You are on page 1of 41

Histol Histopathol (2017) 32: 1239-1279

Histology and
http://www.hh.um.es
Histopathology
From Cell Biology to Tissue Engineering

Review

Morphofunctional basis of the different types


of angiogenesis and formation of postnatal
angiogenesis-related secondary structures
L. Díaz-Flores1, R. Gutiérrez1, M.P. García-Suárez2, F.J. Sáez3,
E. Gutiérrez1, F. Valladares1,6, J.L. Carrasco1, L. Díaz-Flores Jr4 and J.F. Madrid5
1
Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 2Department of Pathology, Hospiten®
Hospitals, Tenerife, 3Department of Cell Biology and Histology, UFI11/44, School of Medicine and Dentistry, University of the Basque
Country, UPV/EHU, Leioa, 4Department of Physical Medicine and Pharmacology, Faculty of Medicine, University of La Laguna,
Tenerife and 5Department of Cell Biology and Histology, School of Medicine, Campus of International Excellence “Campus Mare
Nostrum”, IMIB-Arrixaca, University of Murcia, Murcia and 6CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III,
Madrid, Spain

Summary. We review the morpho-functional basis of sprouting phase, encompassing EC migration (concept
the different types of angiogenesis and report our and characteristics of endothelial tip cells, tip cell
observations, including the formation of angiogenesis- selection, lateral inhibition, localized filopodia
related secondary structures. First of all, we consider the formation, basal lamina degradation and extracellular
following issues: a) conceptual differences between changes facilitating EC migration), EC proliferation
angiogenesis and vasculogenesis, b) incidence of (concept of endothelial stalk cells), pericyte
angiogenesis in pre- and postnatal life, c) regions of mobilization, proliferation, recruitment and changes in
vascular tree with angiogenic capacity, d) cells CD34+ adventitial stromal cells and inflammatory cells,
(endothelial cells, pericytes, CD34+ adventitial stromal tubulogenesis, formation of a new basal lamina, and
cells of the microvasculature and inflammatory cells) vascular anastomosis with capillary loop formation, and
and extracellular matrix components involved in c) vascular remodelling and stabilization phase (concept
angiogenesis, e) events associated with angiogenesis, f) of phalanx cells). Subsequently, the concept, incidence,
different types of angiogenesis, including sprouting and events and mechanisms are considered in the other forms
intussusceptive angiogenesis, and other angiogenic or of angiogenesis. Finally, we contribute the formation of
vascularization forms arising from endothelial precursor postnatal angiogenesis-related secondary structures: a)
cells (postnatal vasculogenesis), vasculogenesis intravascular structures through piecemeal angiogenesis,
mimicry, vessel co-option and piecemeal angiogenesis. including intravascular papillae in vessel tumours and
Subsequently, we consider the specific morpho- pseudotumours (intravascular papillary endothelial
functional characteristics of each type of angiogenesis. hyperplasia, vascular transformation of the sinus in
In sprouting angiogenesis, we grouped the events in lymph nodes, papillary intralymphatic angioendo-
three phases: a) activation phase, which includes thelioma or Dabska tumour, retiform hemangio-
vasodilation and increased permeability, EC, pericyte endothelioma, hemangiosarcoma and lymphangio-
and CD34+ adventitial stromal cell activation, and sarcoma), vascular septa in hemorrhoidal veins and
recruitment and activation of inflammatory cells, b) intravascular projections in some tumours; b) arterial
intimal thickening; c) intravascular tumours and
Offprint requests to: Lucio Díaz-Flores, Departamento de Ciencias pseudotumours (e.g. intravenous pyogenic granulomas
Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, and intravascular myopericytoma); d) vascular
Tenerife, Spain. e-mail: kayto54@gmail.com glomeruloid proliferations; and e) pseudopalisading
DOI: 10.14670/HH-11-923 necrosis in glioblastoma multiform.
124
Angiogenesis and related secondary structures

Key words: Stromal cells, Fibroblasts, Fibrocytes,


Telocytes, Sprouting Angiogenesis, Pericytes, endothelial precursor cell participation. Postnatal
Endothelial cells, Intussusceptive Angiogenesis, vasculogenesis by EPCs).
Vasculogenesis, Piecemeal Angiogenesis
3. Incidence of angiogenesis in pre- and postnatal
life
1. Definition
Angiogenesis is indispensable in embryonic and
Angiogenesis is a process characterized by the foetal development, as well as in a large number of
formation of new vessels from an established normal and pathological processes during postnatal life.
microvasculature and is therefore responsible for Therefore, the area involved in the study of angiogenesis
vascular network expansion and remodelling (Folkman, is far-reaching, covering numerous fields and many
2003). disciplines.
Conventionally, when the term angiogenesis is not During embryonic and foetal development, new
specified, it generally refers to blood vessels (blood vessel formation or neovascularization from pre-existing
vessel angiogenesis). In lymphatic vessels, the more microvasculature is an important event and has been
precise term of lymphangiogenesis is used. studied at morphologic (Fig. 1A) and molecular levels
(for review: Betz et al., 2016).
2. Conceptual differences between angiogenesis and Angiogenesis is generally a quiescent process in the
vasculogenesis adult organism. Nevertheless, it may occur rapidly in
several normal circumstances. For example, the need for
Vasculogenesis and angiogenesis are two principle additional vasculature is imposed on the cyclic evolution
types of mechanisms that intervene in vessel of transient structures in the female reproductive system
development and growth. Classically, the concept of (for review, see Rizov et al., 2017), including the
vasculogenesis was restricted to the embryonic sequential maturation of ovarian pre-ovulatory follicles
development of certain vessels, such as the dorsal aortae (Kim et al., 2017) and subsequent development of the
and the posterior cardinal veins (Sabin, 1920), and is corpora lutea (Berisha et al., 2016; Woad and Robinson,
defined as capillary development (primary capillary 2016), cyclic extensions and repair of the endometrium
plexus) from differentiating endothelial cells (ECs) in (Zhang et al., 2016), decidual transformation,
situ (Risau and Flamme, 1995; Auerbach et al., 1997; Jin implantation and placentation (Okada et al., 2014; Chen
and Patterson, 2009). Currently, the concept of et al., 2017), and mammary gland changes during
vasculogenesis is broader and is considered as occurring pregnancy, lactation and involution (Andres and Djonov,
in pre- and postnatal life, with neo-vessel formation 2010; VanKlompenberg et al., 2016).
from endothelial progenitor cells (EPCs). In the embryo, Furthermore, angiogenesis is an important
vasculogenesis takes place in blood islands, which component of many pathological processes, such as
originate from the splachnopleuric mesoderm. Thus, chronic inflammation, regeneration, wound healing,
after segregation from the mesoderm, some organization of thrombi, collateral circulation
mesenchymal cells transform into nests of isolated cell development and neoplasias. Moreover, angiogenesis
cords of hemangioblasts, which are the precursors of occurs in severa1 conditions in which the term
both ECs and blood cells. The peripheral cells of the ‘angiogenic disease’ has been proposed, since an
angioblastic masses differentiate into ECs, while the abnormality of capillary growth is their principal
blood precursor cells are found in the centre of the blood pathological feature (Folkman, 1989). Angiogenic
islands, where a lumen develops (Axnick and Lammert, diseases include hemangiomas, psoriasis (Heidenreich et
2012). The newly formed lumens soon coalesce (Risau al., 2009; Capkin et al., 2017), scleroderma (Mulligan-
and Flamme, 1995). Finally, smooth muscle cells and Kehoe and Simons, 2008; Manetti et al., 2016), arthritis
pericytes are aggregated from undifferentiated (Szekanecz et al., 2010; Elshabrawy et al., 2015),
mesenchyme. atherosclerotic plaque (Camaré et al., 2017), diabetic
In the current concept of vasculogenesis, new blood retinopathy (Capitão and Soares, 2016), neovascular
vessels may develop from circulating and/or bone glaucoma (Kim et al., 2015) and ischaemic diseases. For
marrow-derived CD34+ EPCs (C-EPCs and BM-EPCs, example, the newly formed capillaries invade the joint
respectively) (Ashara et al., 1997; Gehling et al., 2000; and cartilage in arthritis, and the vitreous in diabetes,
Asahara and Isner, 2002; Grant et al., 2002; Pelosi et al., with secondary effects such as cartilage destruction and
2002), from CD34+, CD110+ cord blood cells (Aoki et bleeding, respectively. The role of angiogenesis in
al., 2004), and from stem cells in a vasculogenic zone neoplasias is of great interest, especially in the
(between vessel adventitia and the media layer) (Zengin progressive growth and metastases of solid tumours
et al, 2006). Therefore, the recruitment, integration, (Folkman, 1985), and has resulted in numerous
migration and maturation of circulating EPCs with descriptive studies on angiogenesis, as well as on the
vascular development is considered to be a type of possible mechanisms involved in this process (for
angiogenesis (see section Postnatal angiogenesis with review: Hillen and Griffioen, 2007; Makrilia et al., 2009;
Krishna Priya et al., 2016; Ronca et al., 2017).
12
Angiogenesis and related secondary

Fig. 1. Angiogenesis in pre- (A) and postnatal (B and C) life. A. An embryonic sprouting vessel (arrow) originating from a vasculogenic vessel (vv). B.
A sprouting vessel (arrow) originating from a postcapillary venule (pv) in the adipose tissue. C. A sprouting vessel (arrow) originating from the rat
femoral vein (fv) induced by peri-venous administration of PGE2 and glycerol. D. Size of the rat femoral artery (black asterisk) and vein (white asterisk)
(scale in mm). Scale bars: A, 20 µm; B, 15 µm; C, 10 µm.
124
Angiogenesis and related secondary structures

4. Regions of vascular tree with angiogenic capacity


(mainly venous side of circulation) and transmission of mechanical forces (Tillet et al.,
2005; von Tell et al., 2006; Díaz-Flores et al., 2011a), b)
The regions with angiogenic capacity are the most release of vesicles (Fig. 2D), such as multivesicular
ubiquitous and have a common characteristic: the bodies, shed vesicles and exosomes, which contain
presence, within or near, of an active preexisting several molecules including microRNA (Dellett et al.,
microvasculature, where angiogenesis begins as a result 2017; Todorova et al., 2017), and c) paracrine regulators,
of neighbouring angiogenic stimuli. Thus, the mother including vascular endothelial growth factor A (VGFA),
vessel microvasculature forms part of a general sphingosine-1-phosphate, angiopoietin 1 and 2,
angiogenic-inflammatory-repair system in which the transforming growth factor beta (TGF beta),
vessels not only intervene in new capillary formation but semaphoring 3-A and several cytokines (Caporali et al.,
also in the recruitment of inflammatory cells and in the 2017). These cells have the ability to modulate cellular
contribution of matrix-forming cells (fibroblasts- interactions during angiogenesis and vessel stabilization
myofibroblasts, osteoblasts, chondroblasts), and (Diaz-Flores et al., 1991; 2009a).
contractile and adipose cells. Interestingly, sprouting The major fibroblastic cell components of adventitial
angiogenesis mainly occurs in the venous side of the vessel walls are CD34+, CD31-, CD146- and CD45-,
circulation, above all in the postcapillary venules (Fig. and are considered MSC progenitors (Corselli et al.,
1B) (Ausprunk and Folkman, 1977; Díaz-Flores et al., 2012; Lin and Lue, 2013; Díaz-Flores et al., 2014;
1992, 1994a) with a negative pressure, an important fact 2015a,b, 2016a,b). These cells have received numerous
in initial stages of this process, in which sprouts are names, including fibroblasts, fibrocytes, dendrocytes,
immature (Díaz-Flores et al., 1992; 1994a) (see below). keratocytes, telocytes and stromal, dendritic, adventitial,
Although some authors restrict the origin of newly supraadventitial, perivascular, paravascular and
formed vessels, pointing out that they only arise from the delimiting cells (for review: CD34+ stromal cells/
microvessels of the venous side of the circulation, fibroblasts/fibrocytes/telocytes, Díaz-Flores et al., 2014).
vessels of greater calibre, such as the rat femoral vein, Some authors point out that the microcirculation
with a discontinuous internal elastic lamina and smooth contains no CD34+ adventitial stromal cells. However,
muscle cells in their media layer, are also capable of these cells extend continuously through most of the
contributing to angiogenesis (Fig. 1C,D) (Diaz-Flores et vascular system. Therefore, a very thin rudimentary
al., 1994a,b, 2017; Madrid et al., 1998). adventitia is found in several microvessels (Fig. 2A),
In avascular tissues (e.g. cartilage and cornea) and in except for some small vessels in certain locations.
fibrin deposits (e.g. wounds), vessels can only originate Examples of microvessels with CD34+ adventitial
from neighbouring vascularized tissues (Díaz-Flores et stromal cells are those of the reticular dermis and
al., 2012a). In this case, the intensity of vascular submucosa, muscular propria, and serosa of the
penetration into an avascular tissue depends on the intestinal wall and gallbladder (Fig. 2A-C). Examples of
properties of the latter (e.g. inhibitory action of microvessels without CD34+ fibroblasts are those of the
antiangiogenic substances, as occurs in cartilage). papillary dermis, and the intestinal and gallbladder
mucosa (Fig. 2E) (Díaz-Flores et al., 2014). The
presence of these ‘delimiting fibroblasts’ around small
5. Cells and extracellular matrix involved in vessels is easily demonstrated by electron microscopy
angiogenesis (Fig. 2C). In the adventitia of large vessels, CD34+
fibroblasts/stromal cells appear in several layers, which
Cells involved in angiogenesis include those of the decrease in number as vessel size reduces and, when
vessel wall, circulating precursor cells, inflammatory present in the capillaries, are arranged in a single layer.
cells and cancer cells. The cells in the vessel wall are Although CD34 is also intensely expressed in ECs, both
ECs (intimal layer), pericytes/smooth muscle cells types of cells are easily distinguished by this different
(mural cells in the media layer) and CD34+ fibroblasts/ response to CD31 (anti-CD31 stains ECs but not CD34+
stromal cells (adventitial or external layer) (Fig. 2A-C). adventitial stromal cells), and by their characteristics and
The participation of ECs and pericytes/smooth muscle respective locations in the vessel wall. Pericytes and
cells in angiogenesis has received great attention in the SMCs are positive for anti-αSMA and negative for anti-
literature (for review: Diaz-Flores et al., 2009a; Ribatti CD34. Since CD34 is expressed in stromal cells of the
et al., 2011); not so the CD34+ adventitial adventitia and in ECs of the tunica intima, the CD34-
fibroblasts/stromal cells. Here we consider the stained vessels show a double-ring appearance (stained
coordination and interactions between pericytes and endothelium and adventitia), owing to two concentric
ECs, and the role of CD34+ adventitial stromal cells. circles sandwiching the unstained media layer (smooth
The coordination and interactions between pericytes muscle or pericytic layer) (Fig. 2A) (Lin et al, 2010;
and ECs may be as follows: a) direct contact, which Maumus et al., 2011; Díaz-Flores et al., 2014). A similar
includes interdigitations (peg and socket) (Fig. 2D), gap image is obtained with double stained (CD34/brown and
junctions, and adherent and occluding plaques, leading αSMA/red) vessels, although with red staining in the
to intercellular communications, passage of molecules media layer (Fig. 2B). Some CD34+ fibroblasts/stromal
cells in the vascular wall may extend their processes into
12
Angiogenesis and related secondary

Fig. 2. Vessel wall


cells involved in
angiogenesis.
A. CD34 expression in
a microvessel with a
double ring
appearance (CD34
stained endothelial -
ec- and stromal
adventitial cells -sc-
sandwiching the
unstained mural cells
of the media layer).
B. A double stained
vessel with anti-CD34
(staining ECs -ec-and
stromal adventitial
cells -sc - brown) and
anti-αSMA (staining
mural cells -mc- red).
Note the double ring
appearance of CD34
stained cells
sandwiching αSMA
mural cells of the
media layer.
C. Electron
photomicrograph
showing projections of
adventitial stromal
cells (sc), a pericyte
(p) and endothelial
cells (ec).
D. Interdigitations (peg
and socket) (arrow)
between the process
of a pericyte (p) and
an endothelial cell
(ec). A multivesicular
body in formation is
also observed in this
interdigitation. E. In
the mucosae of the
bladder, double
staining with anti-
CD34 (brown) and
anti-αSMA (red) show
capillaries devoid of
CD34+ stromal cells.
Only one vessel
presents αSMA+
perivascular mural
cells (asterisk). Scale
bars: A, 10 µm; B, 15
µm; C, 2 µm; D, 1 µm;
E, 35 µm.
124
Angiogenesis and related secondary structures

surrounding tissues and vice versa. Frequently, these


extensions associate with other tissue cells (native or 7.1.2. Events of new blood vessel formation in
recruited). sprouting angiogenesis
The other cells involved in angiogenesis will be
considered in other sections, thus circulating precursor Sprouting angiogenesis is a complex multistep
cells in “Postnatal angiogenesis with endothelial process, in which the following overlapping events have
precursor cell participation”, the inflammatory cells in been described: a) activation of vascular cells, b) EC
“Sprouting angiogenesis” and the cancer cells in migration, c) proteolytic destruction of extracellular
“Vasculogenic mimicry”. matrix, including basement membrane degradation, d)
The pre-existing or incorporated components in the EC proliferation, e) pericyte mobilization, proliferation
extracellular matrix comprise collagens (including and recruitment, f) vascular lumen formation, g) changes
collagen I, III, IV, V, VIII, XV, XVIII – mainly collagen in extracellular matrix with development of a new
IV), elastin, laminins, perlecan, hyaluronan, fibronectin, basement membrane, and h) vascular maturation and
entactin/nidogen, TsP, SPARC, and plasminogen. In the remodelling, with capillary network formation, eventual
basement membrane of quiescent microvasculature there organization of larger microvessels, and involution of
are several proteins, including laminins, type IV most newly formed vessels. Vasodilation and an
collagens, perlecan, nidogens, SPARC, fibulins, inflammatory response may precede and accompany the
trombospondins, and collagen types VIII, XV and process. Furthermore, during angiogenesis and vascular
XVIII, (for review: Seano and Primo, 2015; Randles et involution, other phenomena may occur in the
al., 2017). During angiogenesis, these molecules may interstitium, mainly the participation of precursor
undergo local modifications (matricryptic sites of Davis) stromal cells, which may originate matrix-forming cells
(Davis et al., 2000) by the action of proteases, such as (fibroblast-myofibroblasts, osteoblasts, chondroblasts),
MMPs, and their extracellular matrix fragments contractile cells (SMCs, myofibroblasts and myointimal
originate matrikines (Bellón et al., 2004), which are pro- cells) and adipose cells.
or anti-angiogenic factors (see below). In this review, we group these events in three
phases: Activation phase, Sprouting phase and
6. Events associated with angiogenesis Stabilization phase.

Angiogenesis is a complex biological process, which 7.1.3. Activation phase


may be associated with coagulation, inflammation,
immunity, tissue repair and remodelling. Despite the Vascular dilation, increased vascular permeability,
continuous and orderly nature of the process, these activation of vascular cells and diapedesis of leukocytes
findings occur in several overlapping phases, according form part of the activation phase. Below, we outline
to predominant mechanisms. Therefore, several these findings, which precede and accompany sprouting
phenomena, such as vascular dilation and increased angiogenesis.
vascular permeability, coagulation, activation of
numerous cell types, including vascular and 7.1.3.1. Vasodilation and increased permeability
inflammatory cells, as well as modifications of the
extracellular matrix, take place during new-vessel Dilation and increased permeability of the blood
formation. venules and capillaries occur rapidly in the initial phase
of angiogenesis (Fig. 3A), in which the opening of
7. Types of angiogenesis interendothelial cell junctions is observed (Fig. 3B).
Indeed, in response to proangiogenic factors, such as
Several types of angiogenesis have been described, VEGF (see below), capillaries and postcapillary venules
including sprouting and intussusceptive angiogenesis, show vasodilation and increased vascular permeability.
and other forms of vascularization by means of For example, in corneal vascularization induced by
circulating endothelial precursor cells (EPCs) (postnatal silver nitrate, dilation takes place within an hour
vasculogenesis), vasculogenic mimicry and vascular co- (McCracken et al., 1979) and intensifies progressively.
option. Following, we describe these types of The changes in vascular permeability during
angiogenesis, and we also consider a recently reported angiogenesis were studied using the intravenous
form: piecemeal angiogenesis, which intervenes in the injection of different markers, such as colloidal carbon
formation of angiogenesis-related secondary structures. or Monastral blue to label leaky vessels (Fig. 3C)
(Garbett and Gibbins, 1987; Diaz-Flores et al., 1992).
7.1. Sprouting angiogenesis Increased vascular permeability is also demonstrated
by the presence of extravascular fibrin and dilated
7.1.1. Definition lymphatics. Extravascular fibrin deposits, which are
found during inflammation, wound healing (Madri and
Sprouting angiogenesis is the process by which Pratt, 1988) and in the periphery of tumours (Nagy et al.,
vessel sprouts grow out of a pre-existing vasculature. 1989), are important during angiogenesis. Thus,
neovascularization is induced by fibrin gels in vitro and
12
Angiogenesis and related secondary

Fig. 3. Phenomena in the activation phase of angiogenesis. A. A dilated and congestive capillary between oedematous spaces. Note perivascular
activated CD34+ adventitial stromal cells (arrows). B. Ultrastructural image of the opening of the interendothelial junctions (arrow). C. Demonstration of
vascular permeability during angiogenesis. Under electron microscopy, Monastral Blue particles (asterisk) are observed between endothelial cells (ec)
and pericytes (p) in a leaky vessel. D. A dilated vessel showing early migration of leukocytes (L) between an opened interendothelial cell junction
(arrow). Note an activated CD34+ adventitial stromal cell (sc) with a nucleus increased in size and with a prominent nucleolus. E. Portion of an
activated mast cell with degranulation between a process of a stromal cell (sc) and pericytes (p). Endothelial cell: ec). Scale bars: A, 25 µm; B, D, E, 1
µm; C, 2 µm.
124
Angiogenesis and related secondary structures

in vivo (Dvorak et al., 1987). Also, it has been


demonstrated that cultured ECs synthesize fibronectin
and that growing capillaries produce fibronectin in situ coverage, leading to vessel destabilization (Greenberg et
(Clark et al., 1982a). al., 2008). In addition, VEGF induces overexpression of
Ang2 in ECs, which also facilitates pericyte dissociation,
and vessel destabilization and regression (Zhang et al.,
7.1.3.2. Activation of ECs 2003; Cao et al., 2007). Pericyte dissociation may also
be facilitated by migrating macrophages.
In pre-existing vasculature (parent vessels), the
earliest morphological changes in the normally quiescent
ECs consist of hypertrophy with bulging in the vascular 7.1.3.4. Activation of CD34+ adventitial stromal
cells
lumen, nuclear enlargement, nucleolar prominence,
dispersal of the ribosomes into their free form, and an
increase in the number of organelles and projections In this phase, CD34+ adventitial stromal cells
formation (Ausprunk and Folkman 1977; McCracken et separate from the vascular wall, increase in size and
al., 1979; Díaz-Flores et al., 1992, 1994a). The ECs appear as large, plump, stellate or ovoid cells in
projecting towards the interstitium have been described oedematous spaces (Fig. 3A). The nuclei also increase in
as filopodia (Kurz et al., 1996; Ruhrberg et al., 2002). size and show one or two prominent nucleoli (Fig. 3D)
Increased endothelial DNA-synthesis occurs at the onset (Díaz-Flores et al., 2014, 2015a,b, 2016a,c). These cells
of angiogenesis (Burger et al., 1983; Díaz-Flores et al., secrete VEGF, bFGF, PDGF-α and IL-8, which
1992) and an increased proliferative index is contribute to the induction of an angiogenic phenotype
demonstrated with Ki67. In addition, modifications of in ECs, and promote angiogenesis in vivo and in vitro
the tight junctions (organized by claudins, occludins and (Hartlapp et al., 2001; Miranville et al., 2004) (e.g.
junctional adhesion molecules) and adherens junctions adipose derived stromal cells support postnatal
(organized by catenins and cadherins), as well as neovascularization - Miranville et al., 2004).
extracellular and intracellular associations play an
important role in EC activation (Dejana et al., 2008; 7.1.3.5. Recruitment and activation of inflammatory
Dejana and Orsenigo, 2013; Dejana and Lampugnani, cells
2014). All these phenomena facilitate the recruitment of
inflammatory cells (Sprague and Khalil, 2009) and lead Inflammation may play an important role in
to local deposition of serum proteins, originating a angiogenesis, with vascular and leukocyte
fibrin-rich provisional matrix (Mehta and Malik, communication, and inflammation-induced angiogenesis
2006). (for review, Szade et al., 2015; Kreuger and Phillipson,
2016). Intravascular accumulation of platelets and
Tissue hypoxia activates ECs and angiogenesis- polymorphonuclear leukocytes (PMNs) occurs within a
promoting signals, mainly VEGF, through stimulation of few hours, with early diapedesis of leukocytes outside
VEGFR2. VEGF acts not only in this EC activation, but the vessel lumen. Thus, before and during vascular
also in other phases of vessel sprouting (see below). sprouting, inflammatory cells are observed adhering to
the endothelium of the parent vessels, as well as passing
7.1.3.3. Activation of pericytes through the endothelial junctions and the pericyte-
endothelial space. Between 1 and 6 h after angiogenic
During the initial phase of angiogenesis, activated stimuli, the PMNs predominate. Thereafter, the number
pericytes in pre-existing vasculature bulge, shorten their of monocytes/macrophages increases, while the number
processes and increase their somatic volume, adopting of PMNs decreases dramatically. Frequently, the
an angiogenic phenotype (Díaz-Flores et al., 1992, monocytes/macrophages, either individually or in small
1994a). Their nuclei display some folding and the clusters of two or three, simultaneously appear trapped
nucleoli become prominent. The cytoplasm acquires between the ECs and the pericytes, within the basement
numerous ribosomes, either singly or in aggregates, and membrane (Diaz-Flores et al., 2009b).
multiple profiles of rough endoplasmic reticulum are
observed. Micropinocytic vesicles are widely distributed VEGF, CSF-1 (colony stimulating factor 1), PIGF
along the cell membrane. Moreover, pericytes may show (placental growth factor) and chemokines participate in
increased DNA synthesis, and a sudden and intense leukocyte recruitment. For example, in monocytes,
pericyte proliferation occurs (Diaz-Flores et al., 1992). CXC, CC, C and CX3C chemokines act through
Proliferation markers, such as Ki-67, also demonstrate a transmembrane G protein-coupled receptors, mainly
higher proliferative index in pericytes. In the parent CCL2 (C-C chemokine ligand 2)/CCR2 (Ding et al.,
vessels, the pericyte basement membrane is disrupted 2012). The new vessels may arise following the
and fragmented, sometimes away from the pericyte cell margination and diapedesis of the leukocytes, which do
surface. Many pericytes project into the perivascular not seem to be essential for the initiation and
space and appear detached from the vessel walls (Díaz- continuation of angiogenesis. For example, corneal
Flores et al., 1992). vascularization has been produced in the absence of
leukocytes in rats and rabbits (Sholley et al., 1978).
VEGF is a negative regulator of pericyte function Nevertheless, leukocytes may have a facilitatory or
and vessel maturation, and facilitates the loss of pericyte augmentative role in vascularization, by the secretion of
cytokines, chemokines, growth factors and matrix-
12
Angiogenesis and related secondary

degrading enzymes (see inflammatory cells in sprouting


phase). (Gerhardt et al., 2003; Weavers and Skaer, 2014). In
Activated mast cells (Fig. 3E) play an important role angiogenic sprouts, endothelial tip cells are therefore
in angiogenesis by producing and releasing pro- leader migrating (invading) cells (Fig. 4A,B) that sense
angiogenic factors (for review: Maltby et al., 2009; the environment, and precede and guide endothelial stalk
Ribatti and Crivellato, 2012a; Cimpean et al., 2017). cells. In the initial phase of neo-vascularization, tip cells
Thus, mast cells may release VEGF stored in their degrade cell-cell junctions (VE-cadherin and tight
granules or in a degranulation-independent form, junction protein-1), and contribute to extracellular
enhancing EP2 receptor by PGE2 (Boesiger et al., 1998; matrix proteolysis (Arroyo and Iruela-Arispe, 2010) and
Abdel-Majid and Marshall, 2004). Other angiogenic degradation of the vascular basement membrane of the
mediators stored in their granules, such as histamine, parent vessel. Subsequently, they protrude through the
heparin, IL-8, TNF-alpha, TGF-beta, NGF and FGF-2 vessel wall and begin to migrate in a polarized form into
may be released by degranulation (Kanbe et al., 2000). the interstitial space towards the angiogenic stimulus.
Finally, mast cell granules contain tryptase and chymase, Tip cells show an EC transient modified phenotype
which participate in the degradation of extracellular (De Smet et al., 2009), with numerous free
matrix components. In particular, tryptase stimulates the polyribosomes and abundant intermediate filaments (Fig.
proliferation of endothelial stalk cells and facilitates 4A), as well as filopodia that sense attractant or repellent
tubulogenesis, activation of MMPs, and VEGF and signals and guide polarized migration (Ruhrberg et al.,
FGF-2 release (Blair et al., 1997). 2002; Gerhardt et al., 2003; Lu et al., 2004; De Smet et
al., 2009; Geudens and Gerhardt, 2011). They proliferate
7.1.4. Sprouting phase minimally and express the tyrosine-kinase receptor
VEGFR2, VEGFR3/FIT-4, Delta-like 4 (DLL4), PDGF
This phase includes EC migration, extracellular beta, EC specific molecule I ECM-I1), homolog b
matrix changes with basement membrane degradation (netrin receptor UNC5b) and angiomotin. Notch-
facilitating EC migration, EC proliferation, pericyte and signalling activity is low (Gerhardt et al., 2003; Claxton
CD34+ stromal cell mobilization, proliferation, and Fruttiger, 2004; Lu et al., 2004; Siekmann and
differentiation and recruitment, inflammatory cell Lawson, 2007; Suchting et al., 2007; Hellstrom et al.,
participation, tubulogenesis (vascular lumen formation), 2007a; Tammela et al., 2008; Zheng et al., 2014a). In
formation of a new basement membrane and sprouting early migration, ECs show extracellular-matrix-
connection. During the process in which the ECs degrading podosomes on their surface. These specialized
protrude and the vascular basement membrane is cell-matrix contacts contain adhesive (e.g. stalin,
degraded, scant microscopic bleeding may occur. vinculin, paxilin) and proteolytic (e.g. MT1-MMP)
molecules, surrounding a core rich in actin (Moreau et
7.1.4.1. EC migration al., 2006; Osiak et al., 2005). Individual podosomes and
podosome rosettes are induced by VEGF-A, which up-
Traditionally, it was considered that blood vessels regulates integrin α6β1 (Primo et al., 2010; Seano et al.,
grew through EC movement (His, 1868). This concept of 2014), and participate in the degradation of the basement
EC migration is maintained as an important step during membrane and in the control of blood vessel branching
angiogenesis (Ausprunk and Folkman, 1977). Most (Seano et al., 2014).
researchers agree that these changes precede endothelial Attractant signals, mainly VEGF-A, and repulsive
replication, and that migration and mitoses are signals, mainly semaphoring 3A, guide sprout migration
independent phenomena (Sholley et al., 1977a,b; Wall et (see below). The chemotactic behaviour of ECs at the
al., 1978). Therefore, angiogenic stimuli may operate tips of growing vessels is facilitated by the secretion of
through chemotaxis, and EC mitosis may be a secondary plasminogen activator and collagenases (Moscatelli et
event (Sholley et al., 1977a,b). al., 1981). EC migration, in response to the extracellular
The current concept of endothelial tip, stalk and matrix, depends on the integrin family of cell adhesion
phalanx cells is important to understand these findings receptors (Leavesley et al., 1993). Indeed, EC
(Gerhardt et al., 2003; Ribatti and Crivellato, 2012b) attachment, spreading and migration are mediated by
(see below). In this phase, endothelial tip cells are integrins alfa2B1 and alfa4B3 (Leavesley et al., 1993).
predominantly involved in EC migration, whereas stalk EC migration on collagen and vitronectin is mediated by
cells are involved in proliferation and have the capacity alfa4B3 and occurs in a calcium-dependent mechanism,
to acquire a lumen-forming cell phenotype. Tip cells while collagen recognition by alfa2B1 promotes EC
were previously known as migrating, leader and migration in the absence of calcium (Leavesley et al.,
projecting ECs. The term filopodia was also applied 1993).
(Kurz et al,, 1996; Ruhrberg et al, 2002). Important findings during EC migration are tip cell
selection with lateral inhibition, localized filopodia
7.1.4.1.1. Concept and characteristics of endothelial tip formation and sprouting guidance.
cells. Currently, the term tip cells is used for specialized
cells at the tips of a wide variety of developing 7.1.4.1.2. Tip cell selection and lateral inhibition. Not all
structures, predominantly of tube or branched formations activated ECs acquire the tip cell phenotype, since the
ECs that participate in migration are selected by a
12
Angiogenesis and related secondary

Fig. 4. EC migration in sprouting phase. A. An endothelial tip cell (tc), devoid of basement membrane, is observed in an angiogenic vessel. Note
numerous free polyribosomes and abundant filaments. In the insert, an image under light microscopy of a migrating CD34-stained EC. B. Migrating
CD34+ ECs with projections in their tips (arrows). C. Double stained microscopic section with anti-CD34 (staining endothelial cells - brown) and anti-
αSMA (staining pericytes - red). Note the absence of pericytes around the migrating and neighbouring ECs (arrow). Scale bars: A, 1 µm; B, C, 14 µm.
12
Angiogenesis and related secondary

process in which EC stimulated migration and lateral


inhibition occur. VEGF, Dll4 (delta-like 4)/Notch Remodelling of the cytoskeleton, mainly down-
signalling and the ligand Jagged I participate in this regulation of myosin II contraction, determined by
process of tip cell phenotype specification and lateral extracellular matrix properties, facilitates filopodia
inhibition with stalk cell phenotype acquisition formation (Gerhardt et al., 2003; Fisher et al., 2009;
(Hellström et al., 2007b; Leslie et al., 2007; Lobov et al., Myers et al., 2011). Heparin-binding VEGF isoforms,
2007; Siekmann and Lawson, 2007; Suchting et al., ephrin B2 and a small GTPasa of the Rho family
2007; Bentley et al., 2008; Benedito et al., 2009; De (CDC42) mediate internalization of VEGFR2, and
Smet et al., 2009; Potente et al., 2011). VEGF activates participate in the directionality and extension of
VEGFR2 (De Smet et al., 2009) and is an important filopodia, as well in actin remodelling (Ruhrberg et al.,
inducer of tip cell selection and formation. Thus, only 2002; Etienne-Manneville, 2004; Sawamiphak et al.,
ECs exposed to the highest VEGF levels become tip 2010; Fantin et al., 2015). The non-tyrosine kinase
cells (VEGF activation of VEGFR2 predominates in this transmembrane protein, neuropilin 1(NRP1) is essential
mechanism). VEGFR2 signalling induces Dll4 for extracellular matrix-induced CDC42 activation
expression, which binds to NOTCH receptor on adjacent (Fantin et al., 2015). Pericytes may or may not be
ECs and activates NOTCH signalling. The latter inhibits present near the tip cells (Fig. 4C).
VEGFR2 and VEGFR3 in these adjacent ECs,
preventing tip cell formation and promoting a stalk cell 7.1.4.1.4. Basement membrane degradation and
phenotype in immediately neighbouring ECs (Williams extracellular changes facilitating EC migration. During
et al., 2006a; Hellstrom et al., 2007b; Leslie et al., 2007; angiogenesis, local proteolysis of the basement
Lobov et al., 2007; Suchting et al., 2007). The ligand membrane of the parent vessels is observed, which is a
Jagged I antagonizes Dll4 and controls the tip cell necessary step for EC migration (Ausprunk and
number (Benedito et al., 2009). Determination of stalk Folkman, 1977). Complete disintegration occurs on the
cell phenotype is also facilitated by bone morphogenetic side closest to the angiogenic stimulus, coinciding with
protein signalling pathway (Moya et al., 2012). Taking those areas in which the ECs start to grow outwards
into consideration the above, the mechanism that (Fig. 4A), while subtle alterations appear around the
facilitates the selection of the tip cells may be a small whole circumference of the parent vessel. Therefore, EC
imbalance in local VEGF concentration. A time-based sprouts have no basement membrane, but a
formulation of EC behaviour (temporal adaptation) has homogeneous provisional substratum with altered
been hypothesized for the cell decisions (Bentley and proteoglycans (Clark et al., 1982b). Proteases act in this
Chakravartula, 2017). Therefore, this competitive process (Moscatelli and Rifkin, 1988; Ghajar et al.,
cellular mechanism limits the number of sprouts 2008; van Hinsbergh and Koolwijk, 2008), and matrix
(excessive branching) (Hellström et al., 2007b; Lobov et metalloproteinases (MMPs) and ADAM family proteins
al., 2007; Suchting et al., 2007; Jakobsson et al., 2010; are the main proteases of the metzincin superfamily that
Geudens and Gerhardt, 2011) and facilitates sprout control capillary morphogenesis. MMPs involved in
guidance and the change of tip cells for other cells these functions include membrane-type matrix
within the stalk region (relative levels of VEGFR1 and metalloproteinases (MT1-MMP, MT2-MMP/MMP-15,
VEGFR2) (see below). FGF might also control MT3-MMP), and MMP-2, MMP-3, MMP-7, MMP-9
endothelial tip cell migration (Vitorino and Meyer, and MMP-13. Among these MMPs, the MT1-MMP is
2008). Moreover, several inflammatory signals that act the main pericellular fibrinolisin and collagenase
in vascular permeability also participate in EC activation involved in EC sprouting (Hiraoka et al., 1998; Zhou et
and in extracellular matrix proteolysis by endothelial tip al., 2000; Yana et al., 2007), and crosstalk between neo-
cells. These factors include TNF alpha, CCL2/MPC-1, vessels and mural cells directs the site-specific
bradiquinin, nitric oxide, PGE2 and sphingosine-1- expression of MT1-MMP to endothelial tip cells (Yana
phosphate (SIP) (Langlois et al., 2004; Galvez et al., et al., 2007). The extracellular-matrix-degrading
2005; Lee et al., 2006; Petrovic et al., 2007; Alfranca et podosomes (see above) participate in the degradation of
al., 2008; Sainson et al., 2008). Likewise, the action of the basement membrane (Primo et al., 2010; Seano et al.,
some inflammatory cells (e.g. monocyte/macrophage 2014), since individual podosomes and podosome
MPPs) induces tip cell phenotype, guides tip cells and rosettes, induced by VEGF-A, which up-regulate
cooperates with neovascularization (Anghelina et al., integrin α6β1, contain proteolytic molecules, mainly
2006; Chung et al., 2009). MT1-MMP (Primo et al., 2010; Seano et al., 2014).
Likewise, the components of the microvascular
7.1.4.1.3. Localized filopodia formation in tip cells. extracellular matrix, such as laminins and collagens
Filopodia are slender, polarized, highly dynamic plasma- undergo dramatic changes (Nicosia and Madri, 1987).
membrane protrusions (microspikes) (Fig. 4B), which The peptides released by partial proteolysis of
contain actin filaments, establish adhesions with extracellular matrix macromolecules (matrikines, see
components of the extracellular matrix, sense their above) (Maquart et al., 2004) may have pro- and/or anti-
environment (like antennae) and participate in cell angiogenic activity, promote protease and cytokine
migration. production, and/or facilitate neutrophil migration and
monocyte recruitment (Bellon et al., 2004; Adair-Kirk
12
Angiogenesis and related secondary

and Senior, 2008; Monboisse et al., 2014).


vessel sprout (Zeng et al., 2007). Conversely, in perfused
7.1.4.2. EC proliferation vessels, through CD42 activation, VEGFR2, PECAM1
and VE-cadherin sense the blood flow, and the plane of
Mature ECs, normally in a resting state, have an EC division is determined by the shear-stress direction
extremely slow turnover rate (2 months or more) (Tzima et al 2003, 2005; Gomes et al., 2005).
(Folkman and Cotran, 1976; Chen et al., 1995). Therefore, when an entire endothelial tip cell
Therefore, angiogenesis is generally a quiescent process migrates into the interstitium, it is followed by another
in the healthy adult organism. Nevertheless, ECs can stalk cell, and EC sprouts are formed in the perivascular
quickly convert to a proliferative state during stroma. Endothelial stalk cells align with others to create
angiogenesis in several processes, such as endothelium either a solid sprout or one with an intercellular slit-like
repopulation in organ transplants, repair of large vessel lumina. The proliferation, elongation and rearrangement
defects and thrombi rechannelling (Cavallo et al., 1973; of endothelial stalk cells progressively lengthen the
Folkman, 1984). However, EC proliferation is not sprout (the sprout extension and tubulogenesis being
absolutely essential, since angiogenesis has been shown interlinked processes) (Aydogan et al., 2015). The
to take place even in the absence of EC replication presence of abundant contractile intermediate filaments
(Sholley et al., 1984). Thus, initial sprouts may progress in the endothelium of the sprouts might be important for
without cell division, although proliferation is required the extension and migration of these sprouts (Sauteur et
for sustained sprouting (Ausprunk and Folkman, 1977). al., 2014).
As mentioned above, endothelial DNA synthesis occurs
in parent vessels before sprouting and, according to 7.1.4.3. Pericyte mobilization, proliferation and
some authors, as early as 6 to 8 hours after an angiogenic recruitment in the sprouting phase of angiogenesis
stimulus is applied (Cavallo et al., 1973). EC mitoses
appear in both the parent and newly formed vessels. Pericyte mobilization, proliferation and association
Classically, it was pointed out that mitoses occur at the with EC sprouts occur during the sprouting phase of
tip of the sprouts (Clark and Clark, 1939, Hadfield, angiogenesis (Rhodin and Fujita, 1989; Schlingemann et
1951), but it is now accepted that when capillary sprout al., 1990, 1991, 1996; Díaz-Flores et al., 1991, 1992,
budding begins, endothelial proliferation takes place in 1994a; Nehls et al., 1992; Wesseling et al., 1995;
cells that follow the endothelial tip cells. In other words, Morikawa et al., 2002; Gerhardt and Betsholtz, 2003;
the zone of replication is closer to the parent vessel. McDonald and Choyke, 2003). In addition, pericyte
recruitment and EC investment include alignment,
7.1.4.2.1. Concept of endothelial stalk cells. Cells with contacts and phenotype changes. Although EC sprouts
capacity to proliferate, and to form tubes and branches may initially form without pericyte involvement
are currently named endothelial stalk cells (see above) (representing a plasticity window, allowing ECs to
(Gerhardt and Betsholtz, 2005). Thus, these cells remodel), pericytes are also among the first cells to
proliferate facilitating sprout extension, establish invade newly vascularized tissues (Díaz-Flores et al.,
intercellular junctions, develop basement membrane and 1991; Nehls et al., 1992; Reynolds et al., 2000). Most of
are coated by pericytes (Fig. 5A-C) (Eiken and Adams, the authors are of the opinion that the investment of
2010; Geudens and Gerhardt, 2011; Wacker and sprouts by pericytes occurs around trailing trunk ECs
Gerhardt, 2011). (stalk cells). Therefore, endothelial tube formation is
Stalk cell proliferation depends on VEGF followed by investment of pericytes, which use EC
concentration, while tip cell migration depends on a sprouts as migration clues. In the association of pericytes
VEGF gradient (Ruhrberg et al., 2002; Gerhardt et al., with the endothelium, cytoplasmic processes of pericytes
2003). VEGF induces filopodia formation and the and ECs have been observed penetrating each other (peg
expression of Dll4 in tip cells, and bridge formation with and socket) (Fig. 2D) (Diaz-Flores et al., 2011a).
filopodia of other tip cells during vessel sprout Pericytes are recruited in the vascular sprouts (Fig.
connection (Bentley et al., 2009). The ability of 5A-C) in response to growth factors, such as PDGF and
angiogenic stimuli to induce replication in confluent ECs TGF beta, and angiopoietin 1 and sphingosine 1
is associated with disruption of cell-cell contacts phosphate (Nicosia and Villaschi, 1995; Abramsson et
(Bavisotto et al., 1990). Likewise, the replicative state al., 2002; Hashizume and Ushiki, 2002; Bergers and
and its ability to respond to endogenous mitogens may Song, 2005; Hoffmann et al., 2005). Nitric oxide also
depend on cytoskeletal organization, such as mediates pericyte recruitment (Kashiwagi et al., 2005).
microtubule destabilization or changes in the cell shape This association between pericytes and ECs is key to
(Liaw and Schwartz, 1993). Finally, collagen in the vascular maturation during remodelling and to vascular
interstitium may influence EC proliferation (Madri and patterning, diameter regulation, vessel stabilization and
Stenn, 1982; Schor et al., 1983). endothelial cell survival (Hellström et al., 2001;
The orientation of stalk cell divisions is important Gerhardt and Betsholtz, 2003; Betsholtz et al., 2005;
and is also regulated by VEGF gradient (Zeng et al., Gerhardt and Semb 2008). Although the mere presence
2007). During sprout extension in absence of blood flow, of pericytes appears not to protect vessels from
the plane of division is perpendicular to the axis of the undergoing aggression after inhibition of VGFG
(Morikawa et al., 2002; Inai et al., 2004), pericyte
12
Angiogenesis and related secondary

Fig. 5. Endothelial
stalk cells in
sprouting phase of
angiogenesis.
A, B. Vascular
sprouts (arrows) are
observed in double
stained sections with
anti-CD34 (staining
endothelial stalk cells
- brown) and anti-
αSMA (staining
pericytes - red). Note
in A that sprouts
originate from one
side of the parent
vessels, probably
leading to angiogenic
stimulation.
C. Ultrastructural
image of endothelial
stalk cells (ec)
around a narrow or
virtual lumen (L).
Interendothelial cell
junctions (arrows)
and a mitotic
endothelial stalk cell
are observed. Also
note developing
basement
membrane, a
perivascular cell (p)
and projections of
perivascular cells.
Observe the contacts
between the
perivascular cell and
endothelial stalk
cells. Insert 1: Detail
of the interendothelial
cell junctions
(arrows). Insert 2:
Pericytes (stained in
red with anti-αSMA)
and endothelial stalk
cells (stained in
brown with anti-
CD34) under light
microscopy. Scale
bars: A, 20 µm; B, 30
µm; C, 2 µm.
12
Angiogenesis and related secondary

coverage intervenes favourably in vessel protection


during development (Chan-Ling et al., 2004) and granulation tissue formation, in which there is a loss of
prevents vessel regression (Benjamin et al., 1998, 1999; CD34 expression and a gain of αSMA expression in
Enge et al., 2002). Pericyte-endothelial maturation successive steps, including irregular cell labelling
appears to be more important than simple pericyte intensity for anti-CD34, co-localization of CD34 and
coverage (Hoffmann et al., 2005), and TGF beta 1 actin, and finally loss of CD34 expression and gain of
participates in this process of pericyte differentiation αSMA (Fig. 6C). Moreover, there are simultaneous
from precursor cells (Darland and D’Amore, 2001; changes in ultrastructural morphology of the cells, which
Darland et al., 2003). Recently, it has been demonstrated show characteristics of fibroblasts, protomyofibroblasts
that nerve injury-induced protein, Ninjurin (Ninj1), is a and myofibroblasts, with a progressive increase in
factor to regulate angiogenesis through the function of intracytoplasmic microfilament bundles (first pre-
pericytes (Ninj 1 negatively regulates the formation of stressing with β and γ actin in protomyofibroblasts, and
neovessels) (Matsuki et al., 2015). later with typical αSMA+ stress filaments in
Alternatively, pericytes may locate at the growing myofibroblasts) (Díaz-Flores et al., 2015a,b). Likewise,
front of the endothelial sprouts, participating in there is an increase in organelles involved in protein
angiogenesis by determining the location of sprout synthesis, with remodelling of the extracellular matrix.
formation and by guiding newly formed vessels (Nehls These activated and migrating CD34+ stromal cells
et al., 1992; Tsuzuki and Sasa, 1994; Ozerdem et al., show PDGFR α and β, and secrete vascular endothelial
2001; Morikawa et al., 2002; Ozerdem and Stallcup, growth factors (VEGF), epidermal growth factor (EGF),
2004). Therefore, pericytes could be the first to extend nitric oxide synthase (iNOS), macrophage inflammatory
beyond sprouting ECs, promoting EC survival and protein 1α (MIP-1 α), MIP-2 and cytokines, IL-2, IL-6,
guiding EC migration (Amselgruber et al., 1999; IL-10 and Il-13 (Suciu et al., 2010; Chen et al., 2013;
Morikawa et al., 2002; Darland et al., 2003; Kale et al., Zheng et al., 2014b,c; Albulescu et al., 2015).
2005). Moreover, pericytes and macrophages can invade
tissues in the absence of ECs and can form tubes 7.1.4.5. Inflammatory cells in sprouting phase
(Moldovan et al., 2000; Anghelina et al., 2002, 2004;
2006; Ozerdem and Stallcup, 2004), enabling the Diapedesis of leukocytes (neutrophils, macrophages
posterior penetration of ECs. In this case, pericytes and and lymphocytes) precedes and accompanies
macrophages act as a scaffold along which ECs migrate angiogenesis during repair. The transmigration of these
during sprouting. cells through blood vessel walls is not passive, since
One question is the source of pericytes in the newly ECs, pericytes, CD34+ SFCs and migrating cells interact
formed vessels, which may originate from pre-existing by means of a cascade of signalling events, in different
pericytes or other precursor cells, including bone regulatory mechanisms. Thus, neutrophils are a source
marrow-derived precursor cells and CD34+ stromal cells of VEGF-A and Interleukin-8 (Li et al., 2003; Ohki et
(Nakayasu, 1988; Rhodin and Fujita, 1989; Diaz-Flores al., 2005; Schruefer et al., 2006), macrophages for
et al., 1991; Nehls et al., 1992; Rajantie et al., 2004; Interleukin-1, TNF α, PDGF-AB, TGF α and β, FGF β,
Lamagna and Bergers, 2006; Bexell et al., 2009). VEGF, EGF, prostaglandin, reactive oxygen species and
Among other functions attributed to pericytes is a several pro-angiogenic chemokines via regulation of the
role in angiogenesis regulation. The absence of pericytes chemokine receptor CXCR4 by hypoxia (Polverini et al.,
in EC sprouts is believed to stimulate EC mitosis, while 1977; DiPietro and Polverini, 1993; Schioppa et al 2003;
their presence in older regions of the growing capillaries Moldovan and Moldovan, 2005). T-lymphocytes
may inhibit EC proliferation and migration (Ordlidge produce interferon gamma, Interleukins and TNF-α (Du
and D'Amore, 1987; Sato and Rifkin, 1989). et al., 2012; Dunk et al., 2012). Platelets contribute with
PDGF-AB, TGF β, epidermal growth factor (EGF)/
7.1.4.4. Perivascular CD34+ stromal cells in the TNF-α, tromboxane and insulin-like growth factor.
sprouting phase of angiogenesis Moreover, platelet derived phospholipids and
microparticles play an important role as regulators of
Migration, proliferation and differentiation into other angiogenesis (Walsh et al., 2015).
cell types may occur in the activated and dissociated During the initial phase of angiogenesis, the
perivascular CD34+ stromal cells during sprouting migrating monocytes/macrophages may also contribute
angiogenesis. The migration of these cells from their to the dissociation and detachment of the activated
pre-existing vascular niches takes place in a modified pericytes and CD34+ SFCs in parent vessels before
extracellular matrix in which oedematous spaces and a vascular sprouts (Díaz-Flores et al., 2009b, 2011b,
provisional matrix with fibrin and fibronectin are 2014). Indeed, we have described morphologic
present. During migration, these cells show shortening associations between monocytes/macrophages and
of their processes, present mitoses (Fig. 6A) with a high resident cells (pericytes and CD34+ SFCs) during this
proliferative index (demonstrated in double staining with stage. These associations continue when the perivascular
Ki-67 and CD34) (Fig. 6B) (Díaz-Flores et al., 2015a,b) cells are detached from the vessel wall (Fig. 6D), and
and acquire transitional cell forms in several processes. when they acquire transitional cell forms with
An example of transitional cell forms occurs in myofibroblasts, suggesting interactive cooperation in
migration, differentiation and functional activity (like a
12
Angiogenesis and related secondary

Fig. 6. Perivascular CD34+ stromal cell and macrophage behaviour, and tubulogenesis in sprouting phase of angiogenesis. A. One CD34+
perivascular stromal cell in mitosis (arrow) around a microvessel (asterisk). B. Double staining with anti-ki67 and anti-CD34 reveals ki-67 expression in
nuclei of CD34+ perivascular stromal cells (arrows). C. Double staining with anti-CD34 (brown) and anti-αSMA (red) shows stromal cells expressing
either CD34 or αSMA. D. Double staining with anti-CD68 (staining macrophages – red) and anti-CD34 (staining stromal cells – brown) reveal
association between macrophages and stromal cells. E. Ultrastructural image of vascular lumen formation (tubulogenesis) (asterisks) between
endothelial stalk cells (ec). Pericyte: p. Scale bars: A, 10 µm; B, 15 µm; C, 25 µm; D, 20 µm; E, 1 µm.
12
Angiogenesis and related secondary

tug-macrophage towing a ship-perivascular cell-


myofibroblast) (Díaz-Flores et al., 2009b). these initial stages (Nicosia and Madri, 1987).
Progressively, the deposits of fibronectin decrease,
7.1.4.6. Tubulogenesis (vascular lumen formation) becoming discontinuous, while laminin and type IV
collagen increase, accumulating and forming a
In the formation of the lumen during sprouting continuous feltwork in the subendothelial space. In the
angiogenesis, ECs form tubular channels (Fig. 6E) late stages of angiogenesis, increased amounts of types I
capable of carrying blood. This process requires a and III collagen in the perivascular space are observed.
complex mechanism and the participation of numerous In some conditions, greatly thickened and/or
molecules (for review: Iruela-Arispe and Davis, 2009). multilayered basement membrane is present in mature
Current and classical studies, and according to the five vessels, probably by repeated episodes of EC death and
potential morphogenetic processes described for regrowth (Díaz-Flores et al., 1994a). Pericyte and
different lumen and tubule formation (Lubarsky and endothelial cells participate in the formation of mature
Krasnow, 2003), two mechanisms have been considered basement membrane. Thus, pericytes stimulate vascular
in vascular sprout lumenogenesis: cell hollowing or basal membrane formation by specific induction of
intracellular vacuolization and budding or cord fibronectin, nidogen-1, perlecan and laminin isoforms
hollowing. In the cell hollowing or intracellular (Stratman et al., 2009). Likewise, connective tissue
vacuolization, contiguous endothelial stalk cells growth factor (CCN2) is essential for basement
originate exocytotic vacuoles/vesicles, which membrane formation and pericyte adhesion (Hall-Glenn
interconnect and lead to a luminal space (Folkman and et al., 2012).
Haudenschild, 1980; Furusato et al., 1984). In the
budding or cord hollowing mechanism, initial 7.1.4.8. Vascular anastomosis and capillary loop
intercellular channelling occurs by curvature of the formation
trailing trunk ECs (stalk ECs) (Wakui, 1988; Jin et al.,
2005; Axnick and Lammert, 2012; Yu et al., 2015). In Vascular anastomosis occurs as a result of the
general, it is accepted that the lumen of the capillary formation of stable contacts between ECs of two
sprout is formed between the adjacent endothelial angiogenic sprouts and/or between sprouts and
processes, which bud off the wall of the parent vessels functional blood vessels (Fig. 7A). The connection
and conserve their cellular polarity (Wakui, 1988). Thus, between sprouts is initiated by contacting tip cell
the capillary sprout lumen may be an elongation of the filopodia and is followed by the formation of adherent
parent vessel lumen (Wakui et al., 1988). Other authors junctions, in which cDh5/VE-cadherin plays an
are of the opinion that the new vessel lumen appears first important role (Lenard et al., 2013). When the fusion is
in the sprout, merging later with the mother vessel with a functional vessel, tip cell filopodia in angiogenic
(Folkman, 1984). sprout and ECs of the target vessels form adhesive sites
An important finding, which has received little (apical membrane initiation sites) (Betz et al., 2016).
attention in vascular tubulogenesis in vivo, is the origin Macrophages may act as bridging cells between tip cells
of sprouts from the venous side of circulation with for vascular anastomosis down-stream of VEGF-
negative blood pressure (see above). In our view, mediated tip cell induction (Maden et al., 2009).
although ECs have cell-cell interactions in early When vascular anastomosis occurs, capillary loops
lumenogenesis, they are devoid of sufficient support. are formed. Other capillary sprouts then appear from
Therefore, low physical forces facilitate structural these loops to form a plexus. It has been suggested that
preservation of the developing endothelial tubes. When pericytic processes, which seem to bridge the gap
tip cells join to form capillary loops and contact with the between the leading edges of opposing endothelial
arterial side of circulation, the gradual impact of positive sprouts, may serve as guiding structures for loop
blood pressure occurs in the newly formed vessels and formation (Nehls et al., 1992).
contributes to remodelling them (Díaz-Flores et al., Vessels originating from the venous side of the
2009b). circulation form vascular networks, which connect then
to the arterial side and become functional. Subsequently,
7.1.4.7. Changes in extracellular matrix. Formation remodelling of circulation occurs to adapt to tissue needs
of a new basement membrane (see below).

Initially, active ECs secrete matrix-degrading 7.1.4.2. Phase of vascular remodelling and
enzymes, such as plasminogen activator and collagenase, stabilization
causing fragmentation of the basement membrane (see
above). Thus, in the initial stages of the developing We have considered three main phases in
microvessels, fibronectin is a predominant component of angiogenesis for a better understanding of this process,
the provisional matrix, making up a delicate fibrillary although the phenomena in each phase overlap with the
network. Fibrils of type IV and V collagen, patchy next. Thus, most vascular remodelling and stabilization
amorphous deposits of laminin and rare to absent fibrils phenomena initiate in the sprouting phase (see above).
of type I and III collagen have also been described in Therefore, in this section, we address only the most
outstanding phenomena in this final phase of
12
Angiogenesis and related secondary

Fig. 7. Sprouting angiogenesis: vascular anastomosis, remodelling and stabilization. A. Double staining showing CD34+ endothelial cells (brown)
anastomosing to (arrow) microvessels (v) with periendothelial αSMA+ mural cells (red). B, C. Ultrastructural images in which regressing vessels with
EC degenerative phenomena (B) (arrow) and with intravascular accumulations of platelets (C) are observed. D. Orderly arrangement of phalanx
endothelial cells (ec), which show a sessile morphology. Mural cells: mc. Scale bars: A, 15 µm; B-D, 2 µm.
12
Angiogenesis and related secondary

angiogenesis.
(Fig. 7C). We have hypothesized that this accumulation
7.1.4.2.1. Vascular remodelling. During angiogenesis, a of factor-releasing platelets during the massive
dense network of neovessels is generally created and is regression of neocapillaries may convert highly
followed by a process of remodelling, in which many vascularized zones (as occurs in granulation tissue) into
vessels undergo involution and branching remodelling. a ‘paracrine transitional organ’, facilitating fibroblast/
In this mechanism, pruning of excessive blood vessels is myofibroblast proliferation (Díaz-Flores et al., 2009b).
essential. Two different types of pruning have been Subsequently, homogenized platelets, endothelial cell
described, depending on whether vessels are lumenized debris and basement membrane residues are observed in
or not (Kochhan et al., 2013; Franco et al., 2015; Lenard the regions affected by vascular remodelling. This
et al., 2015; Betz et al, 2016). When vessels are massive regression of capillaries originates capillary-free
lumenized, pruning involves cell-self fusion (Lenard et zones and marked reduction in overall vascular density
al., 2015) and formation of a unicellular tube, in which (Benjamin et al., 1998).
the transcellular lumen collapses (vessel constriction and
occlusion), followed by apical membrane retraction and 7.1.4.2.2. Vascular maturation and stabilization.
reduction in cell-cell contacts, with EC apoptosis and/or Maturation and stabilization of persistent vessels
migration. This process is similar to intussusceptive involves maturation regulating molecules, recruitment of
angiogenesis (see below), in which intravascular pillars pericytes (see above), extracellular matrix formation,
may form in vessels originated by sprouting and vessel type (arteries, arterioles, capillaries, venules
angiogenesis (Djonov et al., 2001; Makanya et al., 2005; and veins) and organ-specific differentiation. Maturation
Hlushchuk et al., 2011). In non-lumenized vessel regulating molecules include angiopoietins and PDGFs
pruning, cell rearrangements occur with formation of a (Potente et al., 2011). Pericytes act in vessel stabilization
unicellular bridge, followed by apical compartment by the release of stabilizing factors including
separation, a reduction in cell-cell contacts, EC angiopoietin-1 and TMP3 (Suri et al., 1996).
apoptosis and/or migration and detachment of the vessel. Angiopoietin-1 induces DLL4 expression through TIE2-
In both types of pruning, residual ECs may migrate and receptor. DLL4 activates Notch signalling which down-
return to neighbouring vessels by a reverse sprouting regulates VEGRF2, thereby preventing further sprouting
mechanism (Chen et al., 2012; Udan et al., 2013). and inducing the expression of NRARP, which enhances
Elevated levels of oxygen, low VEGF levels, WNT signalling, increasing tight junction stabilization.
angiopoietin II signals and vessel flow changes play an In addition, pericytes stimulate vascular basal membrane
important role in vascular pruning (Claxton and formation (Stratman et al., 2009) (see above). Likewise,
Fruttiger, 2005; Lange et al., 2009; Hlushchuk et al., pericytes prevent regression by releasing inhibitors of
2011). Indeed, pruning involves EC apoptosis (Fig. 7B) metalloproteinases (e.g. tissue inhibitor of
(high oxygen level—Lange et al., 2009—and metalloproteinase 3). However, there are apparently
macrophage through WNT7b—Lobov et al., 2005— conflicting data in the role of pericytes in vessel
induced apoptosis), endothelial migration-dependent stabilization (von Tell et al., 2006). Although pericyte
regression (apoptosis-independent vascular regression) coating may prevent vessel regression (Benjamin et al.,
(Dimmeler and Zeiher, 2000; Wietecha et al., 2013) and 1998), pericytes have been observed in immature,
‘reverse intussusception’ (intussusceptive vascular mature and involutive microvessels (Díaz-Flores et al.,
pruning) (Hlushchuk et al., 2011) (for review: Ricard 1991; Inai et al., 2004). Moreover, pericyte-like cells
and Simons, 2015). Intraluminal flow changes (flow persist in some involutive vessels presenting EC
level, orientation and periodicity) also regulate cellular apoptosis.
rearrangement during blood vessel pruning and
branching remodelling, in which the axial polarity Concept of phalanx cells
between the Golgi and nucleus of ECs determines
polarized EC migration (Le Noble et al., 2004; Lee et al, During vessel maturation and stabilization, ECs may
2011; Chen et al., 2012; Tirziu et al., 2012; Kochhan et adopt a thickly apposed, orderly cobblestone-like
al., 2013; Udan et al., 2013; García and Larina, 2014; appearance, acquiring a more sessile cell fate (Fig. 7D)
Lenard et al., 2015; Franco et al., 2015; Ricard and and acquiescent phenotype, as well as the capacity to
Simons, 2015) Thus, it has been proposed that EC sense and regulate perfusion, and to inhibit metastasis in
migration and incorporation into high-flow segments tumours (De Bock et al., 2009; Mazzone et al., 2009). In
depend on Notch pathway signalling (Phng et al., 2009) this stage, these cells are known as endothelial phalanx
and low blood flow, resulting in a smaller total vessel cells, since their alignment and characteristics resemble
area and an increase in blood flow. Alternatively, EC those of ancient Greek soldiers forming military
apoptosis and intussusceptive regression are triggered by phalanxes.
low VEGF levels, resulting in lower blood flow and a
smaller total vessel area (Ricard and Simons, 2015; 7.2. Intussusceptive angiogenesis
Lenard et al., 2015; Franco et al., 2015).
Involutive vessels may also show marked 7.2.1. Definition and nomenclature
intravascular accumulation of platelets (platelet thrombi)
Intussusceptive angiogenesis is the process by which
pre-existing vessels split or remodel through the
12
Angiogenesis and related secondary

formation of transluminal tissue pillars, leading to


expansion (intussusceptive microvascular growth), unmodified. Intussusceptive angiogenesis phenomena
arborisation (intussusceptive arborisation) or branching may occur together with sprouting angiogenesis,
remodelling (intussusceptive branching remodelling) of predominantly in the later stages of growth and
the pre-existing vasculature (Caduff et al., 1986; Burri remodelling (Makanya et al., 2005; Macchiarelli et al.,
and Tarek, 1990; Patan et al., 1996, 2001b; Augustin, 2006).
2001; Djonov et al., 2003; Burri et al., 2004; Makanaya Paku et al. (2011) have followed the mechanisms for
et al., 2009; Paku et al., 2011). pillar formation during tumour-induced intussusceptive
Other terms used to describe intussusceptive angiogenesis, observing suction and subsequent
angiogenesis are intussusception/ intussuceptional transport of perivascular collagen bundles into the vessel
angiogenesis (Caduff et al., 1986), non-sprouting lumen. For this mechanism, the authors coined the term
angiogenesis, splitting angiogenesis, inverse sprouting “inverse sprouting”.
angiogenesis (Paku et al., 2011), intraluminal or internal Taking into account the three main forms of
splitting angiogenesis (Zhou et al., 1998; Williams et al., intussusceptive angiogenesis (intussusceptive
2006b) and longitudinal splitting or longitudinal division microvascular growth, intussusceptive arborisation and
angiogenesis (Egginton, 2009). In addition, and intussusceptive branching remodelling), in
depending on the final results of the process, the intussusceptive microvascular growth, expansion of the
aforementioned terms, intussusceptive microvascular microvasculature occurs through the development of
growth, intussusceptive arborisation and intussusceptive numerous pillars, which fuse and split the pre-existing
branching, are also specifically used for the three main vessels (Makanya et al., 2009; Mentzer and Konerding,
forms of intussusceptive angiogenesis. 2014). In intussusceptive arborisation, series of pillars
participate in vascular branch angles (Ackermann et al.,
7.2.2. Incidence 2014). Finally, in intussusceptive branching remodelling,
two forms may optimize the vascularization: branching
Intussusceptive angiogenesis may occur in pattern or branching pruning (see above). Changes in
developing tissues and in several pathologic processes, blood flow and in metabolic needs occur in all types of
including tumours. Thus, intussusceptive angiogenesis intussusceptive angiogenesis (Djonov et al., 2003).
has been described during development of lung, kidney, Experimentally, we demonstrated that perivenous
ovary, retina, bone and skeletal muscle, among other application of PGE2 and glycerol originates intense
tissues and organs (Macchiarelli et al., 2006; Andres and vascularization of the wall of the rat femoral vein from
Djonov, 2010; De Spiegelaere et al., 2010, 2012), and its intimal endothelial cells (Díaz-Flores et al., 1994b;
experimentally in hypoxic and inflammatory processes 2011a; 2017). In these conditions, the intraparietal
(Konerding et al., 2010), as well as in liver cirrhosis microvessels interconnect, encircle and separate
(Van Steenkiste et al., 2010). Several tumours also show components of the vein wall, giving rise to intravenous
intussusceptive angiogenesis, including colon, renal and projections, in which the EC cover is formed from
mammary carcinomas, gliomas and lymphomas (Patan outgrowing ECs and the core by the incarcerated
et al., 1996; Djonov et al., 2001; Nico et al., 2010; components of the vein wall, including pericyte-like
Ribatti and Djonov, 2012). cells (Díaz-Flores et al., 1994b, 2011a, 2017). The
results resemble the pillars/folds formed by
7.2.3. Events in intussusceptive angiogenesis intussusceptive angiogenesis in the vein of the ovarian
pedicle after ovariectomy or tumour implantation (Patan,
The events in intussusceptive angiogenesis mainly 2000, 2001a,b).
refer to pillar formation, in which four successive phases
have been described (Burri and Tarek, 1990; Patan et al., 7.2.4. Mechanisms
1997; Burri and Djonov, 2002; Djonov et al., 2000,
2003; Egginton et al., 2009; Styp-Rekowska et al., 2011; Several molecules and blood flow participate in the
Paku et al., 2011): 1) intraluminal endothelial pillar regulation of intussusceptive angiogenesis. Variable
formation (trans-capillary inter-endothelial bridge) from expression of VEGF and VEGF receptors (VEGFR1 and
endothelial cells of the opposite sites of a vessel wall VEGFR2) may condition sprouting or intussusceptive
(contact between opposite capillary walls) (Fig. 8A), 2) angiogenesis (Makanya et al., 2005; De Spiegelaere et
reorganization of the junctions between the pillar al., 2010; Hlushchuk et al., 2011; Gianni-Barrera et al.,
endothelial cells, which increase in size, flatten and 2013). Hypoxia-inducible factor 2 alpha, angiopoietin 1
adopt a bilayer arrangement with a central virtual core, and 2, FGF (fibroblast growth factor 2), and platelet-
3) central core perforation and invasion by pericyte and derived growth factor beta (PDGF beta) also act in
myofibroblast extensions with extracellular matrix intussusceptive angiogenesis regulation (Patan, 1998;
formation, including collagen fibres (Fig. 8B), and 4) Augustin et al., 2001; Kurz et al., 2003; Makanya et al.,
increased size of the pillar, fusion with other pillars and 2005, 2007; Taylor et al., 2010). Blood flow changes
splitting of the vessel lumen. Throughout this process, (e.g. shear stress and/or cyclic stretch) play an important
vessel permeability and the basement membrane remain role in vascular plexus remodelling and differentiation of
venules and arterioles (Djonov et al., 2002; Le Noble et
12
Angiogenesis and related secondary

Fig. 8. Intussusceptive, mimicry and co-option forms of vascularization. A. In double staining with anti-CD34 (staining ECs: brown) and anti-αSMA
(staining pericytes: red), an endothelial pillar (transcapillary interendothelial bridge) is observed (arrow). B, insert. ultrastructural images of cross-
sectioned pillars (arrows), covered by endothelial cells -ec-, and with central core formation (presence of collagen fibres -co). C. channels containing
red cells and lined with melanotic cells and some melanophages (vascular mimicry) in a melanoma. D. observe connection between endothelial-lined
vasculature (elv) and a channel lined with melanotic cells (clm), and with red cells in its lumen. E. melanotic cells forming cuff around vessels (arrows)
are observed in a melanoma (co-option form of vascularization). Scale bars: A, C-E, 25 µm; B, 1 µm.
12
Angiogenesis and related secondary

al., 2005; Filipovic et al., 2009; Tsuda et al., 2009; Lee


et al., 2010, 2011; Styp-Rekowska et al., 2011). including VEGF, participate in the release and
mobilization of EPCs from their niches (Asahara et al.,
7.3. Postnatal angiogenesis with endothelial 1999). Factors other than the VEGF involved in release
precursor cell participation (postnatal and mobilization of EPCs include SDF-1 (stromal
vasculogenesis by EC precursors – EPCs) derived-1) through CXR-4, FGF, osteopontin, PLGF
(placental growth factor), angiopoietin-1 and
7.3.1. Concept and incidence macrophage colony stimulating-factor (Takahashi et al.,
1999; Moore et al., 2001). Moreover, VEGF and SDF-1
Angiogenesis with participation of endothelial promote MMP9/endothelial nitric oxidase synthase-
precursor cells (EPCs) can be defined as the process by mediated release of membrane-bound kid ligand
which these precursor cells enter the circulation from (transformation to a soluble form).
their niches (bone marrow, vessel wall ECs, cord blood),
incorporate into the pre-existing vasculature, and 7.3.3.2. Recruitment and integration of EPCs within
differentiate into mature ECs, contributing to the newly angiogenic vasculature
formed vasculature.
The contribution of EPCs in angiogenesis is difficult When EPCs reach sites of injury, they participate in
to establish, and the intensity of their participation is the restoration of vascular integrity and angiogenesis.
controversial, mainly in tumours (Kim et al., 2003; Thus, it has been demonstrated that EPCs can integrate
Larrivee et al., 2005). into blood vessels of neovascularized ischemic limbs in
different experimental models (Asahara et al 1999;
7.3.2. Characteristics of EPCs Kalka et al., 2000). HMGB1 (high-mobility group box
1) activates integrin-dependent homing of EPCs
As the name suggests, EPCs are precursor cells (Chavakis et al., 2007), and P-selectin and E-selectin
capable of differentiating into mature ECs and may act also act in the adhesion of EPCs (Vajkoczy et al., 2003).
in vasculogenesis and angiogenesis (pre- and postnatal Using a parabiosis model, EPC location was
angiogenesis, also considered postnatal vasculogenesis). demonstrated at sites of intussusceptive angiogenesis
In successive works, the characteristics of EPCs were (Chamoto et al., 2013).
established mainly by the expression of specific markers
(Asahara et al., 1997; Shi et al., 1998; Gehling et al., 7.3.3.3. Differentiation of EPCs into mature EPCs
2000; Peichev et al., 2000). Moreover, as these cells
mature, they lose some markers and acquire others. When EPCs in blood vessels lose their progenitor
Thus, expression in the cells of CD34+, AC133+ and characteristics, they acquire those of mature ECs,
VEGFR2+ has been considered representative of a including expression of EC markers, such as endothelial
highly proliferative EPC population, whereas CD34+, nitric oxide synthase, von Willebrand factor and VE-
AC133-, VEGFR2+ cells represent more mature cadherin (Lin et al., 2000). VEGF participates in this
circulatory EPCs (Asahara et al., 1997, Shi et al., 1998; differentiation of EPCs (Gehling et al., 2000).
Gehling et al., 2000; Peichev et al., 2000). In more
mature EPCs, other markers may be observed, including 7.4. Vasculogenic mimicry (vascular mimicry)
CD31, CD144 (VE-cadherin), CD62E (E-selectin) and
CD184 (chemokine receptor CXCR-4). 7.4.1. Concept

7.3.3. Events and mechanisms in angiogenesis Vasculogenic mimicry or vascular mimicry is the
with EPC participation generation of perfusable, non-endothelial-lined tube-like
or channel structures, which are generally formed by: a)
The principal events in angiogenesis in which EPCs highly plastic migratory/invasive tumour cells that
participate are as follows: a) EPC release from the bone upregulate endothelial-selective markers, and b)
marrow niche, and incorporation into the blood remodelled PAS+ extracellular matrix on the inner wall
(circulating EPCs), b) recruitment and integration within of the tube-like structures (Maniotis et al., 1999; Paulis
angiogenic vasculature (incorporation into the vascular et al., 2010). The network formed by these structures is
endothelium of EPCs) and c) differentiation into mature connected with or adjacent to blood vessels, transporting
ECs. blood or fluid from connecting or adjacent leaky vessels,
7.3.3.1. Release of EPCs and incorporation into the respectively (Maniotis et al., 1999; Paulis et al., 2010).
blood Therefore, the networks of channels originated by
vascular mimicry may form part of the complex
This phase includes detachment of c-kit positive vasculature in certain tumours, in which all the types of
cells from their niches, movement of EPCs to the angiogenesis described here participate (sprouting and
vascular zone and incorporation in the circulation intussusceptive angiogenesis, postnatal vasculogenesis
(Heissig et al., 2002). In general, several factors, by precursor ECs, vessel co-option and vascular
mimicry) (Döme et al., 2007; Hillen and Griffioen,
2007;
12
Angiogenesis and related secondary

Carmeliet and Jain, 2011). This complex vasculature


explains the difficulty of disrupting tumour growth using important role in vascular mimicry (Hendrix et al., 2001;
anti-angiogenic therapy (Dey et al., 2015), since the loss Hess et al., 2001, 2003, 2006a,b).
of one type of angiogenesis may be compensated by
other types (e.g. resistance to anti-VEGF therapy in 7.4.5. Functionality
glioblastoma multiform - Soda et al., 2013).
Vascular mimicry acts as a complementary perfusion
7.4.2. Incidence pathway of the tumours, facilitates metastasis (Hendrix
et al., 2003) and is important for the evaluation of
Vascular mimicry formed by tumour cells has been clinical tumour outcome (poor clinical outcome) (Cao et
described in several types of aggressive tumours. al., 2013). As mentioned above, perfusion in vascular
Likewise, non-endothelial or tumoral cells may also mimicry may occur through interconnecting channel
participate in vascular mimicry, including fetal cells of structures and endothelial-lined vasculature (Fig. 8D) or
the trophoblastic lineage in the hemochorial maternal through the transportation of fluid from adjacent leaking
vascular spaces (Rai and Cross, 2014) and macrophages vessels.
(Barnett et al., 2016) (see other forms of vascular
mimicry). Most vascular mimicry studies have been 7.4.6. Other forms of vascular mimicry by non-
carried out on highly invasive melanoma (Fig. 8C,D), in tumour cells
vivo and in vitro. In addition to melanoma, vascular
mimicry has been observed in several neoplastic In both tumours and angiogenesis in in vivo models,
processes, such as carcinomas of the lung, breast, ovary, it has been demonstrated that macrophages may form
skin, kidney, prostate, liver and bladder, gliomas (above primitive non-endothelial, functionally connected
all in glioblastomas), and sarcomas, including synovial channels (Barnett et al., 2016). Hypoxia inducible
sarcoma, alveolar rhabdomyosarcoma, osteosarcoma, factors play an important role in the formation of these
mesothelioma and Ewing tumour (Maniotis et al., 1999; channels (Barnett et al., 2016).
Paulis et al., 2010).
7.5. Vessel co-option
7.4.3. Events, and characteristics of tumour cells
and extracellular matrix forming vascular channels 7.5.1. Concept

The most important findings in vascular channels are Vessel co-option is the process by which tumour
those related to tumour cells (Fig. 8C) and extracellular cells grow using pre-existing vessels (tumour cells co-
matrix. Tumour cells involved in vascular mimicry opt host vessels). Therefore, this process is not a true
exhibit high plasticity and down-regulation of the angiogenic mechanism, since the neoplasic cells grow
original phenotype markers, showing some over and along vessels (tumour cells parasitize on the
characteristics of embryonic progenitors and ECs pre-existing vasculature).
(Bittner et al., 2000; Seftor et al., 2002; Zhang et al.,
2007; Paulis et al., 2010). Moreover, these tumour cells 7.5.2. Incidence
have anti-coagulant properties and act in matrix
remodelling, providing a perfusion pathway. Thus, in Tumours in well-vascularized organs, such as the
melanoma, the tumour cells that form vascular channels lung and brain, can be a substrate for this type of growth
show down-regulation of genes related to a melanocytic (Wesseling et al., 1994; Pezzella et al., 1997), mainly
phenotype (e.g. tyrosinase and melan A) (Demou and during initial tumour phases. This process has been
Hendrix, 2008), while expression of endothelium- described in glioblastoma, melanoma, Kaposi sarcoma,
associated genes occurs (e.g. CD34, CD105, neurophilin and in a model of neuroblastoma, non-small cell
1, E-selectin, VE-cadherin and tyrosine-kinase receptor carcinoma, Lewis lung carcinoma and murine ovarian
1). Extracellular matrix in the networks of tubular cancer (Pezzella et al., 1997; Holash et al., 1999; Döme
structures shows PAS positivity and presence of et al., 2002; Kim et al., 2002; Zhang et al., 2003). A
collagen IV and VI, laminin, heparan-sulphate similar mechanism has been suggested for
proteoglycans and other components. myofibroblasts during wound healing (Kilarski et al.,
2009), although this finding may be the expression of
7.4.4. Mechanisms differentiating precursor adventitial cells (Díaz-Flores et
al., 2014). We have observed vessel co-option in
Signalling pathways in vascular mimicry are highly melanoma implanted in mice, particularly following
complex (for revision: Seftor et al., 2012). Briefly, prior radiation of the tumour bed before implantation,
hypoxia participates in tumour cell plasticity, and in human glioblastoma multiforme. In the latter, the
contributing to the phenotype switch of tumour cells growing tumour cells emit numerous processes around
(Mihic-Probst et al., 2012). VE-cadherin and EphA2, the pre-existing vessels, which follow an undulating
P13K (phosphoinositide 3 route. Subsequently, vessel regression is observed. In
– kinase) and FAK (focal adhesion kinase) play an this case, cell projections form what is described as
palisade necrosis (see below).
12
Angiogenesis and related secondary

7.5.3. Events and mechanisms in vessel co-option


they are surrounded by modified SMCs, which adopt
Tumours with vessel co-option show viable tumour pericytic characteristics, c) interconnection between
cells forming cuffs around the co-opted vessels (Fig. vascular sprouts (anastomosing channels) in the vein
8E). In some of these tumours, perivascular cuffs appear wall forming microvessel networks, in which their slit-
between necrotic areas. An important finding is the like or more apparent lumens connect to the vein lumen,
involution of the pre-existing co-opted vessels (Holash d) presence of papillary structures (Fig. 9A), whose
et al., 1999; Kim et al., 2002; Zhang et al., 2003), in endothelial cover is formed from one side of the pairs of
which loss of pericytes and smooth muscle cells occurs migrating ECs in the innermost networks of the vein
with regression of the ECs. The balance of angiopoietin wall, and whose cores are formed from stromal wall
II and VEGF is involved in the regression of the co- components of the original vessel (extracellular matrix,
opted vessels. Angiopoietin II and VEGF expression modified SMCs and/or other microvessels).
facilitate vessel sprouting, while angiopoietin II Taking into account the above, the anastomosing
expression in the absence or marked decrease of VEGF vascular channels that penetrate the vessel wall and
induces vessel regression (Holash et al., 1999; Kim et perivascular structures remain connected to the vessel
al., 2002). These regressive phenomena have been lumen, form networks encircling components of the vein
considered as a host defence mechanism (Hillen and wall or perivascular structures and give rise to
Griffioen, 2007) and may lead to the angiogenic apparently free papillae. Therefore, in the papillae, the
formation of secondary structures (see below). EC cover is formed of ECs from these anastomosing
vascular channels, and the core is formed by the
7.6. Piecemeal angiogenesis incarcerated components of the vein wall and
perivascular structures. These components may vary
7.6.1. Concept depending on the depth of microvessel penetration in the
vessel wall (penetration is scant when papillae with little
Piecemeal angiogenesis is a mechanism by which a stromal support are formed). Proliferating endothelial
variable number of intravascular papillary structures cells may even fold onto themselves, originating tuft-like
form in tumours, pseudo-tumours, reactive processes, structures, or onto thrombotic fibrin components. This
and malformations of the blood and lymphatic vessels, pathogenic mechanism of vascular network and papillae
and in the intravascular incorporation of different tissue formation combines the sprouting and intussusceptive
components, including neoplasic cells. Through this type mode of angiogenesis (Díaz-Flores et al., 1994b; Patan,
of angiogenic mechanism, ECs grow from a pre-existing 2000; Patan et al., 2001a; Burri et al., 2004; Ackermann
vessel, and encircle and split up portions of the wall of et al., 2014) with development of loops in the vein wall
the vessel itself, perivascular tissues and/or fibrin (which may occur on the venous side in vessels of all
components, giving rise to the intravascular papillae sizes).
partially connected to the vessel wall (Díaz-Flores et al.,
2016d, 2017). In these intravascular papillae, ECs form 8. Participation of angiogenesis in formation of
the cover and encircled components the core. secondary structures during postnatal life
This piecemeal angiogenic mechanism has also been
demonstrated experimentally and is of interest to explain Angiogenesis may participate in the formation of
several forms of angiogenic-related intravascular postnatal angiogenesis-related secondary structures,
structures. In this section, we consider the experimental including: a) intravascular structures through piecemeal
studies, while the angiogenic-related structures angiogenesis, such as intravascular papillae in vessel
originated by this process will be outlined in the section tumours and pseudotumours, vascular septa in
“Participation of angiogenesis in formation of secondary hemorrhoidal veins and intravascular projections in
structures during postnatal life”. some tumours, b) arterial intimal thickening, c)
intravascular tumours and pseudotumours, d) vascular
7.6.2. Events and mechanisms in experimental glomeruloid proliferations, and e) pseudo-palisading
piecemeal angiogenesis necrosis. In this review we do not include the
participation of angiogenesis in postnatal formation of
Experimentally, a large number of vessel sprouts and granulation tissue (a provisional tissue during repair) and
intravascular papillae have been demonstrated in the rat mature mesenchymal tissues (connective, bone, cartilage
femoral vein wall after PGE2 and glycerol injection into and adipose), nor the widely studied pericyte and CD34+
the soft tissue surrounding the vein (Díaz-Flores et al., stromal cell plasticity.
1994b, 2011a, 2017). Vascular sprouts emerge from
exuberant EC growth in the intima of the vein, forming 8.1. Angiogenesis in the formation of intravascular
conspicuous microvascular networks in the media layer structures through piecemeal angiogenesis
and papillae with the following sequence: a) activation
of vein ECs, b) migration of intimal ECs through gaps in A piecemeal form of angiogenesis (see above) plays
the discontinuous internal elastic lamina, originating an important role in the formation of several
numerous vascular sprouts in the media layer, where intravascular structures, including intravascular papillae
in vessel tumours and pseudotumours, vascular septa in
12
Angiogenesis and related secondary

Fig. 9. Piecemeal angiogenesis in experimental conditions and in the formation of angiogenesis-related secondary structures (papillae) in vessel
tumours and pseudotumours. A. semithin section showing intravascular papillae (arrows) after PGE2 and glycerol administration around the rat femoral
vein. B. intravascular papillary endothelial hyperplasia. Numerous papillae are observed in a section stained with anti-CD34 (staining ECs: brown) and
anti-αSMA (staining mural cells: red). C. numerous papillae are also observed in a cavernous hemangioma. Note the wall of a vessel with endothelial
(brown) and mural (red) cells (arrow), as well as the papillae in the lumen. D-F. Papillae in a cavernous lymphangioma. Sections stained with
podoplanin show a linear arrangement of papillae, some of which are joined by ECs (F, arrow). Scale bars: A, 25 µm; B, C, 15 µm; D, 35 µm; E, F, 7
µm.
12
Angiogenesis and related secondary

hemorrhoidal veins and intravascular projections of


some tumours (e.g. in minimally invasive follicular compartmentalization of hemorrhoidal veins. In the
carcinoma of thyroid). latter, successive series of secondary papillae may be
arranged in linear fashion, forming several septa with
8.1.1. Intravascular papillae in vessel tumours compartmentalization of the vein lumen (Fig. 10A,B)
and pseudotumours as a piecemeal form of (unpublished observations).
angiogenesis
8.1.3. Piecemeal angiogenesis in vascular
A piecemeal form of angiogenesis that originates invasion/pseudoinvasion.
intravascular papillae occurs in several types of vessel
tumours and pseudotumours, and has been described in In some tumours, angioinvasion/pseudo-
experimental conditions (Díaz-Flores et al., 1994b, angioinvasion may be based on a piecemeal mechanism
2011a, 2016d, 2017). By this procedure, growing ECs of angiogenesis. For example, in minimally invasive
encircle and separate components of the vessel wall, follicular thyroid carcinoma (angioinvasive encapsulated
perivascular tissue and/or fibrin, giving rise to carcinoma), the invasion of small to medium vessels
intravascular structures, partially connected to the within or immediately adjacent to the tumour capsule
vessel wall (Díaz-Flores et al., 1994; 2011a, 2016d, may also be related to the formation of intravascular
2017). structures. Indeed, ECs grow out of the vessel walls and
Examples of vascular pseudotumours, and benign encircle tumour glands following a piecemeal
and malignant vascular tumours with intravascular mechanism (Fig. 10C) (see above). In these conditions,
papillae include intravascular papillary endothelial the glands remain in the core of the newly formed
hyperplasia (IPEH), vascular transformation of the peculiar papillae and therefore isolated from the
sinuses in lymph nodes, papillary intralymphatic circulation. This mechanism may explain how vascular
angioendothelioma (PILA/Dabska tumour), retiform invasion evolves with infrequent metastasis and requires
hemangioendothelioma, hemangiosarcoma and further studies.
lymphangiosarcoma (Clearkin and Enzinger, 1976; Kuo
et al., 1976; Hashimoto et al., 1983; Sanz-Trelles et al., 8.2. Arterial intimal thickening
1997; Bhatia et al, 2006; Neves et al., 2011; Li et al.,
2013; Mota et al., 2013; Kugler et al., 2016; Liu et al., In several forms of intimal thickening (e.g. arterial
2015; Emberger et al., 2009; Kuo et al., 2015; Diaz- segments isolated between ligatures), an intense
Flores et al., 2016d). In IPEH (Diaz-Flores et al., 2016d) intraarterial neovascularization originates from the vasa-
and in other lesions, papillae, which may be numerous vasorum, predominantly in the early phase of intimal
(myriad of papillae), show a cover formed by ECs and a thickening development (Díaz-Flores and Domínguez,
connective or fibrinous core (Fig. 9B-F), as occurs in 1985). We have demonstrated this penetration of
experimental conditions (perivenous administration of microvessels through the arterial wall by injecting
PGE2 and glycerol) (Díaz-Flores et al., 1994b, 2011a, contrast, and subsequent tissue clearance and
2017) (see above). However, there are differences in the observations in HE stained sections (Fig. 11A,B).
number of papillae, as well as variations in papillary Indeed, in occluded arterial segments, the following
size, distribution, arrangement, EC cover and core occurs: a) early capillary ingrowth through the adventitia
components (Fig. 9B-F). The resulting papillae are and tunica media, b) development of numerous
similar to folds and pillars described in the ovarian microvessels in the occluded arterial lumen, originating
pedicle after ovariectomy or tumour implantation (Patan, a granulation-like tissue, c) coalescence of the lumen of
2001a,b), and in florid intussusceptive-like microvessels, forming an axial arterial neolumen, d)
microvascular disangiogenesis of bronchopulmonary involution of most microvessels and persistence of
dysplasia (De Paepe et al., 2017). interstitial cells, especially those with a pericytic aspect,
Therefore, these intravascular projections in vessel which acquire characteristics of intimal cells, and e)
tumours and pseudotumours (papillae for pathologists), presence of intimal thickening and a patent lumen in the
and in intussusceptive angiogenesis (folds and pillars), occluded segment connected to some perforating vessels
present a similar structure, supporting the hypothesis of (Díaz-Flores and Dominguez et al., 1985, Díaz-Flores et
a piecemeal angiogenic mechanism in papillary al., 1990).
formation in the experimental and pathological
conditions, with association of sprouting and 8.3. Formation of intravascular tumours and
intussusceptive types of angiogenesis (Díaz-Flores et al., pseudotumours
2016d, 2017).
Intravascular tumours and pseudotumours may be
8.1.2. Compartmentalization of hemorrhoidal related to angiogenesis. For example, in intravenous
veins pyogenic granuloma and intravascular myopericytoma.
In intravenous pyogenic granuloma, numerous
Closely associated with intravascular papillae is the microvessels with prominent ECs and pericytes form the
intravascular lesion (Fig. 11C,D). In myopericytoma, as
12
Angiogenesis and related secondary

Fig. 10. Piecemeal angiogenesis in compartmentalization of hemorrhoidal veins and in vascular invasion/pseudoinvasion. A, B. images of hemorrhoidal
veins in which successive series of secondary papillae, arranged in a linear fashion (arrow) (A), form several septa (arrows) with compartmentalization
of the vein lumen (B). C. minimally invasive follicular thyroid carcinoma. Detail of an intravascular tumour gland (asterisk), encircled by CD34+
endothelium, as occurs in papillary structures. Note a gland (double asterisk) partially encircled by CD34+ endothelial cells, which originate from the
vessel wall. A and B: double staining with anti-CD34 (brown) and anti-αSMA (red). C: staining with CD34 (brown). Scale bars: A, 15 µm; B, 40 µm; C,
12 µm.
12
Angiogenesis and related secondary

Fig. 11. Angiogenesis


in intimal thickening
formation and in
intravascular tumours.
A, B. in early phases
of intimal thickening
development in
occluded arteries,
microvessel ingrowth
in the arterial lumen
from the vasa vasorum
is demonstrated by
injecting contrast after
tissue clearance (A)
and in an HE stained
section (B). Observe in
B microvessels
crossing the artery wall
(aw), reaching the
intimal thickening (it).
C, D. intravenous
pyogenic granuloma
with numerous
microvessels similar to
those of granulation
tissue. Double staining
with anti-CD34
(staining ECs - brown)
and anti- αSMA
(staining mural
cells/pericytes - red).
Scale bars: A, 0.5 mm;
B, 40 µm; C, 0.1 mm;
D, 35 µm.
12
Angiogenesis and related secondary

occurs with arterial intimal thickening, intravascular


neovascularization plays an important role, and these extravascular tissue; c) incorporation of proliferating
lesions are therefore related (Diaz-Flores et al., 2011c, pericytes, which intermingle with the ECs; d) formation
2012b). This hypothesis is supported by the following: of a multilayered basement membrane between ECs and
a) frequent participation of intimal thickening as a pericytes; e) partial occlusion of the mother vessel lumen
myopericytoma component, b) phenotypic similarities and formation of small channels in the developing
between the myoid cells (myopericytes) of the tumour glomeruloid structures and f) development of
and the neointimal (myointimal) cells of the concomitant microvessels with erythrocytes containing lumens,
intimal thickening, and c) possibility of a common cell originating characteristic glomeruloid bodies. In
origin for both lesions and of an angiogenic pattern in glioblastoma multiforme, we have observed (non-
their initial stages (Díaz-Flores et al., 2011c, 2012b). published observations) that several glomeruloid
microvascular structures may appear along a pre-existing
8.4. Glomeruloid vascular proliferations vessel, adopting an arciform distribution. The presence
of glomeruloid microvascular proliferation indicates an
8.4.1. Definition aggressive angiogenic phenotype in human cancers and
is therefore an unfavourable biological marker (Straume
Glomeruloid vascular proliferations or vascular et al., 2002). However, in some conditions, glomeruloid
glomeruloid bodies are structures that resemble the renal structures have been considered as reactive vascular
glomeruli and are formed by closely packed proliferations of undetermined biological importance
anastomosing capillaries, which show irregular and (Kandemir et al., 2014).
tortuous narrow lumina, prominent ECs and pericytes.
8.5. Angiogenesis in the formation of pseudo-
8.4.2. Incidence palisading necrosis

Vascular glomeruloid bodies have been described in Two important structures in glioblastomas are
vascular tumours (frequently associated with POEMS related to angiogenesis: pseudopalisading necrosis and
syndrome – Forman et al., 2007; Yuri et al., 2008; glomeruloid vascular structures described above.
Gonzalez-Guerra et al., 2009) and malformations of the In glioblastoma, two main types of necrosis can be
skin, in glioblastoma multiforme (Fig. 12A,B) (Rojiani observed: pseudopalisading necrosis and large necrosis.
and Dorovini-Zis, 1996), and in other tumours, including Pseudopalisading necrosis in glioblastoma is formed by
breast, lung, endometrium, prostate and meninge a garland-like arrangement of tumour cells (rim of cells)
tumours (Ohtani 1992; Bläker et al., 1999; Straume et around a central degenerative region (Fig. 12C). The
al., 2002; Goffin et al., 2003; Kandemir et al., 2014), as resulting pseudopalisades can be long and undulating or
well as in experimental conditions (e.g. after small and rosette-like. This pseudopalisading
VPF/VEGF-164 gene delivery) (Sundberg et al., 2001). configuration of astrocytic cells is essentially unique for
glioblastoma. The hypothesis of a mixed population of
8.4.3. Characteristics neoplasic and inflammatory cells to explain the
pseudopalisades is not sustainable, since practically all
In glomeruloid bodies, florid microvascular cells in this location are neoplasic astrocytic cells.
proliferation, with packed anastomosing capillaries, Currently, several authors are of the opinion that
shows prominent ECs, which express CD31 and CD34 pseudopalisades could represent an astrocytic cell
(Fig. 12A). These ECs present weak VEGF reactivity population that rapidly migrates away from a
and a Ki-67 proliferation index above 10% (Kandemir et dysfunctional vasculature (from a hypoxic zone) (Brat et
al., 2014). In addition, the pericyte population, with al., 2004; Rong et al., 2006). In pseudopalisades, we
αSMA expression, is relatively important in vessel walls have observed that neoplasic astrocytic cells emit
of glomeruloid structures (Fig. 12B). numerous processes toward an involutive vessel in the
axis of the degenerative central zone. These processes
8.4.4. Mechanisms and sequence of formation and are positive for anti-gliofibrillar acid protein (Fig. 12D
prognostic importance and E) and therefore form most of the degenerative
central zone, in which debris of CD34+ ECs of the
Experimentally, it has been demonstrated that VPF- involutive vessel are also observed (Fig. 12F). Loss of
VEGF-164 is sufficient for induction of glomeruloid pericytes or vascular smooth muscle cells occurs in these
bodies. Likewise, VPF-VEGF-164 is also necessary for vessels. Normally, astrocyte processes envelop synapses,
maintaining these vascular bodies (Sundberg et al., make contact with nodes of Ranvier, form gap junctions
2001). The sequence of glomeruloid body formation is between distal processes of other astrocytes and
as follows: a) vessel dilation and focal accumulation in establish interactions with blood vessels, where they
the vessel EC layer of plump primitive cells, which show form astrocytic endfeet. Our results (obtained in
EC markers, increased expression of VGFR-2 and lack conjunction with Spreafico MA†, nonpublished
of pericyte markers; b) the cells in the accumulations observations) agree with the concept that
proliferate and extend toward the vessel lumen and the pseudopalisades are formed by a mechanism in which
neoplasic astrocytes grow over and along a vessel, which
12
Angiogenesis and related secondary

Fig. 12. Angiogenesis in glomeruloid vascular proliferations and in pseudopalisading necrosis formation. A, B. Glomeruloid bodies with florid
microvascular proliferation, showing prominent CD34+ ECs (A) and an abundant pericyte population (B). C-F. pseudopalisading necrosis in
glioblastoma formed by a garland-like arrangement of tumour cells (rim of cells) around a central degenerative zone (cz), observed in HE (C), anti-
protein gliofibrillar acid (D, E) and in anti-CD34 stained sections. Note that the central degenerative region is formed by the gliofibrillar acid +
projections of the neoplasic astrocytic cells (D, E), which converge in a central vessel (E, arrow). F. Degenerative phenomena (residual
expression of CD34) (arrow) in a central vessel of the pseudopalisading necrosis. Scale bars: A, 20 µm; B, 15 µm; C-E, 50 µm; F, 40 µm.
12
Angiogenesis and related secondary

shows involutive phenomena. Therefore, our


interpretation of the findings is that mummified or Asahara T. and Isner J.M. (2002). Endothelial progenitor cells for
partially disintegrated processes of the astrocytes form vascular regeneration. J. Hematother. Stem Cell Res. 11, 171-178.
the degenerative central zone around vessels (Fig. Asahara T., Murohara T., Sullivan A., Silver M., van der Zee R., Li T.,
12D,E). This concept may also justify the absence of Witzenbichler B., Schatteman G. and Isner J.M. (1997). Isolation of
these specific pseudopalisades in tumours other than putative progenitor endothelial cells for angiogenesis. Science 275,
glioblastomas. The explanation of the involution of the 964-967.
central vessel may be similar to that in vessel co-option Asahara T., Takahashi T., Masuda H., Kalka C., Chen D., Iwaguro H.,
(see above). Inai Y., Silver M. and Isner J.M. (1999). VEGF contributes to
postnatal neovascularization by mobilizing bone marrow-derived
References endothelial progenitor cells. EMBO J. 18, 3964-3972.
Auerbach R., Gilligan B., Lu L.S. and Wang S.J. (1997). Cell
Abdel-Majid R.M. and Marshall J.S. (2004). Prostaglandin E2 induces interactions in the mouse yolk sac: vasculogenesis and
degranulation-independent production of vascular endothelial growth hematopoiesis. J. Cell Physiol. 173, 202-2055.
factor by human mast cells. J. Immunol. 172, 1227-1236. Augustin H.G. (2001). Tubes, branches, and pillars: the many ways of
Abramsson A., Berlin O., Papayan H., Paulin D., Shani M. and Betsholtz forming a new vasculature. Circ. Res. 89, 645-647.
C. (2002). Analysis of mural cell recruitment to tumor vessels. Ausprunk D.H. and Folkman J. (1977). Migration and proliferation of
Circulation 105, 112-117. endothelial cells in preformed and newly formed blood vessels
Ackermann M., Houdek J.P., Gibney B.C., Ysasi A., Wagner W., Belle during tumor angiogenesis. Microvasc. Res. 14, 53-65.
J., Schittny J.C., Enzmann F., Tsuda A., Mentzer S.J. and Axnick J. and Lammert E. (2012). Vascular lumen formation. Curr. Opin.
Konerding M.A. (2014). Sprouting and intussusceptive angiogenesis Hematol. 19, 192-198.
in postpneumonectomy lung growth: mechanisms of alveolar Aydogan V., Lenard A., Denes A.S., Sauteur L., Belting H.G. and
neovascularization. Angiogenesis 17, 541-551. Affolter M. (2015). Endothelial cell division in angiogenic sprouts of
Adair-Kirk T.L. and Senior R.M. (2008). Fragments of extracellular differing cellular architecture. Biol. Open 4, 1259-1269.
matrix as mediators of inflammation. Int. J. Biochem. Cell Biol. 40, Barnett F.H., Rosenfeld M., Wood M., Kiosses W.B., Usui Y., Marchetti
1101-1110. V., Aguilar E. and Friedlander M. (2016). Macrophages form
Albulescu R., Tanase C., Codrici E., Popescu D.I., Cretoiu S.M. and functional vascular mimicry channels in vivo. Sci. Rep. 6, 36659.
Popescu L.M. (2015). The secretome of myocardial telocytes Bavisotto L.M., Schwartz S.M. and Heimark R.L. (1990). Modulation of
modulates the activity of cardiac stem cells. J. Cell Mol. Med. 19, Ca-dependent intercellular adhesion in bovine aortic and human
1783-1794. umbilical vein endothelial cells by heparin-binding growth factors. J.
Alfranca A., López-Oliva J.M., Genís L., López-Maderuelo D., Mirones Cell. Physiol. 143, 39-51
I., Salvado D., Quesada A.J., Arroyo A.G. and Redondo J.M. (2008). Bellon G., Martiny L. and Robinet A. (2004). Matrix metalloproteinases
PGE2 induces angiogenesis via MT1-MMP-mediated activation of and matrikines in angiogenesis. Crit. Rev. Oncol. Hematol. 49, 203-
the TGFbeta/Alk5 signaling pathway. Blood 112, 1120-1128. 220.
Amselgruber W.M., Schäfer M. and Sinowatz F. (1999). Angiogenesis in Benedito R., Roca C., Sorensen I., Adams S., Gossler A., Fruttiger M.
the bovine corpus luteum: an immunocytochemical and and Adams R.H. (2009). The notch ligands Dll4 and Jagged1 have
ultrastructural study. Anat. Histol. Embryol. 28, 157-166. opposing effects on angiogenesis. Cell 137, 1124-1135.
Andres A.C. and Djonov V. (2010). The mammary gland vasculature Benjamin L.E., Hemo I. and Keshet E. (1998). A plasticity window for
revisited. J. Mammary Gland Biol. Neoplasia 15, 319-328. blood vessel remodelling is defined by pericyte coverage of the
Anghelina M., Schmeisser A., Krishnan P., Moldovan L., Strasser R.H. preformed endothelial network and is regulated by PDGF-B and
and Moldovan N.I. (2002). Migration of monocytes/macrophages in VEGF. Development 125, 1591-1598.
vitro and in vivo is accompanied by MMP12-dependent tunnel Benjamin L.E., Golijanin D., Itin A., Pode D. and Keshet E. (1999).
formation and by neovascularization. Cold Spring Harb Symp. Quant Selective ablation of immature blood vessels in established human
Biol. 67, 209-215. tumors follows vascular endothelial growth factor withdrawal. J. Clin.
Anghelina M., Krishnan P., Moldovan L. and Moldovan N.I. (2004). Invest. 103, 159-165.
Monocytes and macrophages form branched cell columns in Bentley K. and Chakravartula S. (2017). The temporal basis of
matrigel: implications for a role in neovascularization. Stem Cells angiogenesis. Philos. Trans. R Soc. Lond. B Biol. Sci. 372,
Dev. 13, 665-676. 20150522.
Anghelina M., Krishnan P., Moldovan L. and Moldovan N.I. (2006). Bentley K., Gerhardt H. and Bates P.A. (2008). Agent-based simulation
Monocytes/macrophages cooperate with progenitor cells during of notch mediated tip cell selection in angiogenic sprout initialisation.
neovascularization and tissue repair: conversion of cell columns into J. Theor. Biol. 250, 25-36.
fibrovascular bundles. Am. J. Pathol. 168, 529-541. Bentley K., Mariggi G., Gerhardt H. and Bates P.A. (2009). Tipping the
Aoki M., Yasutake M. and Murohara T. (2004). Derivation of functional balance: robustness of tip cell selection, migration and fusion in
endotelial progenitor cells from human umbilical cord blood angiogenesis. PLoS Comput Biol. 5, e1000549.
mononuclear cells isolated by a novel cell filtration device. Stem Bergers G. and Song S. (2005). The role of pericytes in blood-vessel
Cells 22, 994-1002. formation and maintenance. Neuro Oncol. 7, 452-464.
Arroyo A.G. and Iruela-Arispe M.L. (2010). Extracellular matrix, Berisha B., Schams D., Rodler D. and Pfaffl M.W. (2016). Angiogenesis
inflammation, and the angiogenic response. Cardiovasc. Res. 86, in the ovary - The most important regulatory event for follicle and
226-235. corpus luteum development and function in cow - An overview. Anat.
Histol. Embryol. 45, 124-130.
12
Angiogenesis and related secondary

Betsholtz C., Lindblom P. and Gerhardt H. (2005). Role of pericytes in


Cancer 49, 3914-3923.
vascular morphogenesis. EXS 94, 115-125.
Capitão M. and Soares R. (2016). Angiogenesis and inflammation
Betz C., Lenard A., Belting H.G. and Affolter M. (2016). Cell behaviors
crosstalk in diabetic retinopathy. J. Cell Biochem. 117, 2443-2453.
and dynamics during angiogenesis. Development 143, 2249-2260.
Capkin A.A., Demir S., Mentese A., Bulut Ç. and Ayar A. (2017). Can
Bexell D., Gunnarsson S., Tormin A., Darabi A., Gisselsson D., Roybon
signal peptide-CUB-EGFdomain-containing protein (SCUBE) levels
L., Scheding S. and Bengzon J. (2009). Bone marrow multipotent
be a marker of angiogenesis in patients with psoriasis? Arch.
mesenchymal stroma cells act as pericyte-like migratory vehicles in
Dermatol. Res. 309, 203-207.
experimental gliomas. Mol. Ther. 17, 183-190.
Caporali A., Martello A., Miscianinov V., Maselli D., Vono R. and Spinetti
Bhatia A., Nada R., Kumar Y. and Menon P. (2006). Dabska tumor
G. (2017). Contribution of pericyte paracrine regulation of the
(endovascular papillary angioendothelioma) of testis: a case report
endothelium to angiogenesis. Pharmacol. Ther. 171, 56-64.
with brief review of literature. Diagn. Pathol. 1, 12.
Carmeliet P. and Jain R.K. (2011). Molecular mechanisms and clinical
Bittner M., Meltzer P., Chen Y., Jiang Y., Seftor E., Hendrix M.,
applications of angiogenesis. Nature 473, 298-307.
Radmacher M., Simon R., Yakhini Z., Ben-Dor A., Sampas N.,
Cavallo T., Sade R., Folkman J. and Cotran R.S. (1973). Ultrastructural
Dougherty E., Wang E., Marincola F., Gooden C., Lueders J.,
autoradiographic studies of the early vasoproliferative response in
Glatfelter A., Pollock P., Carpten J., Gillanders E., Leja D., Dietrich
tumor angiogenesis. Am. J. Pathol. 70, 345-362.
K., Beaudry C., Berens M., Alberts D. and Sondak V. (2000).
Chamoto K., Gibney B.C., Ackermann M., Lee G.S., Konerding M.A.,
Molecular classification of cutaneous malignant melanoma by gene
Tsuda A. and Mentzer S.J. (2013). Alveolar epithelial dynamics in
expression profiling. Nature 406, 536-540.
postpneumonectomy lung growth. Anat. Rec. (Hoboken). 296, 495-
Blair R.J., Meng H., Marchese M.J., Ren S., Schwartz L.B., Tonnesen
503.
M.G. and Gruber B.L. (1997). Human mast cells stimulate vascular
Chan-Ling T., Page M.P., Gardiner T., Baxter L., Rosinova E. and
tube formation. Tryptase is a novel, potent angiogenic factor. J. Clin.
Hughes S. (2004). Desmin ensheathment ratio as an indicator of
Invest. 99, 2691-2700.
vessel stability: evidence in normal development and in retinopathy
Bläker H., Dragoje S., Laissue J.A. and Otto H.F. (1999). Pericardial
of prematurity. Am. J. Pathol. 165, 1301-1313.
involvement by thymomas. Entirely intrapericardial thymoma and a
Chavakis E., Hain A., Vinci M., Carmona G., Bianchi M.E., Vajkoczy P.,
pericardial metastasis of thymoma with glomeruloid vascular
Zeiher A.M., Chavakis T. and Dimmeler S. (2007). High-mobility
proliferations. Pathol. Oncol. Res. 5, 160-163.
group box 1 activates integrin-dependent homing of endothelial
Boesiger J., Tsai M., Maurer M., Yamaguchi M., Brown L.F., Claffey
progenitor cells. Circ. Res. 100, 204-212.
K.P., Dvorak H.F. and Galli S.J. (1998). Mast cells can secrete
Chen Y.L., Jan K.M., Lin H.S. and Chien S. (1995). Ultrastructural
vascular permeability factor/vascular endothelial cell growth factor
studies on macromolecular permeability in relation to endothelial cell
and exhibit enhanced release after immunoglobulin E-dependent
turnover. Atherosclerosis 118, 89-104.
upregulation of fc epsilon receptor I expression. J. Exp. Med. 188,
Chen P.Y., Qin L., Barnes C., Charisse K., Yi T., Zhang X., Ali R.,
1135-1145.
Medina P.P., Yu J., Slack F.J., Anderson D.G., Kotelianski V., Wang
Brat D.J., Castellano-Sanchez A.A., Hunter S.B., Pecot M., Cohen C.,
F., Tellides G. and Simons M. (2012). FGF regulates TGF-β
Hammond E.H., Devi S.N., Kaur B. and Van Meir E.G. (2004).
signaling and endothelial-to-mesenchymal transition via control of
Pseudopalisades in glioblastoma are hypoxic, express extracellular
let-7 miRNA expression. Cell Rep. 2, 1684-1696.
matrix proteases, and are formed by an actively migrating cell
Chen X., Zheng Y., Manole C.G., Wang X. and Wang Q. (2013).
population. Cancer Res. 64, 920-927.
Telocytes in human oesophagus. J. Cell Mol. Med. 17, 1506-1512.
Burger P.C., Chandler D.B. and Klintworth G.K. (1983). Corneal
Chen X., Man G.C.W., Liu Y., Wu F., Huang J., Li T.C. and Wang C.C.
neovascularization as studied by scanning electron microscopy of
(2017). Physiological and pathological angiogenesis in endometrium
vascular casts. Lab. Invest. 48, 169-180.
at the time of embryo implantation. Am. J. Reprod. Immunol. 78,
Burri P.H. and Djonov V. (2002). Intussusceptive angiogenesis--the
e12693.
alternative to capillary sprouting. Mol. Aspects Med. 23, S1-27.
Chung E.S., Chauhan S.K., Jin Y., Nakao S., Hafezi-Moghadam A., van
Burri P.H. and Tarek M.R. (1990). A novel mechanism of capillary
Rooijen N., Zhang Q., Chen L. and Dana R. (2009). Contribution of
growth in the rat pulmonary microcirculation. Anat. Rec. 228, 35-45.
macrophages to angiogenesis induced by vascular endothelial
Burri P.H., Hlushchuk R. and Djonov V. (2004). Intussusceptive
growth factor receptor-3-specific ligands. Am. J. Pathol. 175, 1984-
angiogenesis: its emergence, its characteristics, and its significance.
1992.
Dev. Dyn. 231, 474-488.
Cimpean A.M., Tamma R., Ruggieri S., Nico B., Toma A. and Ribatti D.
Caduff J.H., Fischer L.C. and Burri P.H. (1986). Scanning electron
(2017). Mast cells in breast cancer angiogenesis. Crit. Rev. Oncol.
microscope study of the developing microvasculature in the
Hematol. 115, 23-26.
postnatal rat lung. Anat. Rec. 216, 154-164.
Camaré C., Pucelle M., Nègre-Salvayre A. and Salvayre R. (2017). Clark R.A. and Clark E.L. (1939). Microscopic observations on the
Angiogenesis in the atherosclerotic plaque. Redox Biol. 12, 18-34. growth of blood capillaries in the living mammal. J. Anat. 64, 251-
Cao Y., Sonveaux P., Liu S., Zhao Y., Mi J., Clary B.M., Li C.Y., Kontos 301.
C.D. and Dewhirst M.W. (2007). Systemic overexpression of Clark R.A., DellaPelle P., Manseau E., Lanigan J.M., Dvorak H.F. and
angiopoietin-2 promotes tumor microvessel regression and inhibits Colvin R.B. (1982a). Blood vessel fibronectin increases in
angiogenesis and tumor growth. Cancer Res. 67, 3835-3844. conjunction with endothelial cell proliferation and capillary ingrowth
Cao Z., Bao M., Miele L., Sarkar F.H., Wang Z. and Zhou Q. (2013). during wound healing. J. Invest. Dermatol. 79, 269-276.
Tumour vasculogenic mimicry is associated with poor prognosis of Clark R.A., Quinn J.H., Winn H.J., Lanigan J.M., Dellepella P. and
human cancer patients: a systemic review and meta-analysis. Eur. Colvin R.B. (1982b). Fibronectin is produced by blood vessels in
J. response to injury. J. Exp. Med. 156, 646-651.
12
Angiogenesis and related secondary

Claxton S. and Fruttiger M. (2004). Periodic Delta-like 4 expression in


Anat. Histopathol. 406, 165-177.
developing retinal arteries. Gene Expr. Patterns 5, 123-127.
Diaz-Flores L., Valladares F., Gutierrez R. and Varela H. (1990). The
Claxton S. and Fruttiger M. (2005). Oxygen modifies artery
role of the pericytes of the adventitial microcirculation in the arterial
differentiation and network morphogenesis in the retinal vasculature.
intimal thickening. Histol. Histopathol. 5, 145-153.
Dev. Dyn. 233, 822-828.
Diaz-Flores L., Gutierrez R., Varela H., Rancel N. and Valladares F.
Clearkin K.P. and Enzinger F.M. (1976). Intravascular papillary
(1991). Microvascular pericytes: a review of their morphological and
endothelial hyperplasia. Arch. Pathol. Lab. Med. 100, 441-444.
functional characteristics. Histol. Histopathol. 6, 269-286.
Corselli M., Chen C.W., Sun B., Yap S., Rubin J.P. and Péault B.
Diaz-Flores L., Gutierrez R. and Varela H. (1992). Behavior of
(2012). The tunica adventitia of human arteries and veins as a
postcapillary venule pericytes during postnatal angiogenesis. J.
source of mesenchymal stem cells. Stem Cells Dev. 21, 1299-1308.
Morphol. 213, 33-45.
Darland D.C. and D'Amore P.A. (2001). TGF beta is required for the
Díaz-Flores L., Gutiérrez R. and Varela H. (1994a). Angiogenesis: an
formation of capillary-like structures in three-dimensional cocultures
update. Histol. Histopathol. 9, 807-843.
of 10T1/2 and endothelial cells. Angiogenesis 4, 11-20.
Diaz-Flores L., Gutierrez R., Valladares F., Varela H. and Perez M.
Darland D.C., Massingham L.J., Smith S.R., Piek E., Saint-Geniez M.
(1994b). Intense vascular sprouting from rat femoral vein induced
and D'Amore P.A. (2003). Pericyte production of cell-associated
bypmstaglandins El and E2. Anat. Rec. 238, 66-76.
VEGF is differentiation-dependent and is associated with endothelial
Díaz-Flores L., Gutiérrez R., Madrid J.F., Varela H., Valladares F.,
survival. Dev. Biol. 264, 275-288.
Acosta E., Martín-Vasallo P. and Díaz-Flores L. Jr (2009a).
Davis G.E., Bayless K.J., Davis M.J. and Meininger G.A. (2000).
Pericytes. Morphofunction, interactions and pathology in a quiescent
Regulation of tissue injury responses by the exposure of matricryptic
and activated mesenchymal cell niche. Histol. Histopathol. 24, 909-
sites within extracellular matrix molecules. Am. J. Pathol. 156, 1489-
969.
1498.
Diaz-Flores L. Jr, Gutierrez R., Madrid J.F., Varela H., Valladares F. and
De Bock K., De Smet F., Leite De Oliveira R., Anthonis K. and Carmeliet
Diaz-Flores L. (2009b). Adult stem cells and repair through
P. (2009). Endothelial oxygen sensors regulate tumor vessel
granulation tissue. Front Biosci (Landmark Ed). 14, 1433-1470.
abnormalization by instructing phalanx endothelial cells. J. Mol.
Díaz-Flores L. Jr, Gutiérrez R., Madrid J.F., Sáez F.J., Valladares F.,
Med. (Berl) 87, 561-569.
Villar J. and Díaz-Flores L. (2011a). Peg-and-socket junctions
De Paepe M.E., Benny M.K., Priolo L., Luks F.I. and Shapiro S. (2017).
between smooth muscle cells and endothelial cells in femoral veins
Florid intussusceptive-like microvascular dysangiogenesis in a
are stimulated to angiogenesis by prostaglandin E₂ and
preterm lung. Pediatr. Dev. Pathol. (in press).
glycerols. Histol. Histopathol. 26, 623-630.
De Smet F., Segura I., De Bock K., Hohensinner P.J. and Carmeliet P.
Díaz-Flores L. Jr, Gutiérrez R., Madrid J.F., Álvarez-Argüelles H.,
(2009). Mechanisms of vessel branching: filopodia on endothelial tip
Valladares F. and Díaz-Flores L. (2011b). Adult stem and transit-
cells lead the way. Arterioscler. Thromb. Vasc. Biol. 29, 639-649.
amplifying cells in repair through granulation tissue: Role of the
De Spiegelaere W., Cornillie P., Casteleyn C., Burvenich C. and Van
pericytic vasculature niche. In: Stem Cell, regenerative Medicine.
den Broeck W. (2010). Detection of hypoxia inducible factors and
Chapter 6. Singh S.R. (ed.). Nova Science Publishers.
angiogenic growth factors during foetal endochondral and
Díaz-Flores L., Gutiérrez R., García M.P., Alvarez-Argüelles H., Díaz-
intramembranous ossification. Anat. Histol. Embryol. 39, 376-384.
Flores L. Jr and Madrid J.F. (2011c). Myopericytoma and arterial
De Spiegelaere W., Casteleyn C., Van den Broeck W., Plendl J.,
intimal thickening: the relationship between myopericytes and
Bahramsoltani M., Simoens P., Djonov V. and Cornillie P. (2012).
myointimal cells. J. Cutan. Pathol. 38, 857-864.
Intussusceptive angiogenesis: a biologically relevant form of
Diaz-Flores L. Jr, Gutierrez R., Madrid J.F., Acosta E., Avila J., Diaz-
angiogenesis. J. Vasc. Res. 49, 390-404.
Flores L. and Martin-Vasallo P. (2012a). Cell sources for cartilage
Dejana E. and Lampugnani M.G. (2014). Differential adhesion drives
repair; contribution of the mesenchymal perivascular niche. Front
angiogenesis. Nat. Cell Biol. 16, 305-306.
Biosci (Schol Ed). 4, 1275-1294.
Dejana E. and Orsenigo F. (2013). Endothelial adherens junctions at a
Díaz-Flores L., Gutiérrez R., García M.P., Díaz-Flores L. Jr, Valladares
glance. J. Cell Sci. 126, 2545-2549.
F. and Madrid J.F. (2012b). Ultrastructure of myopericytoma: a
Dejana E., Orsenigo F. and Lampugnani M.G. (2008). The role of
continuum of transitional phenotypes of myopericytes. Ultrastruct.
adherens junctions and VE-cadherin in the control of vascular
Pathol. 36, 189-194.
permeability. J. Cell Sci. 121, 2115-2122.
Díaz-Flores L., Gutiérrez R., García M.P., Sáez F.J., Díaz-Flores L. Jr,
Dellett M., Brown E.D., Guduric-Fuchs J., O'Connor A., Stitt A.W.,
Valladares F. and Madrid J.F. (2014). CD34+ stromal
Medina R.J. and Simpson D.A. (2017). MicroRNA-containing
cells/fibroblasts/fibrocytes/telocytes as a tissue reserve and a
extracellular vesicles released from endothelial colony-forming cells
principal source of mesenchymal cells. Location, morphology,
modulate angiogenesis during ischaemic retinopathy. J. Cell Mol.
function and role in pathology. Histol. Histopathol. 29, 831-870.
Med. (in press).
Díaz-Flores L., Gutiérrez R., Lizartza K., Goméz M.G., García M.del P.,
Demou Z.N. and Hendrix M.J. (2008). Microgenomics profile the
Sáez F.J., Díaz-Flores L. Jr and Madrid J.F. (2015a). Behavior of in
endogenous angiogenic phenotype in subpopulations of aggressive
situ human native adipose tissue CD34+ stromal/progenitor cells
melanoma. J. Cell Biochem. 105, 562-73.
during different stages of repair. Tissue-resident CD34+ stromal
Dey N., De P. and Brian L.J. (2015). Evading anti-angiogenic therapy:
cells as a source of myofibroblasts. Anat. Rec (Hoboken) 298, 917-
resistance to anti-angiogenic therapy in solid tumors. Am. J. Transl.
930.
Res. 7, 1675-1698.
Díaz-Flores L, Gutiérrez R., García M.P., González M., Sáez F.J.,
Diaz-Flores L. and Dominguez C. (1985). Relation between arterial
Aparicio F., Díaz-Flores L. Jr and Madrid J.F. (2015b). Human
intimal thickening and the vasa-vasorum. Virchows Arch. A Pathol.
resident CD34+ stromal cells/telocytes have progenitor capacity and
12
Angiogenesis and related secondary

are a source of αSMA+ cells during repair. Histol. Histopathol 30,


Pflugers Arch. 457, 963-977.
615-627.
Eilken H.M. and Adams R.H. (2010). Dynamics of endothelial cell
Díaz-Flores L., Gutiérrez R., García-Suárez M.P., González M., Díaz-
behavior in sprouting angiogenesis. Curr. Opin. Cell Biol. 22, 617-
Flores L. and Madrid J.F. (2016a). Telocytes as a source of
625.
progenitor cells in regeneration and repair through granulation
Elshabrawy H.A., Chen Z., Volin M.V., Ravella S., Virupannavar S. and
tissue. Curr. Stem Cell Res. Ther. 11, 395-403.
Shahrara S. (2015). The pathogenic role of angiogenesis in
Díaz-Flores L., Gutiérrez R., Díaz-Flores L. Jr, Goméz M.G., Sáez F.J.
rheumatoid arthritis. Angiogenesis 18, 433-448.
and Madrid J.F. (2016b). Behaviour of telocytes during
Emberger M., Laimer M., Steiner H. and Zelger B. (2009). Retiform
physiopathological activation. Semin. Cell Dev. Biol. 55, 50-61.
hemangioendothelioma: presentation of a case expressing D2-40. J.
Díaz-Flores L., Gutiérrez R., González-Gómez M., Díaz-Flores L. Jr,
Cutan. Pathol. 36, 987-990.
Valladares F., Rancel N., Sáez F.J. and Madrid J.F. (2016c).
Enge M., Bjarnegård M., Gerhardt H., Gustafsson E., Kalén M., Asker
Telocyte behaviour during inflammation, repair and tumour stroma
N., Hammes H.P., Shani M., Fässler R. and Betsholtz C. (2002).
formation. Adv. Exp. Med. Biol. 13, 177-191.
Endothelium-specific platelet-derived growth factor-B ablation
Díaz-Flores L., Gutiérrez R., Madrid J.F., García-Suárez M.P.,
mimics diabetic retinopathy. EMBO J. 21, 4307-4316.
González-Álvarez M.P., Díaz-Flores L. Jr and Sáez F.J. (2016d).
Etienne-Manneville S. (2004). Cdc42--the centre of polarity. J. Cell Sci.
Intravascular Papillary Endothelial Hyperplasia (IPEH). Evidence
117, 1291-1300.
supporting a piecemeal mode of angiogenesis from vein
Fantin A., Lampropoulou A., Gestri G., Raimondi C., Senatore V.,
endothelium, with vein wall neovascularization and papillary
Zachary I. and Ruhrberg C. (2015). NRP1 Regulates CDC42
formation. Histol. Histopathol. 31, 1271-1279.
Activation to promote filopodia formation in endothelial tip cells. Cell
Díaz-Flores L., Gutiérrez R., García M.P., Sáez F.J., Díaz-Flores L. Jr.
Rep. 11, 1577-1590.
and Madrid J.F. (2017). Piecemeal mechanism combining sprouting
Filipovic N., Tsuda A., Lee G.S., Miele L.F., Lin M., Konerding M.A. and
and intussusceptive angiogenesis in intravenous papillary formation
Mentzer S.J. (2009). Computational flow dynamics in a geometric
induced by PGE2 and glycerol. Anat. Rec. (Hoboken) (in press).
model of intussusceptive angiogenesis. Microvasc. Res. 78, 286-
Dimmeler S. and Zeiher A.M. (2000). Endothelial cell apoptosis in
293.
angiogenesis and vessel regression. Circ. Res. 87, 434-439.
Fischer R.S., Gardel M., Ma X., Adelstein R.S. and Waterman C.M.
Ding Y., Song N. and Luo Y. (2012). Role of bone marrow-derived cells
(2009). Local cortical tension by myosin II guides 3D endothelial cell
in angiogenesis: focus on macrophages and pericytes. Cancer
branching. Curr. Biol. 19, 260-265.
Microenviron. 5, 225-236.
Folkman J. (1984). What is the role of endothelial cells in angiogenesis.
DiPietro L.A. and Polverini P.J. (1993). Angiogenic macrophages
Lab. Invest. 51, 601-602.
produce the angiogenic inhibitor thrombospondin 1. Am. J. Pathol.
Folkman J. (1985). Tumor angiogenesis. Adv. Cancer Res. 43, 175-203.
143, 678-684.
Folkman J. (1989). Sucessful treatment of an angiogenic disease. N.
Djonov V., Schmid M., Tschanz S.A. and Burri P.H. (2000).
Engl. J. Med. 320, 1211-1212.
Intussusceptive angiogenesis: its role in embryonic vascular network
Folkman J. (2003). Fundamental concepts of the angiogenic process.
formation. Circ. Res. 86, 286-292.
Curr. Mol. Med. 3, 643-651
Djonov V., Andres A.C. and Ziemiecki A. (2001). Vascular remodelling
Folkman J. and Cotran R. (1976). Relation of vascular proliferation to
during the normal and malignant life cycle of the mammary gland.
tumor growth. Int. Rev. Exp. Pathol. 16, 207-248.
Microsc. Res. Tech. 52, 182-189.
Folkman J. and Haudenschild C.C. (1980). Angiogenesis in vitro. Nature
Djonov V.G., Kurz H. and Burri P.H. (2002). Optimality in the developing
288, 551-556.
vascular system: branching remodeling by means of intussusception
Forman S.B., Tyler W.B., Ferringer T.C. and Elston D.M. (2007).
as an efficient adaptation mechanism. Dev. Dyn. 224, 391-402.
Glomeruloid hemangiomas without POEMS syndrome: series of
Djonov V., Baum O. and Burri P.H. (2003). Vascular remodeling by
three cases. J. Cutan. Pathol. 34, 956-957.
intussusceptive angiogenesis. Cell Tissue Res. 314, 107-117.
Franco C.A., Jones M.L., Bernabeu M.O., Geudens I., Mathivet T., Rosa
Döme B., Paku S., Somlai B and Tímár J. (2002). Vascularization of
A., Lopes F.M., Lima A.P., Ragab A., Collins R.T., Phng L.K.,
cutaneous melanoma involves vessel co-option and has clinical
Coveney P.V. and Gerhardt H. (2015). Dynamic endothelial cell
significance. J. Pathol. 197, 355-362.
rearrangements drive developmental vessel regression. PLoS Biol.
Döme B., Hendrix M.J., Paku S., Tóvári J. and Tímár J. (2007).
13, e1002125.
Alternative vascularization mechanisms in cancer: Pathology and
Furusato M., Fukunaga M, Kikuchi Y., Yokota S., Joh K., Aizawa S. and
therapeutic implications. Am. J. Pathol. 170; 1-15.
lshikawa E. (1984). Two- and three dimensional ultrastructural
Du J.W., Xu K.Y., Fang L.Y. and Qi X.L. (2012). Interleukin-17,
observation of angiogenesis in juvenile hemangioma. Virchows
produced by lymphocytes, promotes tumor growth and angiogenesis
Arch. (Cell Pathol.). 46, 229-237.
in a mouse model of breast cancer. Mol. Med. Rep. 6, 1099-1102.
Gálvez B.G., Genís L., Matías-Román S., Oblander S.A., Tryggvason
Dunk C., Smith S., Hazan A., Whittle W. and Jones R.L. (2008).
K., Apte S.S. and Arroyo A.G. (2005). Membrane type 1-matrix
Promotion of angiogenesis by human endometrial lymphocytes.
metalloproteinase is regulated by chemokines monocyte-
Immunol. Invest. 37, 583-610.
chemoattractant protein-1/ccl2 and interleukin-8/CXCL8 in
Dvorak H.F., Hawey V.S., Estrella P., Brown L.F., McDonach J. and
endothelial cells during angiogenesis. J. Biol. Chem. 280, 1292-
Dvorak A.M. (1987). Fibrin containing gels induce angiogenesis.
1298.
lmplications for tumor stroma generation and wound healing. Lab.
Garbett P.K. and Gibbins J.R. (1987). Experimental neovascularization
Invest. 57, 673-686.
in vivo: lhe early changes in a stable adult vasculature responding
Egginton S. (2009). Invited review: activity-induced angiogenesis.
toangiogenic stimulation by a syngeneic neoplasm. Br. J. Exp.
12
Angiogenesis and related secondary

Pathol. 68, 625-635.


Hashimoto H., Daimaru Y. and Enjoji M. (1983). Intravascular papillary
Garcia M.D. and Larina I.V. (2014). Vascular development and
endothelial hyperplasia. A clinicopathologic study of 91 cases. Am.
hemodynamic force in the mouse yolk sac. Front. Physiol. 5, 308.
J. Dermatopathol. 5, 539-546.
Gehling U.M., Ergün S., Schumacher U., Wagener C., Pantel K., Otte
Hashizume H. and Ushiki T. (2002). Three-dimensional cytoarchitecture
M., Schuch G., Schafhausen P., Mende T., Kilic N., Kluge K.,
of angiogenic blood vessels in a gelatin sheet implanted in the rat
Schäfer B., Hossfeld D.K. and Fiedler W. (2000). In vitro
skeletal muscular layers. Arch. Histol. Cytol. 65, 347-357.
differentiation of endothelial cells from AC133-positive progenitor
Heidenreich R., Röcken M. and Ghoreschi K. (2009). Angiogenesis
cells. Blood 95, 3106-3112.
drives psoriasis pathogenesis. Int. J. Exp. Pathol. 90, 232-248.
Gerhardt H. and Betsholtz C. (2003). Endothelial-pericyte interactions in
Heissig B., Hattori K., Dias S., Friedrich M., Ferris B., Hackett N.R.,
angiogenesis. Cell Tissue Res. 314, 15-23.
Crystal R.G., Besmer P., Lyden D., Moore M.A., Werb Z. and Rafii
Gerhardt H. and Betsholtz C. (2005). How do endothelial cells
S. (2002). Recruitment of stem and progenitor cells from the bone
orientate? EXS 94, 3-15.
marrow niche requires MMP-9 mediated release of kit-ligand. Cell
Gerhardt H. and Semb H. (2008). Pericytes: gatekeepers in tumour cell
109, 25-37.
metastasis? J. Mol. Med. (Berl). 86, 135-144.
Hellström M., Gerhardt H., Kalén M., Li X., Eriksson U., Wolburg H. and
Gerhardt H., Golding M., Fruttiger M., Ruhrberg C., Lundkvist A.,
Betsholtz C. (2001). Lack of pericytes leads to endothelial
Abramsson A., Jeltsch M., Mitchell C., Alitalo K., Shima D. and
hyperplasia and abnormal vascular morphogenesis. J. Cell Biol.
Betsholtz C. (2003). VEGF guides angiogenic sprouting utilizing
153, 543-553.
endothelial tip cell filopodia. J. Cell Biol. 161, 1163-1177.
Hellström M., Phng L.K. and Gerhardt H. (2007a). VEGF and Notch
Geudens I. and Gerhardt H. (2011). Coordinating cell behaviour during
signaling: the yin and yang of angiogenic sprouting. Cell Adh. Migr.
blood vessel formation. Development 138, 4569-4583.
1, 133-136.
Ghajar C.M., George S.C. and Putnam A.J. (2008). Matrix
Hellström M., Phng L.K., Hofmann J.J., Wallgard E., Coultas L.,
metalloproteinase control of capillary morphogenesis. Crit. Rev.
Lindblom P., Alva J., Nilsson A.K., Karlsson L., Gaiano N., Yoon K.,
Eukaryot. Gene Expr. 18, 251-278.
Rossant J., Iruela-Arispe M.L., Kalén M., Gerhardt H. and Betsholtz
Gianni-Barrera R., Trani M., Fontanellaz C., Heberer M., Djonov V.,
C. (2007b). Dll4 signalling through Notch1 regulates formation of tip
Hlushchuk R. and Banfi A. (2013). VEGF over-expression in skeletal
cells during angiogenesis. Nature 445, 776-780.
muscle induces angiogenesis by intussusception rather than
Hendrix M.J., Seftor E.A., Meltzer P.S., Gardner L.M., Hess A.R.,
sprouting. Angiogenesis 16, 123-136.
Kirschmann D.A., Schatteman G.C. and Seftor R.E. (2001).
Goffin J.R., Straume O., Chappuis P.O., Brunet J.S., Bégin L.R., Hamel
Expression and functional significance of VE-cadherin in aggressive
N., Wong N., Akslen L.A. and Foulkes W.D. (2003). Glomeruloid
human melanoma cells: role in vasculogenic mimicry. Proc. Natl.
microvascular proliferation is associated with p53 expression,
Acad. Sci. USA 98, 8018-8023.
germline BRCA1 mutations and an adverse outcome following
Hendrix M.J., Seftor E.A., Hess A.R. and Seftor R.E. (2003).
breast cancer. Br. J. Cancer 89, 1031-1034.
Vasculogenic mimicry and tumour-cell plasticity: lessons from
Gomes E.R., Jani S. and Gundersen G.G. (2005). Nuclear movement
melanoma. Nat. Rev. Cancer 3, 411-421.
regulated by Cdc42, MRCK,myosin, and actin flow establishes
Hess A.R. and Hendrix M.J. (2006). Focal adhesion kinase signaling
MTOC polarization in migrating cells. Cell 121, 451-463.
and the aggressive melanoma phenotype. Cell Cycle. 5, 478-480.
González-Guerra E., Haro M.R., Fariña M.C., Martín L., Manzarbeitia L.
Hess A.R., Seftor E.A., Gardner L.M., Carles-Kinch K., Schneider G.B.,
and Requena L. (2009). Glomeruloid haemangioma is not always
Seftor R.E., Kinch M.S. and Hendrix M.J. (2001). Molecular
associated with POEMS syndrome. Clin. Exp. Dermatol. 34, 800-
regulation of tumor cell vasculogenic mimicry by tyrosine
803.
phosphorylation: role of epithelial cell kinase (Eck/EphA2). Cancer
Grant M.B., May W.S., Caballero S., Brown G.A., Guthrie S.M., Mames
Res. 61, 3250-3255.
R.N., Byrne B.J., Vaught T., Spoerri P.E., Peck A.B. and Scott E.W.
Hess A.R., Seftor E.A., Seftor R.E. and Hendrix M.J. (2003).
(2002). Adult hematopoietic stem cells provide functional
Phosphoinositide 3-kinase regulates membrane Type 1-matrix
hemangioblast activity during retinal neovascularization. Nat. Med.
metalloproteinase (MMP) and MMP-2 activity during melanoma cell
8, 607-612.
vasculogenic mimicry. Cancer Res. 63, 4757-4762.
Greenberg J.I., Shields D.J., Barillas S.G., Acevedo L.M., Murphy E.,
Hess A.R., Seftor E.A., Gruman L.M., Kinch M.S., Seftor R.E. and
Huang J., Scheppke L., Stockmann C., Johnson R.S., Angle N. and
Hendrix M.J. (2006). VE-cadherin regulates EphA2 in aggressive
Cheresh D.A. (2008). A role for VEGF as a negative regulator of
melanoma cells through a novel signaling pathway: implications for
pericyte function and vessel maturation. Nature 456, 809-813.
vasculogenic mimicry. Cancer Biol. Ther. 5, 228-233.
Hadfield G. (1951). Granulation tissue. Ann. Roy. Coll. Surg. Eng.
9,397-407. Hillen F. and Griffioen A.W. (2007). Tumour vascularization: sprouting
Hall-Glenn F., De Young R.A., Huang B.L., van Handel B., Hofmann angiogenesis and beyond. Cancer Metastasis Rev. 26, 489-502.
J.J., Chen T.T., Choi A., Ong J.R., Benya P.D., Mikkola H., Iruela- Hiraoka N., Allen E., Apel I.J., Gyetko M.R. and Weiss S.J. (1998).
Arispe M.L. and Lyons K.M. (2012). CCN2/connective tissue growth Matrix metalloproteinases regulate neovascularization by acting as
factor is essential for pericyte adhesion and endothelial basement pericellular fibrinolysins. Cell 95, 365-377.
membrane formation during angiogenesis. PLoS One 7, e30562. His W. (1868). Untersuchungen uber die erste Anlage
Hartlapp I., Abe R., Saeed R.W., Peng T., Voelter W., Bucala R. and desWirbelthierleibes. F.C.W. Vogel. Leipzig.
Metz C.N. (2001). Fibrocytes induce an angiogenic phenotype in Hlushchuk R., Ehrbar M., Reichmuth P., Heinimann N., Styp-Rekowska
cultured endothelial cells and promote angiogenesis in vivo. FASEB B., Escher R., Baum O., Lienemann P., Makanya A., Keshet E. and
J. 15, 2215-2224. Djonov V. (2011). Decrease in VEGF expression induces
intussusceptive vascular pruning. Arterioscler. Thromb. Vasc. Biol.
12
Angiogenesis and related secondary

31, 2836-2844.
D.H., Lee E.S., Kim H.K., Ryu K.W. and Bae J.M. (2003). Circulating
Hoffmann J., Feng Y., vom Hagen F., Hillenbrand A., Lin J., Erber R.,
numbers of endothelial progenitor cells in patients with gastric and
Vajkoczy P., Gourzoulidou E., Waldmann H., Giannis A., Wolburg
breast cancer. Cancer Lett. 198, 83-88.
H., Shani M., Jaeger V., Weich H.A., Preissner K.T., Hoffmann S.,
Kim M., Lee C., Payne R., Yue B.Y., Chang J.H. and Ying H. (2015).
Deutsch U. and Hammes H.P. (2005). Endothelial survival factors
Angiogenesis in glaucoma filtration surgery and neovascular
and spatial completion, but not pericyte coverage of retinal
glaucoma: A review. Surv. Ophthalmol. 60, 524-535.
capillaries determine vessel plasticity. FASEB J. 19, 2035-2036.
Kim S.O., Trau H.A. and Duffy D.M. (2017). Vascular endothelial growth
Holash J., Maisonpierre P.C., Compton D., Boland P., Alexander C.R.,
factors C and D may promote angiogenesis in the primate ovulatory
Zagzag D., Yancopoulos G.D. and Wiegand S.J. (1999). Vessel
follicle. Biol. Reprod. 96, 389-400.
cooption, regression, and growth in tumors mediated by
Kochhan E., Lenard A., Ellertsdottir E., Herwig L., Affolter M., Belting
angiopoietins and VEGF. Science 284, 1994-1998.
H.G. and Siekmann A.F. (2013). Blood flow changes coincide with
Inai T., Mancuso M., Hashizume H., Baffert F., Haskell A., Baluk P., Hu-
cellular rearrangements during blood vessel pruning in zebrafish
Lowe D.D., Shalinsky D.R., Thurston G., Yancopoulos G.D. and
embryos. PLoS One 8, e75060.
McDonald D.M. (2004). Inhibition of vascular endothelial growth
Konerding M.A., Turhan A., Ravnic D.J., Lin M., Fuchs C., Secomb
factor (VEGF) signaling in cancer causes loss of endothelial
T..W, Tsuda A and Mentzer SJ. (2010) Inflammation-induced
fenestrations, regression of tumor vessels, and appearance of
intussusceptive angiogenesis in murine colitis. Anat. Rec.
basement membrane ghosts. Am. J. Pathol. 165, 35-52.
(Hoboken). 293, 849-857.
Iruela-Arispe M.L. and Davis G.E. (2009). Cellular and molecular
Kreuger J. and Phillipson M. (2016). Targeting vascular and leukocyte
mechanisms of vascular lumen formation. Dev. Cell 16, 222-231.
communication in angiogenesis, inflammation and fibrosis. Nat. Rev.
Jakobsson L., Franco C.A., Bentley K., Collins R.T., Ponsioen B.,
Drug Discov. 15, 125-142.
Aspalter I.M., Rosewell I., Busse M., Thurston G., Medvinsky A.,
Krishna Priya S., Nagare R.P., Sneha V.S., Sidhanth C., Bindhya S.,
Schulte-Merker S. and Gerhardt H. (2010). Endothelial cells
Manasa P. and Ganesan T.S. (2016). Tumour angiogenesis-Origin
dynamically compete for the tip cell position during angiogenic
of blood vessels. Int. J. Cancer 139, 729-735.
sprouting. Nat. Cell Biol. 12, 943-953.
Kugler A, Koelblinger P, Zelger B, Ahlgrimm-Siess V. and Laimer M.
Jin S.W. and Patterson C. (2009). The opening act: vasculogenesis and
(2016). Papillary intralymphatic angioendothelioma (PILA), also
the origins of circulation. Arterioscler. Thromb. Vasc. Biol. 29, 623-
referred to as Dabska tumour, in an 83-year-old woman. J. Eur.
629.
Acad. Dermatol. Venereol. 30, e59-e61.
Jin S.W., Beis D., Mitchell T., Chen J.N. and Stainier D.Y. (2005)
Kuo T., Sayers C.P. and Rosai J. (1976). Masson's "vegetant
Cellular and molecular analyses of vascular tube and lumen
intravascular hemangioendothelioma:" a lesion often mistaken for
formation in zebrafish. Development 132, 5199-5209.
angiosarcoma: study of seventeen cases located in the skin and soft
Kale S., Hanai J., Chan B., Karihaloo A., Grotendorst G., Cantley L. and
tissues. Cancer 38, 1227-1236.
Sukhatme V.P. (2005). Microarray analysis of in vitro pericyte
Kuo C.L, Chen P.C, Li W.Y. and Chu P.Y. (2015). Retiform
differentiation reveals an angiogenic program of gene expression.
hemangioendothelioma of the neck. J. Pathol. Transl. Med. 49, 171-
FASEB J. 19, 270-271.
173.
Kalka C., Masuda H., Takahashi T., Kalka-Moll W.M., Silver M.,
Kurz H., Gärtner T., Eggli P.S. and Christ B. (1996). First blood vessels
Kearney M., Li T., Isner J.M. and Asahara T. (2000). Transplantation
in the avian neural tube are formed by a combination of dorsal
of ex vivo expanded endothelial progenitor cells for therapeutic
angioblast immigration and ventral sprouting of endothelial cells.
neovascularization. Proc. Natl. Acad. Sci. USA 97, 3422-3427.
Dev. Biol. 173, 133-147.
Kanbe N., Kurosawa M., Nagata H., Yamashita T., Kurimoto F. and
Kurz H., Burri P.H. and Djonov V.G. (2003). Angiogenesis and vascular
Miyachi Y. (2000). Production of fibrogenic cytokines by cord blood-
remodeling by intussusception: from form to function. News Physiol.
derived cultured human mast cells. J. Allergy Clin. Immunol. 106,
Sci. 18, 65-70.
S85-S90.
Lamagna C. and Bergers G. (2006). The bone marrow constitutes a
Kandemir N.O., Narli Z.I., Kalayci M. and Ozdamar S.O. (2014). A rare
reservoir of pericyte progenitors. J. Leukoc. Biol. 80, 677-681.
pattern of angiogenesis in meningiomas: glomeruloid microvascular
Lange C., Ehlken C., Stahl A., Martin G., Hansen L. and Agostini H.T.
proliferation. Turk. Neurosurg. 24, 765-769.
(2009). Kinetics of retinal vaso-obliteration and neovascularisation in
Kashiwagi S., Izumi Y., Gohongi T., Demou Z.N., Xu L., Huang P.L.,
the oxygen-induced retinopathy (OIR) mouse model. Graefes Arch.
Buerk D.G., Munn L.L., Jain R.K. and Fukumura D. (2005). NO
Clin. Exp. Ophthalmol. 247, 1205-1211.
mediates mural cell recruitment and vessel morphogenesis in
Langlois S., Gingras D. and Béliveau R. (2004). Membrane type 1-
murine melanomas and tissue-engineered blood vessels. J. Clin.
matrix metalloproteinase (MT1-MMP) cooperates with sphingosine
Invest. 115, 1816-1827.
1-phosphate to induce endothelial cell migration and morphogenic
Kilarski W.W., Samolov B., Petersson L., Kvanta A. and Gerwins P.
differentiation. Blood 103, 3020-3028.
(2009). Biomechanical regulation of blood vessel growth during
Larrivée B., Niessen K., Pollet I., Corbel S.Y., Long M., Rossi F.M.,
tissue vascularization. Nat. Med. 15, 657-664.
Olive P.L. and Karsan A. (2005). Minimal contribution of marrow-
Kim E.S., Serur A., Huang J., Manley C.A., McCrudden K.W., Frischer
derived endothelial precursors to tumor vasculature. J Immunol.
J.S., Soffer S.Z., Ring L., New T., Zabski S., Rudge J.S., Holash J.,
175, 2890-2899.
Yancopoulos G.D., Kandel J.J. and Yamashiro D.J. (2002). Potent
le Noble F., Moyon D., Pardanaud L., Yuan L., Djonov V., Matthijsen R.,
VEGF blockade causes regression of coopted vessels in a model of
Bréant C., Fleury V. and Eichmann A. (2004). Flow regulates
neuroblastoma. Proc. Natl. Acad. Sci. USA 99, 11399-11404.
arterial-venous differentiation in the chick embryo yolk sac.
Kim H.K., Song K.S., Kim H.O., Chung J.H., Lee K.R., Lee Y.J., Lee
Development 131, 361-375.
12
Angiogenesis and related secondary

le Noble F., Fleury V., Pries A., Corvol P., Eichmann A. and Reneman
Lu X., Le Noble F., Yuan L., Jiang Q., De Lafarge B., Sugiyama D.,
R.S. (2005). Control of arterial branching morphogenesis in
Bréant C., Claes F., De Smet F., Thomas J.L., Autiero M., Carmeliet
embryogenesis: go with the flow. Cardiovasc. Res. 65, 619-628.
P., Tessier-Lavigne M. and Eichmann A. (2004). The netrin receptor
Leavesley D.I., Schwartz M.A., Rosenfeld M. and Cheresh D.A. (1993).
UNC5B mediates guidance events controlling morphogenesis of the
Integrin beta 1- and beta 3-mediated endothelial cell migration is
vascular system. Nature 432, 179-186.
triggered through distinct signalling mechanisms. J. Cell Biol. 121,
Lubarsky B. and Krasnow M.A. (2003). Tube morphogenesis: making
163-170.
and shaping biological tubes. Cell 112, 19-28.
Lee J.F., Ozaki H., Zhan X., Wang E., Hla T. and Lee M.J. (2006).
Macchiarelli G., Jiang J.Y., Nottola S.A. and Sato E. (2006).
Sphingosine-1-phosphate signaling regulates lamellipodia
Morphological patterns of angiogenesis in ovarian follicle capillary
localization of cortactin complexes in endothelial cells. Histochem.
networks. A scanning electron microscopy study of corrosion cast.
Cell Biol. 126, 297-304.
Microsc. Res. Tech. 69, 459-468.
Lee G.S., Filipovic N., Miele L.F., Lin M., Simpson D.C, Giney B.,
Maden C.H., Fantin A. and Ruhrberg C. (2009). Neuropilin ligands in
Konerding M.A., Tsuda A. and Mentzer S.J. (2010). Blood flow
vascular and neuronal patterning. Biochem. Soc. Trans. 37, 1228-
shapes intravascular pillar geometry in the chick chorioallantoic
1232.
membrane. J. Angiogenes. Res. 2, 11.
Madri J.A. and Stenn K.S. (1982). Aortic endothelial cell migration. l.
Lee G.S., Filipovic N., Lin M., Gibney B.C., Simpson D.C., Konerding
Matrix requirements and composition. Am. J. Pathol. 106, 180-1 86.
M.A., Tsuda A. and Mentzer S.J. (2011). Intravascular pillars and
Madri J.A. and Pratt B.M. (1986). Endothelial cell-matrix interactions: in
pruning in the extraembryonic vessels of chick embryos. Dev. Dyn.
vitro models of angiogenesis. J. Histochem. Cytochem. 34, 85-91.
240, 1335-1343.
Madrid J.F., Diaz-Flores L., Gutierrez R., Varela H., Valladares F.,
Lenard A., Ellertsdottir E., Herwig L., Krudewig A., Sauteur L., Belting
Rancel N. and Rodriguez F. (1998). Participation of angiogenesis
H.G. and Affolter M. (2013). In vivo analysis reveals a highly
from rat femoral veins in the neovascularization of adjacent
stereotypic morphogenetic pathway of vascular anastomosis. Dev.
occluded arteries. Histol. Histopathol 13, 1-11.
Cell 25, 492-506.
Lenard A., Daetwyler S., Betz C., Ellertsdottir E., Belting H.G., Huisken Makanya A.N., Stauffer D., Ribatti D., Burri P.H. and Djonov V. (2005).
Microvascular growth, development, and remodeling in the
J. and Affolter M. (2015). Endothelial cell self-fusion during vascular
embryonic avian kidney: the interplay between sprouting and
pruning. PLoS Biol. 13, e1002126.
Leslie J.D., Ariza-McNaughton L., Bermange A.L., McAdow R., Johnson intussusceptive angiogenic mechanisms. Microsc. Res. Tech. 66,
275-288.
S.L. and Lewis J. (2007). Endothelial signalling by the Notch ligand
Makanya A.N., Hlushchuk R., Baum O., Velinov N., Ochs M. and Djonov
Delta-like 4 restricts angiogenesis. Development 134, 839-844.
V. (2007). Microvascular endowment in the developing chicken
Li Q., Fukuda K., Lu Y., Nakamura Y., Chikama T., Kumagai N. and
embryo lung. Am. J. Physiol. Lung Cell. Mol. Physiol. 292, L1136-
Nishida T. (2003). Enhancement by neutrophils of collagen
L1146.
degradation by corneal fibroblasts. J. Leukoc. Biol. 74, 412-419.
Makanya A.N., Hlushchuk R. and Djonov V.G. (2009). Intussusceptive
Li B., Li Y., Tian X.Y. and Li Z. (2013). Unusual multifocal intraosseous
angiogenesis and its role in vascular morphogenesis, patterning,
papillary intralymphatic angioendothelioma (Dabska tumor) of facial
and remodeling. Angiogenesis 12, 113-123.
bones: a case report and review of literature. Diagn. Pathol. 8, 160.
Makrilia N., Lappa T., Xyla V., Nikolaidis I. and Syrigos K. (2009). The
Liaw L. and Schwartz S.M. (1993). Microtubule disruption
role of angiogenesis in solid tumours: an overview. Eur. J. Intern.
stimulatesDNA synthesis in bovine endothelial cells and potentiates
Med. 20, 663-671.
cellular response to basic fibroblast growth factor. Am. J. Pathol.
Maltby S., Khazaie K. and McNagny K.M. (2009). Mast cells in tumor
143, 937-948.
growth: angiogenesis, tissue remodelling and immune-modulation.
Lin C.S. and Lue T.F. (2013). Defining vascular stem cells. Stem Cells
Biochim. Biophys. Acta 1796, 19-26.
Dev. 22, 1018-1026.
Manetti M., Guiducci S. and Matucci-Cerinic M. (2016). The crowded
Lin Y., Weisdorf D.J., Solovey A. and Hebbel R.P. (2000). Origins of
crossroad to angiogenesis in systemic sclerosis: where is the key to
circulating endothelial cells and endothelial outgrowth from blood. J.
the problem? Arthritis. Res. Ther. 18, 36.
Clin. Invest. 105, 71-77.
Maniotis A.J., Folberg R., Hess A., Seftor E.A., Gardner L.M., Pe'er J.,
Lin C.S., Xin Z.C., Deng C.H., Ning H., Lin G. and Lue T.F. (2010).
Trent J.M., Meltzer P.S. and Hendrix M.J. (1999). Vascular channel
Defining adipose tissue-derived stem cells in tissue and in culture.
formation by human melanoma cells in vivo and in vitro:
Histol. Histopathol. 25, 807-815.
vasculogenic mimicry. Am. J. Pathol. 155, 739-752.
Liu Q., Ouyang R., Chen P. and Zhou R. (2015). A case report of
Maquart F.X., Bellon G., Pasco S. and Monboisse J.C. (2004).
retiform hemangioendothelioma as pleural nodules with literature
Matrikines in the regulation of extracellular matrix degradation.
review. Diagn. Pathol. 10, 194.
Biochimie 87, 353-360.
Lobov I.B., Rao S., Carroll T.J., Vallance J.E., Ito M., Ondr J.K., Kurup
Matsuki M., Kabara M., Saito Y., Shimamura K., Minoshima A.,
S., Glass D.A., Patel M.S., Shu W., Morrisey E.E., McMahon A.P.,
Nishimura M., Aonuma T., Takehara N., Hasebe N. and Kawabe J.
Karsenty G. and Lang R.A. (2005). WNT7b mediates macrophage-
(2015). Ninjurin1 is a novel factor to regulate angiogenesis through
induced programmed cell death in patterning of the vasculature.
the function of pericytes. Circ. J. 79, 1363-1371.
Nature 437, 417-421.
Maumus M., Peyrafitte J.A., D'Angelo R., Fournier-Wirth C., Bouloumié
Lobov I.B., Renard R.A., Papadopoulos N., Gale N.W., Thurston G.,
A., Casteilla L., Sengenès C. and Bourin P. (2011). Native human
Yancopoulos G.D. and Wiegand S.J. (2007). Delta-like ligand 4
adipose stromal cells: localization, morphology and phenotype. Int.
(Dll4) is induced by VEGF as a negative regulator of angiogenic
J. Obes. (Lond) 35, 1141-1153.
sprouting. Proc. Natl. Acad. Sci. USA 104, 3219-3224.
Mazzone M., Dettori D., Leite de Oliveira R., Loges S., Schmidt T.,
12
Angiogenesis and related secondary

Jonckx B., Tian Y.M., Lanahan A.A., Pollard P., Ruiz de Almodovar
(2012). Stalk cell phenotype depends on integration of Notch and
C., De Smet F., Vinckier S., Aragonés J., Debackere K., Luttun A.,
Smad1/5 signaling cascades. Dev. Cell 22, 501-514.
Wyns S., Jordan B., Pisacane A., Gallez B., Lampugnani M.G.,
Mulligan-Kehoe M.J. and Simons M. (2008). Vascular disease in
Dejana E., Simons M, Ratcliffe P., Maxwell P. and Carmeliet P.
scleroderma: angiogenesis and vascular repair. Rheum. Dis. Clin.
(2009). Heterozygous deficiency of PHD2 restores tumor
North Am. 34, 73-79.
oxygenation and inhibits metastasis via endothelial normalization.
Myers K.A., Applegate K.T., Danuser G., Fischer R.S. and Waterman
Cell 136, 839-851.
C.M. (2011). Distinct ECM mechanosensing pathways regulate
McCracken J.S., Burger P.C. and Klintworth G.K. (1979). Morphologic
microtubule dynamics to control endothelial cell branching
observations on experimental comeal vascularization in the rat. Lab.
morphogenesis. J. Cell Biol. 192, 321-334.
Invest. 41, 519-530.
Nagy J.A., Brown L.F., Senger D.R., Lanir N., Van de Water L., Dvorak
McDonald D.M. and Choyke P.L. (2003). Imaging of angiogenesis: from
A.M. and Dvorak H.F. (1989). Pathogenesis of tumor stroma
microscope to clinic. Nat. Med. 9, 713-725.
generation: a critical role for leaky blood vessels and fibrin
Mehta D. and Malik A.B. (2006). Signaling mechanisms regulating
deposition. Biochim. Biophys. Acta 948, 305-326.
endothelial permeability. Physiol. Rev. 86, 279-367.
Nakayasu K. (1988). Origin of pericytes in neovascularization of rat
Mentzer S.J. and Konerding M.A. (2014). Intussusceptive angiogenesis:
cornea. Jpn. J. Ophthalmol. 32, 105-112.
expansion and remodeling of microvascular networks. Angiogenesis
Nehls V., Denzer K. and Drenckhahn D. (1992). Pericyte involvement in
17, 499-509.
capillary sprouting during angiogenesis in situ. Cell Tissue Res. 270,
Mihic-Probst D., Ikenberg K., Tinguely M., Schraml P., Behnke S.,
469-474.
Seifert B., Civenni G., Sommer L., Moch H. and Dummer R. (2012).
Neves R.I., Stevenson J., Hancey M.J., Vangelisti G., Miraliakbari R.,
Tumor cell plasticity and angiogenesis in human melanomas. PLoS
Mackay D. and Clarke L. (2011). Endovascular papillary
One 7, e33571.
angioendothelioma (Dabska tumor): underrecognized malignant
Miranville A., Heeschen C., Sengenès C., Curat C.A., Busse R. and
tumor in childhood. J. Pediatr. Surg. 46, e25-28.
Bouloumié A. (2004). Improvement of postnatal neovascularization
Nico B., Crivellato E., Guidolin D., Annese T., Longo V., Finato N.,
by human adipose tissue-derived stem cells. Circulation 110, 349-
Vacca A. and Ribatti D. (2010). Intussusceptive microvascular
355.
growth in human glioma. Clin. Exp. Med. 10, 93-98.
Moldovan L. and Moldovan N.I. (2005). Role of monocytes and
Nicosia R.F. and Madri J.A. (1987). The microvascular extracellular
macrophages in angiogenesis. EXS 94, 127-146.
matrix. Developmental changes during angiogenesis in the aortic
Moldovan N.I., Goldschmidt-Clermont P.J., Parker-Thornburg J.,
ring-plasma clot model. Am. J. Pathol. 128, 78-90.
Shapiro S.D. and Kolattukudy P.E. (2000). Contribution of
Nicosia R.F. and Villaschi S. (1995). Rat aortic smooth muscle cells
monocytes/macrophages to compensatory neovascularization: the
become pericytes during angiogenesis in vitro. Lab. Invest. 73, 658-
drilling of metalloelastase-positive tunnels in ischemic myocardium.
666.
Circ Res. 87, 378-384.
Ohki Y., Heissig B., Sato Y., Akiyama H., Zhu Z., Hicklin D.J., Shimada
Monboisse J.C., Oudart J.B., Ramont L., Brassart-Pasco S. and
K., Ogawa H., Daida H., Hattori K. and Ohsaka A. (2005).
Maquart F.X. (2014). Matrikines from basement membrane
Granulocyte colony-stimulating factor promotes neovascularization
collagens: a new anti-cancer strategy. Biochim. Biophys. Acta 1840,
by releasing vascular endothelial growth factor from neutrophils.
2589-2598.
FASEB J. 19, 2005-2007.
Moore M.A., Hattori K., Heissig B., Shieh J.H., Dias S., Crystal R.G. and
Ohtani H. (1992). Glomeruloid structures as vascular reaction in human
Rafii S. (2001). Mobilization of endothelial and hematopoietic stem
gastrointestinal carcinoma. Jpn. J. Cancer Res. 83, 1334-1340.
and progenitor cells by adenovector-mediated elevation of serum
Okada H., Tsuzuki T., Shindoh H., Nishigaki A., Yasuda K. and Kanzaki
levels of SDF-1, VEGF, and angiopoietin-1. Ann. NY Acad. Sci. 938,
H. (2014). Regulation of decidualization and angiogenesis in the
36-45.
human endometrium: mini review. J. Obstet. Gynaecol. Res. 40,
Moreau V., Tatin F., Varon C., Anies G., Savona-Baron C. and Génot E.
1180-1187.
(2006). Cdc42-driven podosome formation in endothelial cells. Eur.
Orlidge A. and D'Amore P.A. (1987). lnhibition of capillary endothelial
J. Cell Biol. 85, 319-325.
cell growth by pericytes and smooth muscle cells. J. Cell Biol. 105,
Morikawa S., Baluk P., Kaidoh T., Haskell A., Jain R. and McDonald D.
1455-1462.
(2002). Abnormalities in pericytes on blood vessels and endothelial
Osiak A.E., Zenner G. and Linder S. (2005). Subconfluent endothelial
sprouts in tumors. Am. J. Pathol. 160, 985-1000.
cells form podosomes downstream of cytokine and RhoGTPase
Moscatelli D. and Rifkin D.B. (1988). Membrane and matrix localization
signaling. Exp. Cell Res. 307, 342-353.
of proteinases: A common theme in tumor invasion and
Ozerdem U and Stallcup W.B. (2004). Pathological angiogenesis is
angiogenesis. Biochim. Biophys. Acta 948, 67-85.
reduced by targeting pericytes via the NG2 proteoglycan.
Moscatelli D., Gross J.L. and Rifkin D.B. (1981). Angiogenic factors
Angiogenesis 7, 269-276.
stimulate plasminogen activator and collagenase production
Ozerdem U., Grako K.A., Dahlin-Huppe K., Monosov E. and Stallcup
bycapillary endothelial cells. J. Cell Biol. 91, 201a
W.B. (2001). NG2 proteoglycan is expressed exclusively by mural
Mota A., Argenziano G., Zalaudek I., Piana S., Longo C., Moscarella E.
cells during vascular morphogenesis. Dev. Dyn. 222, 218-227.
and Lallas A. (2013). Clinical, dermoscopic and histopathologic
Paku S., Dezso K., Bugyik E., Tóvári J., Tímár J., Nagy P., Laszlo V.,
findings of retiform hemangioendothelioma. Dermatol. Pract.
Klepetko W. and Döme B. (2011). A new mechanism for pillar
Concept. 3, 11-14.
formation during tumor-induced intussusceptive angiogenesis:
Moya I.M., Umans L., Maas E., Pereira P.N., Beets K., Francis A., Sents
inverse sprouting. Am. J. Pathol. 179, 1573-1585.
W., Robertson E.J., Mummery C.L., Huylebroeck D. and Zwijsen A.
Patan S. (1998). TIE1 and TIE2 receptor tyrosine kinases inversely
12
Angiogenesis and related secondary

regulate embryonic angiogenesis by the mechanism of


P. (2004). Adult bone marrow-derived cells recruited during
intussusceptive microvascular growth. Microvasc. Res. 56, 1-21.
angiogenesis comprise precursors for periendothelial vascular mural
Patan S. (2000). Vasculogenesis and angiogenesis as mechanisms of
cells. Blood 104, 2084-2086.
vascular network formation, growth and remodeling. J. Neurooncol.
Randles M.J., Humphries M.J. and Lennon R. (2017). Proteomic
50, 1-15.
definitions of basement membrane composition in health and
Patan S., Munn L.L. and Jain R.K. (1996). Intussusceptive
disease. Matrix Biol. 58, 12-28.
microvascular growth in a human colon adenocarcinoma xenograft:
Reynolds L.P., Grazul-Bilska A.T. and Redmer D.A. (2000).
a novel mechanism of tumor angiogenesis. Microvasc. Res. 51, 260-
Angiogenesis in the corpus luteum. Endocrine 12, 1-9.
372.
Rhodin J.A.G. and Fujita H. (1989). Capillary growth in the mesentery of
Patan S., Haenni B. and Burri P.H. (1997). Implementation of
normal young rats. lntravital video and electron microscope
intussusceptive microvascular growth in the chicken chorioallantoic
analyses. J. Submicrosc. Cytol. Pathd. 21, 1-34.
membrane (CAM). Microvasc. Res. 53, 33-52.
Ribatti D and Crivellato E. (2012a). Mast cells, angiogenesis, and
Patan S., Munn L.L., Tanda S., Roberge S., Jain R.K. and Jones R.C.
tumour growth. Biochim. Biophys. Acta 1822, 2-8.
(2001a). Vascular morphogenesis and remodeling in a model of
Ribatti D. and Crivellato E. (2012b). "Sprouting angiogenesis", a
tissue repair: blood vessel formation and growth in the ovarian
reappraisal. Dev. Biol. 372, 157-165.
pedicle after ovariectomy. Circ. Res. 89, 723-731.
Ribatti D. and Djonov V. (2012). Intussusceptive microvascular growth
Patan S., Tanda S., Roberge S., Jones R.C., Jain R.K. and Munn L.L.
in tumors. Cancer Lett. 316, 126-131.
(2001b). Vascular morphogenesis and remodeling in a human tumor
Ribatti D., Nico B. and Crivellato E. (2011). The role of pericytes in
xenograft: blood vessel formation and growth after ovariectomy and
angiogenesis. Int. J. Dev. Biol. 55, 261-268.
tumor implantation. Circ. Res. 89, 732-739.
Ricard N. and Simons M. (2015). When it is better to regress: dynamics
Paulis Y.W., Soetekouw P.M., Verheul H.M., Tjan-Heijnen V.C. and
of vascular pruning. PLoS Biol. 13, e1002148.
Griffioen A.W. (2010). Signalling pathways in vasculogenic mimicry.
Risau W. and Flamme I. (1995). Vasculogenesis. Annu. Rev. Cell Dev.
Biochim. Biophys. Acta 1806, 18-28.
Biol. 11, 73-91.
Peichev M., Naiyer A.J., Pereira D., Zhu Z., Lane W.J., Williams M., Oz
Rizov M., Andreeva P. and Dimova I. (2017). Molecular regulation and
M.C., Hicklin D.J., Witte L., Moore M.A. and Rafii S. (2000).
role of angiogénesis in reproduction. Taiwan J. Obstet. Gynecol. 56,
Expression of VEGFR-2 and AC133 by circulating human CD34(+)
127-132.
cells identifies a population of functional endothelial precursors.
Rojiani A.M. and Dorovini-Zis K. (1996). Glomeruloid vascular structures
Blood 95, 952-958.
in glioblastoma multiforme: an immunohistochemical and
Pelosi E., Valtieri M., Coppola S., Botta R., Gabbianelli M., Lulli V.,
ultrastructural study. J. Neurosurg. 85, 1078-1084.
Marziali G., Masella B., Müller R., Sgadari C., Testa U., Bonanno G.
Ronca R., Benkheil M., Mitola S., Struyf S. and Liekens S. (2017).
and Peschle C. (2002). Identification of the hemangioblast in
Tumor angiogenesis revisited: Regulators and clinical implications.
postnatal life. Blood 100, 3203-3208.
Med. Res. Rev. (in press).
Petrovic N., Schacke W., Gahagan J.R., O'Conor C.A., Winnicka B.,
Rong Y., Durden D.L., Van Meir E.G. and Brat D.J. (2006).
Conway R.E., Mina-Osorio P. and Shapiro L.H. (2007). CD13/APN
'Pseudopalisading' necrosis in glioblastoma: a familiar morphologic
regulates endothelial invasion and filopodia formation. Blood 110,
feature that links vascular pathology, hypoxia, and angiogenesis. J.
142-150.
Neuropathol. Exp. Neurol. 65, 529-539.
Pezzella F., Pastorino U., Tagliabue E., Andreola S., Sozzi G.,
Ruhrberg C., Gerhardt H., Golding M., Watson R., Ioannidou S.,
Gasparini G., Menard S., Gatter K.C., Harris A.L., Fox S., Buyse M.,
Fujisawa H., Betsholtz C. and Shima D.T. (2002). Spatially restricted
Pilotti S., Pierotti M. and Rilke F. (1997). Non-small-cell lung
patterning cues provided by heparin-binding VEGF-A control blood
carcinoma tumor growth without morphological evidence of neo-
vessel branching morphogenesis. Genes Dev. 16, 2684-2698.
angiogenesis. Am. J. Pathol. 151, 1417-1423.
Sabin F.R. (1920). Studies on the origin of the blood vessels and of
Phng L.K., Potente M., Leslie J.D., Babbage J., Nyqvist D., Lobov I.,
redblood corpuscles as seen in the living blastodenn of chick during
Ondr J.K., Rao S., Lang R.A., Thurston G. and Gerhardt H. (2009).
thesecond day of incubation. Contr. Embryol. 9, 215-262.
Nrarp coordinates endothelial Notch and Wnt signaling to control
Sainson R.C., Johnston D.A., Chu H.C., Holderfield M.T., Nakatsu M.N.,
vessel density in angiogenesis. Dev. Cell 16, 70-82.
Crampton S.P., Davis J., Conn E. and Hughes C.C. (2008). TNF
Polverini P.J., Cotran R.S. and Sholley M.M. (1977). Endothelial
primes endothelial cells for angiogenic sprouting by inducing a tip
proliferation in the delayed hypersensitivity reaction: an
cell phenotype. Blood 111, 4997-5007.
autoradiographic study. J. Immunol. 118, 529-537.
Sanz-Trelles A., Rodrigo-Fernandez I., Ayala-Carbonero A. and
Potente M., Gerhardt H. and Carmeliet P. (2011). Basic and therapeutic
Contreras-Rubio F. (1997). Retiform hemangioendothelioma. A new
aspects of angiogenesis. Cell 146, 873-887.
case in a child with diffuse endovascular papillary endothelial
Primo L., Seano G., Roca C., Maione F., Gagliardi P.A., Sessa R.,
proliferation. J. Cutan. Pathol. 24, 440-444.
Martinelli M., Giraudo E., di Blasio L. and Bussolino F. (2010).
Sato Y. and Rifkin D.B. (1989). lnhibition of endothelial cell movement
Increased expression of alpha6 integrin in endothelial cells unveils a
by pericytes and smooth muscle cells: Activation of a latent
proangiogenic role for basement membrane. Cancer Res. 70, 5759-
transforrning growth factor-bl-like molecule by plasmin during
5769.
coculture. J. Cell Biol. 109, 309-315.
Rai A. and Cross J.C. (2014). Development of the hemochorial maternal
Sauteur L., Krudewig A., Herwig L., Ehrenfeuchter N., Lenard A.,
vascular spaces in the placenta through endothelial and
Affolter M. and Belting H.G. (2014). Cdh5/VE-cadherin promotes
vasculogenic mimicry. Dev. Biol. 387, 131-141.
endothelial cell interface elongation via cortical actin polymerization
Rajantie I., Ilmonen M., Alminaite A., Ozerdem U., Alitalo K. and Salven
during angiogenic sprouting. Cell Rep. 9, 504-513.
12
Angiogenesis and related secondary

Sawamiphak S., Seidel S., Essmann C.L., Wilkinson G.A., Pitulescu


occur without proliferation of endothelial cells. Lab. Invest. 51, 624-
M.E., Acker T. and Acker-Palmer A. (2010). Ephrin-B2 regulates
634.
VEGFR2 function in developmental and tumour angiogenesis.
Siekmann A.F. and Lawson N.D. (2007). Notch signalling and the
Nature 465, 487-491.
regulation of angiogenesis. Cell Adh. Migr. 1, 104-106.
Schioppa T., Uranchimeg B., Saccani A., Biswas S.K., Doni A.,
Soda Y., Myskiw C., Rommel A. and Verma I.M. (2013). Mechanisms of
Rapisarda A., Bernasconi S., Saccani S., Nebuloni M., Vago L.,
neovascularization and resistance to anti-angiogenic therapies in
Mantovani A., Melillo G. and Sica A. (2003). Regulation of the
glioblastoma multiforme. J. Mol. Med. (Berl) 91, 439-448.
chemokine receptor CXCR4 by hypoxia. J. Exp. Med. 198, 1391-
Sprague A.H. and Khalil R.A. (2009). Inflammatory cytokines in vascular
1402.
dysfunction and vascular disease. Biochem. Pharmacol. 78, 539-
Schlingemann R.O., Rietveld F.J.R., de Waal R.M.W., Ferrone S. and
552.
Ruiter D.J. (1990). Expression of the high molecular weight
Stratman A.N., Malotte K.M., Mahan R.D., Davis M.J. and Davis G.E.
melanoma-associated antigen by pericytes during angiogenesis in
(2009). Pericyte recruitment during vasculogenic tube assembly
tumors and in healing wounds. Am. J. Pathol. 136, 1393-1405.
stimulates endothelial basement membrane matrix formation. Blood
Schlingemann R.O., Rietveld F.J.R., Kwaspen F., van de Kerkhof
114, 5091-5101.
P.C.M., de Waal R.M.W. and Ruiter D.J. (1991). Differential
Straume O., Chappuis P.O., Salvesen H.B., Halvorsen O.J., Haukaas
expresion of markers for endothelial cells, pericytes, and basal
S.A., Goffin J.R, Bégin L.R., Foulkes W.D. and Akslen L.A. (2002).
lamina in the microvasculature of tumors and granulatlon tissue.
Prognostic importance of glomeruloid microvascular proliferation
Am.J. Pathol. 138, 1335-1347.
indicates an aggressive angiogenic phenotype in human cancers.
Schlingemann R.O., Oosterwijk E., Wesseling P., Rietveld F.J. and
Cancer Res. 62, 6808-6811.
Ruiter D.J. (1996). Aminopeptidase a is a constituent of activated
Styp-Rekowska B., Hlushchuk R., Pries A.R. and Djonov V. (2011).
pericytes in angiogenesis. J. Pathol. 179, 436-442.
Intussusceptive angiogenesis: pillars against the blood flow. Acta
Schor A.M., Schor S.L. and Allen T.D. (1983). Effects of culture
Physiol. (Oxf) 202, 213-223.
conditions on the proliferation morphology and migration of bovine
Suchting S., Freitas C., le Noble F., Benedito R., Bréant C., Duarte A.
aortic endothelial cells. J. Cell Sci. 62, 267-285.
and Eichmann A. (2007). The Notch ligand Delta-like 4 negatively
Schruefer R., Sulyok S., Schymeinsky J., Peters T., Scharffetter-
regulates endothelial tip cell formation and vessel branching. Proc.
Kochanek K. and Walzog B. (2006). The proangiogenic capacity of
Natl. Acad. Sci. USA 104, 3225-3230.
polymorphonuclear neutrophils delineated by microarray technique
Suciu L., Popescu L.M., Gherghiceanu M., Regalia T., Nicolescu M.I.,
and by measurement of neovascularization in wounded skin of
Hinescu M.E. and Faussone-Pellegrini M.S. (2010). Telocytes in
CD18-deficient mice. J. Vasc. Res. 43, 1-11.
human term placenta: morphology and phenotype. Cells Tissues
Seano G. and Primo L. (2015). Podosomes and invadopodia: tools to
Organs 192, 325-339.
breach vascular basement membrane. Cell Cycle 14, 1370-1374.
Sundberg C., Nagy J.A., Brown L.F., Feng D., Eckelhoefer I.A.,
Seano G., Chiaverina G., Gagliardi P.A., di Blasio L., Puliafito A.,
Manseau E.J., Dvorak A.M. and Dvorak H.F. (2001). Glomeruloid
Bouvard C., Sessa R., Tarone G., Sorokin L., Helley D., Jain R.K.,
microvascular proliferation follows adenoviral vascular permeability
Serini G., Bussolino F. and Primo L. (2014). Endothelial podosome
factor/vascular endothelial growth factor-164 gene delivery. Am. J.
rosettes regulate vascular branching in tumour angiogenesis. Nat.
Pathol. 158, 1145-1160.
Cell Biol. 16, 931-941.
Suri C., Jones P.F., Patan S., Bartunkova S., Maisonpierre P.C., Davis
Seftor E.A., Meltzer P.S., Kirschmann D.A., Pe'er J., Maniotis A.J., Trent
S., Sato T.N. and Yancopoulos G.D. (1996). Requisite role of
J.M., Folberg R. and Hendrix M.J. (2002). Molecular determinants of
angiopoietin-1, a ligand for the TIE2 receptor, during embryonic
human uveal melanoma invasion and metastasis. Clin. Exp.
angiogenesis. Cell 87, 1171-1180.
Metastasis 19, 233-246.
Szade A., Grochot-Przeczek A., Florczyk U., Jozkowicz A. and Dulak J.
Seftor R.E., Hess A.R., Seftor E.A., Kirschmann D.A., Hardy K.M.,
(2015). Cellular and molecular mechanisms of inflammation-induced
Margaryan N.V. and Hendrix M.J. (2012). Tumor cell vasculogenic
angiogenesis. IUBMB Life 67, 145-159.
mimicry: from controversy to therapeutic promise. Am. J. Pathol.
Szekanecz Z., Besenyei T., Szentpétery A. and Koch A.E. (2010).
181, 1115-1125.
Angiogenesis and vasculogenesis in rheumatoid arthritis. Curr.
Shi Q., Rafii S., Wu M.H., Wijelath E.S., Yu C., Ishida A., Fujita Y.,
Opin. Rheumatol. 22, 299-306.
Kothari S., Mohle R., Sauvage L.R., Moore M.A., Storb R.F. and
Takahashi T., Kalka C., Masuda H., Chen D., Silver M., Kearney M.,
Hammond W.P. (1998). Evidence for circulating bone marrow-
Magner M., Isner J.M. and Asahara T. (1999). Ischemia- and
derived endothelial cells. Blood 92, 362-367.
cytokine-induced mobilization of bone marrow-derived endothelial
Sholley M.M., Cavallo T. and Cotran R.S. (1977a). Endothelial cell
progenitor cells for neovascularization. Nat. Med. 5, 434-438.
proliferation in inflammation. l. Autoradiographic studies following
Tammela T., Zarkada G., Wallgard E., Murtomäki A., Suchting S.,
themal injured to the skin of nomal rats. Am. J. Pathol. 89, 277-296.
Wirzenius M., Waltari M., Hellström M., Schomber T., Peltonen R.,
Sholley M.M., Gimbrone M.A. and Cotran R.S. (1977b). Cellular
Freitas C., Duarte A., Isoniemi H., Laakkonen P., Christofori G., Ylä-
migration and replication in endothelial regeneration: a study using
Herttuala S., Shibuya M., Pytowski B., Eichmann A., Betsholtz C.
irradiated endothelial cultures. Lab. Invest. 36, 18-25.
and Alitalo K. (2008). Blocking VEGFR-3 suppresses angiogenic
Sholley M.M., Gimbrone M.A. and Cotran R.S. (1978). The effects of
sprouting and vascular network formation. Nature 454, 656-660.
leukocyte depletion on corneal neovascularization. Lab. Invest. 38,
Taylor A.C., Seltz L.M., Yates P.A. and Peirce S.M. (2010). Chronic
32-40.
whole-body hypoxia induces intussusceptive angiogenesis and
Sholley M.M., Ferguson G.P., Seibel H.R., Montuor J.L. and Wilson J.D.
microvascular remodeling in the mouse retina. Microvasc. Res. 79,
(1984). Mechanisms of neovascularization. Vascular sprouting can
93-101.
12
Angiogenesis and related secondary

Tillet E., Vittet D., Féraud O., Moore R., Kemler R. and Huber P. (2005).
Wall R.T., Harker L.A., Quadracci L.J. and Striker G.E. (1978). Factors
N-cadherin deficiency impairs pericyte recruitment, and not
influencing endothelial cell proliferation in vitro. J. Cell Physiol. 96,
endothelial differentiation or sprouting, in embryonic stem cell-
203-213.
derived angiogenesis. Exp. Cell Res. 310, 392-400.
Walsh T.G., Metharom P. and Berndt M.C. (2015). The functional role of
Tirziu D., Jaba I.M., Yu P., Larrivée B., Coon B.G., Cristofaro B.,
platelets in the regulation of angiogenesis. Platelets 26, 199-211.
Zhuang Z.W., Lanahan A.A., Schwartz M.A., Eichmann A. and
Weavers H. and Skaer H. (2014). Tip cells: master regulators of
Simons M. (2012). Endothelial nuclear factor-κB-dependent
tubulogenesis? Semin. Cell Dev. Biol. 31, 91-99.
regulation of arteriogenesis and branching. Circulation 126, 2589-
Wesseling P., van der Laak J.A., de Leeuw H., Ruiter D.J. and Burger
2600.
P.C. (1994). Quantitative immunohistological analysis of the
Todorova D., Simoncini S., Lacroix R., Sabatier F. and Dignat-George
microvasculature in untreated human glioblastoma multiforme.
F. (2017). Extracellular vesicles in angiogenesis. Circ. Res. 120,
Computer-assisted image analysis of whole-tumor sections. J.
1658-1673.
Neurosurg. 81, 902-909.
Tsuda A., Turhan A., Konerding M., Ravnic D., Hanidziar D., Lin M. and
Wesseling P., Schlingemann R.O., Rietveld F.J., Link M., Burger P.C.
Mentzer S.J. (2009). Bimodal oscillation frequencies of blood flow in
and Ruiter D.J. (1995). Early and extensive contribution of
the inflammatory colon microcirculation. Anat. Rec. (Hoboken) 292,
pericytes/vascular smooth muscle cells to microvascular proliferation
65-72.
in glioblastoma multiforme: an immuno-light and immuno-electron
Tsuzuki H. and Sasa S. (1994). Ultrastructural observation of capillary
microscopic study. J. Neuropathol. Exp. Neurol. 54, 304-310.
sprouts in the dental organs of rat molars. Kaibogaku Zasshi 69,
Wietecha M.S., Cerny W.L. and DiPietro L.A. (2013). Mechanisms of
684-696.
vessel regression: toward an understanding of the resolution of
Tzima E., Kiosses W.B., del Pozo M.A. and Schwartz M.A. (2003).
angiogenesis. Curr. Top. Microbiol. Immunol. 367, 3-32.
Localized cdc42 activation, detected using a novel assay, mediates
Williams C.K., Li J.L., Murga M., Harris A.L. and Tosato G. (2006a). Up-
microtubule organizing center positioning in endothelial cells in
regulation of the Notch ligand Delta-like 4 inhibits VEGF-induced
response to fluid shear stress. J. Biol. Chem. 278, 31020-
endothelial cell function. Blood 107, 931-939.
31023.
Williams J.L., Weichert A., Zakrzewicz A., Da Silva-Azevedo L., Pries
Tzima E., Irani-Tehrani M., Kiosses W.B., Dejana E., Schultz D.A.,
A.R., Baum O. and Egginton S. (2006b). Differential gene and
Engelhardt B., Cao G., DeLisser H. and Schwartz M.A. (2005). A
protein expression in abluminal sprouting and intraluminal splitting
mechanosensory complex that mediates the endothelial cell
forms of angiogenesis. Clin. Sci. (Lond) 110, 587-595.
response to fluid shear stress. Nature 437, 426-431.
Woad K.J. and Robinson R.S. (2016). Luteal angiogenesis and its
Udan R.S., Vadakkan T.J. and Dickinson M.E. (2013). Dynamic
control. Theriogenology 86, 221-228.
responses of endothelial cells to changes in blood flow during
Yana I., Sagara H., Takaki S., Takatsu K., Nakamura K., Nakao K.,
vascular remodeling of the mouse yolk sac. Development 140,
Katsuki M., Taniguchi S., Aoki T., Sato H., Weiss S.J. and Seiki M.
4041-4050.
(2007). Crosstalk between neovessels and mural cells directs the
Vajkoczy P., Blum S., Lamparter M., Mailhammer R., Erber R.,
site-specific expression of MT1-MMP to endothelial tip cells. J. Cell
Engelhardt B., Vestweber D. and Hatzopoulos A.K. (2003). Multistep
Sci. 120, 1607-1614.
nature of microvascular recruitment of ex vivo-expanded embryonic
Yu J.A., Castranova D., Pham V.N. and Weinstein B.M. (2015). Single-
endothelial progenitor cells during tumor angiogenesis. J. Exp. Med.
cell analysis of endothelial morphogenesis in vivo. Development
197, 1755-1765.
142, 2951-2961.
van Hinsbergh V.W. and Koolwijk P. (2008). Endothelial sprouting and
Yuri T., Yamazaki F., Takasu K., Shikata N. and Tsubura A. (2008).
angiogenesis: matrix metalloproteinases in the lead. Cardiovasc.
Glomeruloid hemangioma. Pathol. Int. 58, 390-395.
Res. 78, 203-212.
Zeng G., Taylor S.M., McColm J.R., Kappas N.C., Kearney J.B.,
Van Steenkiste C., Trachet B., Casteleyn C., van Loo D., Van
Williams L.H., Hartnett M.E. and Bautch V.L. (2007). Orientation of
Hoorebeke L., Segers P., Geerts A., Van Vlierberghe H. and Colle I.
endothelial cell division is regulated by VEGFsignaling during blood
(2010). Vascular corrosion casting: analyzing wall shear stress in
vessel formation. Blood 109, 1345-1352.
the portal vein and vascular abnormalities in portal hypertensive and
Zengin E., Chalajour F., Gehling U.M., Ito W.D., Treede H., Lauke H.,
cirrhotic rodents. Lab. Invest. 90, 1558-1572.
Weil J., Reichenspurner H., Kilic N. and Ergün S. (2006). Vascular
VanKlompenberg M.K., Manjarín R., Donovan C.E., Trott J.F. and
wall resident progenitor cells: asource for postnatal vasculogenesis.
Hovey R.C. (2016). Regulation and localization of vascular
Development 133, 1543-1551.
endothelial growth factor within the mammary glands during the
Zhang L., Yang N., Park J.W., Katsaros D., Fracchioli S., Cao G.,
transition from late gestation to lactation. Domest. Anim. Endocrinol.
O'Brien-Jenkins A., Randall T.C., Rubin S.C. and Coukos G. (2003).
54, 37-47.
Tumor-derived vascular endothelial growth factor up-regulates
Vitorino, P. and T. Meyer. (2008). Modular control of endothelial sheet
angiopoietin-2 in host endothelium and destabilizes host
migration. Genes Dev. 22, 3268-3281.
vasculature, supporting angiogenesis in ovarian cancer. Cancer
von Tell D., Armulik A. and Betsholtz C. (2006). Pericytes and vascular
Res. 63, 3403-3412.
stability. Exp. Cell Res. 312, 623-629.
Zhang S., Zhang D. and Sun B. (2007). Vasculogenic mimicry: current
Wacker A. and Gerhardt H. (2011). Endothelial development taking
status and future prospects. Cancer Lett. 254, 157-164.
shape. Curr. Opin. Cell Biol. 23, 676-685.
Zhang Y., Lin X., Dai Y., Hu X., Zhu H., Jiang Y. and Zhang S. (2016).
Wakui S. (1988). Two and three dimensional ultrastructural observation
Endometrial stem cells repair injured endometrium and induce
of two cell type angiogenesis in human granulation tissue. Virchows
angiogenesis via AKT and ERK pathways. Reproduction 152, 389-
Arch. (B) 56, 127-139.
402.
12
Angiogenesis and related secondary

Zheng Y., Chen X., Qian M., Zhang M., Zhang D., Bai C., Wang Q. and
Zhou A., Egginton S., Hudlická O. and Brown M.D. (1998). Internal
Wang X. (2014a). Human lung telocytes could promote the
division of capillaries in rat skeletal muscle in response to chronic
proliferation and angiogenesis of human pulmonary microvascular
vasodilator treatment with alpha1-antagonist prazosin. Cell Tissue
endothelial cells in vitro. Mol. Cell. Ther. 1, 2-3.
Res. 293, 293-303.
Zheng Y., Cretoiu D., Yan G., Cretoiu S.M., Popescu L.M., Fang H. and
Zhou Z., Apte S.S., Soininen R., Cao R., Baaklini G.Y., Rauser R.W.,
Wang X. (2014b). Protein profiling of human lung telocytes and
Wang J., Cao Y. and Tryggvason K. (2000). Impaired endochondral
microvascular endothelial cells using iTRAQ quantitative proteomics.
ossification and angiogenesis in mice deficient in membrane-type
J. Cell. Mol. Med. 18, 1035-1059.
matrix metalloproteinase I. Proc. Natl. Acad. Sci. USA 97, 4052-
Zheng Y., Cretoiu D., Yan G., Cretoiu S.M., Popescu L.M. and Wang X.
4057.
(2014c). Comparative proteomic analysis of human lung telocytes
with fibroblasts. J. Cell. Mol. Med. 18, 568-589.
Accepted July 19, 2017

You might also like