
286:123–136 53. Resnick N, Yahav H, Schubert S, Wolfovitz E, Shay A (2000)
33. Djonov V, Baum O, Burri PH (2003) Vascular remodeling by Signalling pathways in vascular endothelium activated by shear
intussusceptive angiogenesis. Cell Tissue Res 314:107–117 stress: relevance to atherosclerosis. Curr Opin Lipidol 11:167–177
34. Makanya AN, Stauffer D, Ribatti D, Burri PH, Djonov V (2005) 54. Ando J, Yamamoto K (2011) Effects of shear stress and stretch
Microvascular growth, development, and remodeling in the on endothelial function. Antioxid Redox Signal 15:1389–1403
embryonic avian kidney: the interplay between sprouting and 55. Baxter LT, Jain RK (1988) Vascular-permeability and interstitial
intussusceptive angiogenic mechanisms. J Electron Microsc Tech diffusion in superfused tissues—a two-dimensional model. Mi-
66:275–288 crovasc Res 36:108–115
35. Eilken HM, Adams RH (2010) Dynamics of endothelial cell 56. Ravnic DJ, Konerding MA, Huss HT, Wolloscheck T, Pratt JP,
behavior in sprouting angiogenesis. Curr Opin Cell Biol 22: Mentzer SJ (2007) Murine microvideo endoscopy of the colonic
617–625 microcirculation. J Surg Res 142:97–103
36. Wacker A, Gerhardt H (2011) Endothelial development taking 57. Ravnic DJ, Konerding MA, Tsuda A, Jiang X, Huss HT, Pratt JP,
shape. Curr Opin Cell Biol 23:676–685 Mentzer SJ (2007) Structural adaptations in the murine colon
37. Lee CY, Bautch VL (2011) Ups and downs of guided vessel microcirculation associated with hapten-induced inflammation.
sprouting: the role of polarity. Physiology 26:326–333 Gut 56:518–523
38. Iruela-Arispe ML, Davis GE (2009) Cellular and molecular 58. Turhan A, Konerding MA, Tsuda A, Ravnic DJ, Hanidizar D, Lin
mechanisms of vascular lumen formation. Dev Cell 16:222–231 MY, Mentzer SJ (2007) Bridging mucosal vessels associated with
39. Ackermann M, Tsuda A, Secomb TW, Mentzer SJ, Konerding rhythmically oscillating blood flow in murine colitis. Anat Rec
MA (2013) Intussusceptive remodeling of vascular branch angles 291:74–92
in chemically-induced murine colitis. Microvasc Res 87:75–82 59. Tsuda A, Turhan A, Konerding MA, Ravnic DJ, Hanidziar D, Lin
40. Lee GS, Filipovic N, Miele LF, Simpson DC, Lin M, Konerding M, Mentzer SJ (2009) Bimodal oscillation frequencies of blood
MA, Tsuda A, Mentzer SJ (2010) Blood flow shapes intravascular flow in the inflammatory colon microcirculation. Anat Rec
pillar geometry in the chick chorioallantoic membrane. J Angi- 292:65–72
ogenes Res 2:11–20 60. Miele LF, Turhan A, Lee GS, Lin M, Ravnic DJ, Tsuda A,
41. Filipovic N, Tsuda A, Lee GS, Miele L, Lin M, Konerding MA, Konerding MA, Mentzer SJ (2009) Blood flow patterns spatially
Mentzer SJ (2009) Computational flow dynamics in a geometric associated with platelet aggregates in murine colitis. Anat Rec
model of intussusceptive angiogenesis. Microvasc Res 78:286–293 292:1143–1153
42. Hudlicka O, Brown MD, Egginton S (2004) Microcirculation in 61. Stump MM, Jordan GL Jr, Debakey ME, Halpert B (1963)
muscle. Myology: basic and clinical. McGraw-Hill, London, Endothelium grown from circulating blood on isolated intravas-
pp 511–533 cular Dacron hub. Am J Pathol 43:361–367

123
Angiogenesis

62. O’Neal RM, Jordan GL Jr, Rabin ER, Debakey ME, Halpert B 66. Chamoto K, Gibney BC, Lee GS, Lin M, Simpson DC, Vos-
(1964) Cells grown on isolated intravascular Dacron hub; an winckel R, Konerding MA, Tsuda A, Mentzer SJ (2012)
electron microscopic study. Exp Mol Pathol 90:403–412 CD34? progenitor to endothelial cell transition in post-pneu-
63. Gibney B, Chamoto K, Lee GS, Simpson DC, Miele L, Tsuda A, monectomy angiogenesis. Am J Respir Cell Mol Biol 46:283–289
Konerding MA, Wagers A, Mentzer SJ (2012) Cross-circulation 67. Chamoto K, Gibney BC, Lee GS, Ackermann M, Konerding MA,
and cell distribution kinetics in parabiotic mice. J Cell Physiol Tsuda A, Mentzer SJ (2013) Migration of CD11b? accessory
227:821–828 cells during murine lung regeneration. Stem Cell Res 10:267–277
64. Bunster E, Meyer RK (1933) An improved method of parabiosis.
Anat Rec 57:339–343
65. Swenson ES, Price JG, Brazelton T, Krause DS (2007) Limita-
tions of green fluorescent protein as a cell lineage marker. Stem
Cells 25:2593–2600

123

You might also like