View/Open - Lirias

advertisement
Bringing new life to damaged bone: the importance of angiogenesis
in bone repair and regeneration
Steve Stegena,b, Nick van Gastela,b and Geert Carmelieta,b
aLaboratory
of Clinical and Experimental Endocrinology, Department of Clinical and Experimental
Medicine, KU Leuven, 3000 Leuven, Belgium
bPrometheus,
Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
Corresponding Author:
Geert Carmeliet, M.D., Ph.D.
Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and
Experimental Medicine, KU Leuven
O&N1 Herestraat 49 bus 902, 3000 Leuven, Belgium
Tel.: 0032 16 330 731
Fax: 0032 16 330 718
e-mail: geert.carmeliet@med.kuleuven.be
Disclaimer: The authors declare no conflict of interest
1
Abstract
Bone has the unique capacity to heal without the formation of a fibrous scar, likely because
several of the cellular and molecular processes governing bone healing recapitulate the
events during skeletal development. A critical component in bone healing is the timely
appearance of blood vessels in the fracture callus. Angiogenesis, the formation of new blood
vessels from pre-existing ones, is stimulated after fracture by the local production of
numerous angiogenic growth factors. The fracture vasculature not only supplies oxygen and
nutrients, but also stem cells able to differentiate into osteoblasts and in a later phase also
the ions necessary for mineralization.
This review provides a concise report of the regulation of angiogenesis by bone cells, its
importance during bone healing and its possible therapeutic applications in bone tissue
engineering.
Keywords: bone healing, fracture, angiogenesis, VEGF, hypoxia, tissue engineering
Highlights:
-
An adequate angiogenic response, together with abundant mesenchymal stem cells
and mechanical stabilization, is crucial for successful bone healing.
-
The vasculature supplies oxygen and nutrients during bone repair, and may serve as
a niche for osteoprogenitor cells.
-
The timely establishment of a vascular system in tissue-engineered constructs
ensures cell survival and thus improves ultimately bone formation.
-
Preclinical data indicates the benefit of angiogenic factors for bone repair, but
translation into the clinic remains challenging.
2
Abbreviations
αSMA
alpha Smooth Muscle Actin
BMP
Bone Morphogenetic Protein
BOEC
Blood Outgrowth Endothelial Cells
Dll4
Delta-like protein 4
EC
Endothelial Cells
EPC
Endothelial Progenitor Cells
FGF
Fibroblast Growth Factor
FGFR
Fibroblast Growth Factor Receptor
HIF
Hypoxia Inducible Factor
HRE
Hypoxia Responsive Element
miRNA
micro RNA
MMP
Matrix Metalloproteinase
MSC
Mesenchymal Stem Cells
PHD
Prolyl Hydroxylase Domain protein
PlGF
Placental Growth Factor
VEGF
Vascular Endothelial Growth Factor
VEGFR
Vascular Endothelial Growth Factor Receptor
3
Introduction
The establishment of a functional vascular system is crucial during organ development as
well as during tissue repair. In bone, blood vessels not only serve as a source of oxygen and
nutrients, but also supply calcium and phosphate, the building blocks for mineralization. In
addition, blood vessels in the bone marrow appear to have a major role as a niche for both
the bone-forming skeletal stem cells and blood-forming hematopoietic stem cells [1]. The
connection between angiogenesis and osteogenesis is also evident during the healing of
bone fractures, as a timely and coordinated angiogenic response is of vital importance for
successful bone repair [2]. Increasing insight into the molecular and cellular processes
orchestrating the angiogenic cascade may help to develop novel treatments for fracture
healing, especially for clinical situations with a limited angiogenic host response such as
large bone defects or fractures with severe soft tissue trauma. In this review, we will highlight
the current knowledge of developmental skeletal angiogenesis and its regulation by
angiogenic growth factors, as well as the importance of the vascular system during bone
regeneration.
Angiogenesis: a critical process during facture repair
The molecular basis of physiological angiogenesis
When an organism or tissue grows beyond a size where passive diffusion for the exchange
of oxygen, nutrients and metabolic waste products becomes insufficient, the need for the
development of a vascular system emerges. Angiogenesis is the process by which the
organism establishes new blood vessels from pre-existing ones. Recent studies have
highlighted the fundamental aspects of vessel formation, including vasculogenic assembly,
vessel sprouting, lumen formation and vascular remodeling [3, 4].
Sprouting of endothelial cells (EC) is the first in a sequence of events during angiogenesis
[5]. In a resting vessel, both endothelial and mural cells form a basement membrane around
the vessel tube, preventing resident EC to migrate. When new blood vessels are needed in a
4
growing tissue, local production of angiogenic growth factors is enhanced, which trigger the
EC to degrade the basement membrane extracellular matrix, a process mediated by matrix
metalloproteinases (MMPs). The action of these MMPs further fortifies the angiogenic
response by releasing proangiogenic factors that were stored within the matrix. Attracted by
the angiogenic signals, some EC become motile and express filopodia. These so-called tip
cells form the leading front of the newly developing vessel; the trailing EC are called stalk
cells. The key angiogenic factor Vascular Endothelial Growth Factor (VEGF) stimulates tip
cell induction and filopodia formation via VEGF receptor (VEGFR) signaling [4]. The filopodia
on the tip cells ‘sense’ the environment for attractive cues, guiding the sprouts into the tissue
stroma. Stalk cells are equipped to form tubes and branches, mediated by their high
proliferative capacity and the ability to stabilize the newly formed lumen. The priming of EC
into tip or stalk cells is also dependent on the Notch pathway, in close intimacy with VEGFR
signaling. In tip cells, VEGF signaling enhances Delta-like protein 4 (Dll4) expression, a
Notch ligand. Dll4 activates Notch signaling in the neighboring EC, thereby preventing their
switch to a tip cell phenotype and favoring a stalk cell phenotype. Novel vessel circuits are
created via the interaction of two neighboring tip cells (i.e. anastomosis). These connections
are then stabilized by several processes including the deposition of extracellular matrix, the
recruitment of pericytes, reduced endothelial cell proliferation and increased formation of cell
junctions [6]. In addition to the maturation of the endothelium, vascular branches are
remodeled to match its rigidity to the local tissue needs.
Angiogenesis and bone repair
The timely formation of new blood vessels is a critical process during embryonic and fetal
development [7, 8]. In the adult, angiogenesis occurs during physiological processes like
wound healing, the menstrual cycle, and pregnancy but also in specific diseases such as
intraocular neovascular disorders and tumorigenesis. As opposed to soft-tissue healing,
bone can regenerate itself without the formation of fibrous scar tissue and hereby maintains
its physiological and mechanical characteristics. Normal fracture healing in adults occurs
5
through intramembranous or endochondral bone formation, closely mimicking skeletal
development in the embryo [9]. Intramembranous ossification is characterized by direct
formation of bone by committed osteoprogenitor cells and mesenchymal stem cells (MSC)
from the periosteum. During endochondral ossification, MSC differentiate into chondrocytes,
which in turn produce a cartilaginous matrix. Consecutively, this matrix undergoes
calcification and is eventually replaced by bone [10]. The scar-less regeneration of fractured
bones through the endochondral pathway might be attributed to the avascular nature of the
cartilage template. In fact, chondrocytes are metabolically well adapted to survive in poorly
oxygenated regions and still produce the extracellular matrix needed for mineral deposition,
and hereby likely contribute to optimal fracture healing.
Despite the remarkable regenerative capacity of bone tissue, fracture healing fails in about
10% of the cases leading to delayed union or non-union. Adequate vascularization has been
shown to be critical for successful bone healing, next to the presence of osteoprogenitor cells
and mechanical stabilization. Indeed, inappropriate blood vessel supply is a major cause of
delayed union or non-union during fracture healing [11]. More precisely, when fracture is
associated with large vascular injuries, the rate of impaired healing is as high as 46%,
exceeding by far the global 10% non-union rate [2]. In addition, inhibition of angiogenesis
during fracture repair in animal models resulted in the formation of fibrous scar tissue,
resembling human atrophic non-union [12].
Therefore, treatment modalities that promote tissue vascularization possibly provide a central
strategy to accelerate the healing response and tissue regeneration.
6
Factors mediating angiogenesis during fracture healing
During the last years, extensive research has lead to the discovery of a plethora of
angiogenesis-stimulating growth factors that are important during bone development and
normal fracture healing. Some of these factors stimulate angiogenesis directly by inducing
neoangiogenesis and include factors such as VEGF, Placental Growth Factor (PlGF),
Fibroblast Growth Factor (FGF) or members of the Transforming Growth Factor beta family.
Others have angiogenic properties and mainly regulate the production of angiogenic
molecules; examples are Bone Morphogenetic Proteins (BMPs), angiopoietin, PlateletDerived Growth Factor and Insulin-like Growth Factor family members (Table 1). Here, we
will focus on the most extensively studied angiogenic growth factors VEGF, PlGF and FGF.
Vascular Endothelial Growth Factor
One of the most extensively studied angiogenic growth factors is VEGF, an endothelial cellspecific mitogen. VEGF proteins are secreted by cells involved in skeletal development and
repair, including EC, macrophages, fibroblasts, smooth muscle cells, osteoblasts and
hypertrophic chondrocytes [13, 14]. Different from bone development, the early stage of
fracture repair is characterized by hematoma formation, which has strong proangiogenic
properties, predominantly due to the presence of VEGF [15, 16] (Figure 2). The importance
of an appropriate VEGF-mediated stimulation of angiogenesis during fracture healing is
eminent, as inhibition of VEGF activity through treatment with a soluble VEGFR or VEGF
antagonist resulted in impaired healing of femoral fractures and cortical bone defects in mice,
whereas local administration of VEGF lead to improved successful bone repair in both
models [15].
VEGF can influence bone regeneration by affecting bone cells indirectly or directly (Figure 3).
First, through its action on EC, VEGF induces the angiogenic process. Bone forming
precursor cells possibly migrate concomitantly with these blood vessels to the fracture callus,
7
where they differentiate into osteoblasts. Secondly, through an angiocrine mechanism, VEGF
can stimulate EC to produce osteogenic cytokines that promote the differentiation of
progenitor cells into osteoblasts [17]. Lastly, VEGF may also directly influence osteoblast
function. In accordance, osteoblasts not only produce VEGF, they also respond to VEGF
itself, which regulates chemotaxis, proliferation and differentiation of osteoblasts [15, 18-21].
In accordance, VEGF inhibition leads to decreased differentiation of in vitro cultured primary
osteoblasts. In addition, conditional deletion of VEGFR-1 or VEGFR-2 [20-23] in
osteoprogenitors (Osterix-Cre) decreases trabecular bone mass in mice [24]. In this model,
the differentiation of osteoblasts lacking VEGFR-1 or VEGFR-2 was reduced, possibly due to
a decrease in VEGF-mediated RUNX2 induction. However, more experimental evidence is
warranted to elucidate the direct effect of VEGF signaling on osteoblast function during
development and particularly during fracture healing.
Taken together, VEGF activity is indispensable for normal angiogenesis, callus formation and
mineralization in response to injury. These findings support also the notion that VEGF
production is one of the major mechanisms by which angiogenesis and osteogenesis are
tightly coupled during bone repair.
Placental Growth Factor
PlGF, originally discovered in the human placenta, is a VEGF homolog. In contrast to VEGF
which can bind to both VEGFR-1 and VEGFR-2, PlGF binds exclusively to VEGFR-1 [25].
Although PlGF mediates physiological events during angiogenesis, inflammation and
endochondral ossification, it appears to be redundant during development. Indeed, PlGF
deficient mice are viable and show no organ abnormalities. In contrast, PlGF is
nonredundant during several pathological conditions including ischemia, tumor growth or
colitis. Several stimuli are known to induce the expression of PlGF and its receptor in disease
conditions, ranging from hypoxia, growth factors, and hormones to oncogenes.
8
In accordance with this concept, VEGFR-1 and PlGF expression is increased during fracture
healing [23, 26] and Plgf inactivation in mice resulted in reduced fracture healing because of
a decrease in the inflammatory response, osteogenic differentiation and callus remodeling
[26].
Fibroblast Growth Factor
Members of the FGF family regulate proliferation, migration and differentiation of many
different cell types, including skeletal (precursor) cells. Ample in vitro and in vivo evidence
demonstrates a clear proangiogenic role of several members of the FGF family. Most
research has focused on FGF-1 and FGF-2, showing that these factors regulate several
aspects of the angiogenesis process, including endothelial cell proliferation, migration,
integrin and cadherin receptor expression, and intercellular gap-junction communication
(reviewed in [27]). Furthermore, FGF-2 can also induce angiogenesis indirectly through
VEGF/VEGFR signaling: (i) FGF-2 modulates VEGF expression in EC, (ii) VEGFR-2
antagonists inhibit not only VEGF but also FGF-2-induced angiogenesis, and (iii) FGF-2
increases the expression of FGF receptors (FGFRs) as wells as VEGFRs in EC [27].
Several FGF ligands, including FGF-2, FGF-9, and FGF-18, as well as their corresponding
receptors FGFR-1, FGFR-2 and FGFR-3, are involved in skeletal development [28-30] and
fracture repair [31]. Recently, it was suggested that FGF-9 is necessary for the establishment
of a vascular network during fracture healing [32]. However, given the strong mitogenic
character of FGFs, the effect on other cells including progenitor cells [33], chondrocytes [34],
osteoblasts [34] or osteoclasts [35] cannot be excluded. Therefore, more insight in the direct
or indirect regulation of endothelial or osteolineage cell function by FGFs during fracture
healing using cell-specific transgenic knockout models is required.
Matrix metalloproteinases
Cartilage and bone are rich in extracellular matrix and the remodeling of this matrix is critical
for development and repair. MMPs are enzymes mediating localized proteolytic modification
9
of this extracellular matrix, allowing (i) cartilage and bone remodeling and (ii) release of
stored angiogenic signaling molecules and subsequent blood vessel invasion.
During skeletal development, mice lacking membrane-type 1 (MT1) MMP or MMP-9 display
growth plate abnormalities in the long bones and impaired bone formation, concomitant with
a delay in angiogenesis [36-38]. The function of MMP-9 during development was mirrored
during fracture repair. Mmp9-/- mice display delayed bone healing, linked to a defect in the
removal of matrix deposited by the hypertrophic chondrocytes and a delay in vascular
invasion. Surprisingly, this defect was not explained by a decrease in Vegf expression, nor its
receptor. In contrast, MMP-9 likely regulates the bioavailability of VEGF as exogenous
application of VEGF compensated for the lack of Mmp9 [39], although decisive proof is
lacking.
In addition, identifying other angiogenic growth factors and proteases will contribute in
establishing a more complete model of how remodeling of the extracellular matrix and
angiogenesis are linked during bone healing.
Novel insights in the stimulation of angiogenesis during bone repair
Recent research has characterized additional, novel angiogenic factors that are secreted by
bone-forming cells. Kim et al. identified DJ-1 as an angiogenic factor produced by human
MSC [40], which induces a direct angiogenic response in EC through activation of FGFR1
and regulates angiogenesis indirectly by stimulating VEGF production by osteoblasts. In
addition, DJ-1 stimulates the differentiation of MSC and exogenous administration of DJ-1
enhances bone regeneration in a rodent model of bone fracture repair.
Over the last several years, an increasing amount of evidence emerged implicating
microRNAs (miRNAs) in the control of angiogenesis at multiple levels. miRNAs are
expressed in blood vessel constituting cell types such as EC and vascular smooth muscle
cells and have key roles in both development and disease. For example, deletion of Dicer, an
enzyme required for miRNA biogenesis, results in early embryonic lethality, whereas mice
homozygous for a hypomorphic allele of Dicer survive to mid-gestation but have major
10
defects in angiogenesis [41]. In addition, alterations in miRNA expression have been
observed in several pathologies linked to the vascular system, including cancer [42] and
diabetic retinopathy [43].
Besides an effect on angiogenic events, miRNAs appear also to regulate chondrocyte,
osteoblast and osteoclast differentiation [44-46], implying important roles in fracture repair.
Until very recently, the link between miRNAs and angiogenesis during bone healing
remained uncertain. Murata et al. showed that plasma levels of various miRNAs were altered
after fracture, including miRNA-16, miRNA-19b-1, miRNA25, miRNA92a, miRNA101 and
miRNA-129-5p [47]. Interestingly, inhibition of miRNA-92a using systemic and local
administration of antimir markedly enhanced angiogenesis in a murine fracture model,
leading to improved bone repair.
In summary, our knowledge on the role of angiogenic growth factors during bone healing is
progressively increasing, but it has mainly been focused on members of the VEGF and FGF
families. Endorsing insight in the regulation of angiogenesis by other factors, including novel
cytokines and miRNAs, may aid the development of new therapies promoting angiogenesis
during (compromised) fracture healing.
Oxygen as a key determinant of angiogenesis during bone healing
One of the immediate events after a fracture is the rupture of the local (micro)vasculature
resulting in the formation of a hematoma, and these two aspects lead to limited perfusion at
the fracture site and regional hypoxia [48] (Figure 2). Hypoxia Inducible Factor (HIF)
functions as the main transcription factor that regulates cellular responses to hypoxia [49].
HIF is a basic, heterodimeric, helix-loop-helix protein consisting of two subunits, HIF-α and
HIF-β. Under aerobic conditions, HIF-α is hydroxylated by prolyl hydroxylase domain proteins
(PHDs), which use oxygen and α-ketoglutarate as substrates. Hydroxylated HIF-α interacts
with the von Hippel–Lindau protein, the substrate-recognition subunit of an ubiquitin-protein
11
ligase that targets HIF-α for proteasomal degradation. In response to hypoxia, the oxygen
sensing PHDs will become inactive and HIF binds to a specific hypoxia responsive element
(HRE; typical HRE sequence is ACGTG) in the promoter region of genes involved in
anaerobic metabolism (glucose transporter 1, lactate dehydrogenase A), erythropoiesis
(erythropoietin) and vascular response (VEGF, FGF-2, heme oxygenase-1, inducible nitric
oxide synthase).
The function of HIF in the regulation of angiogenesis under physiological and pathological
conditions has been well established (reviewed in [49]). Numerous studies have reported HIF
to be expressed during fracture healing [50, 51] suggesting that activation of the HIFsignaling pathway to induce a VEGF-mediated angiogenic response might be an attractive
therapy to accelerate bone healing. Indeed, genetic overexpression of HIF-1α in mature
osteoblasts of mice was reported to improve bone formation and angiogenesis in a
distraction osteogenesis model, whereas osteoblasts lacking HIF-1α failed to bridge the
distraction gap [52]. In addition, pharmacological inhibition of the prolyl hydroxylases that
normally target HIF-1α for proteosomal degradation stimulated angiogenesis and bone
formation in a femur fracture and distraction osteogenesis model in mice [52, 53]. These
findings indicate that interfering with the hypoxia signaling pathway stimulates both
endochondral and intramembranous bone formation, accompanied by improved blood vessel
formation.
Taken together, the hypoxia signaling pathway is critical during fracture healing to regulate
the timely formation of a functional vascular network, and targeting this pathway is an
appealing strategy to improve bone healing.
12
The vasculature: a source of skeletal stem cells for bone
regeneration
Given the importance of timely vascularization for successful bone healing, the question
remains how the angiogenic response exactly aids in restoring the damaged bone. One
obvious reason is the supply of oxygen, nutrients and minerals for bone formation. Recently,
the importance of blood vessels in delivering osteoprogenitors to the fracture site has also
become appreciated [54].
Several studies have shown that in bone marrow, MSC occupy a perivascular location [5558]. Different markers have been proposed in an attempt to identify these perivascular cells,
including CD-markers (e.g. CD44 or CD146 [56, 59]), Stro1 [60], Platelet-Derived Growth
Factor Receptor β [55], NG2 [61] and alpha Smooth Muscle Actin (αSMA) [62]. More
compelling evidence has been provided by recent studies showing that MSC in the bone
marrow of mice can be identified by their expression of a Nestin-GFP transgene and that
these cells occupy a perivascular location and express the pericyte markers NG2 and αSMA
[57, 63]. Despite the common consensus that perivascular cells play an important role during
development and in various injured tissues [64, 65], knowledge on their role during fracture
healing is rather limited. Perivascular cells in the periosteum are suggested to be a source of
osteoprogenitor cells during periosteal osteogenesis [66]. Recently, using a cell lineage
tracing approach in mice, it was found that αSMA-positive (αSMA+) perivascular cells exhibit
the ability to differentiate into mature osteoblasts in vitro and in vivo during normal bone
remodeling [67]. In addition, during fracture healing, the majority of callus cells including
chondrocytes and osteoblasts were derived from αSMA+ cells. Although the exact origin of
these αSMA+ cells remains to be determined, they presumably derive from the local
periosteal vasculature, since it has been shown that the periosteum supplies the vast
majority of the cells in the fracture callus [68]. In accordance with this notion, we have
recently shown that a subpopulation of culture-expanded murine periosteal cells localize
13
perivascularly when co-transplanted in vivo with EC, indicating that these cells can
structurally support newly formed blood vessels [69].
Targeting angiogenesis to treat non-healing bone defects
The reestablishment of an adequate blood vessel system is of utmost importance for the
success of bone healing. Long-standing preclinical and clinical evidence has established a
strong correlation between the impairment of vascular function and failure of fracture healing
(delayed union or non-union) [70].
Bone tissue engineering represents an appealing strategy to improve non-healing osseous
defects. Reconstruction of damaged bone using this technique relies on the combination of a
biocompatible scaffold, osteogenic cells and osteogenic or angiogenic growth factors.
Furthermore, mechanical stabilization of the defect and an adequate host response are
critical to ensure successful bone formation [71, 72]. Despite the clear potential of
engineered constructs, translation into the clinic remains difficult. One of the major limitations
of this approach is the inability to provide sufficient blood vessel supply during the early
stages of bone regeneration, reflected by the limited survival of the osteogenic cells early
after implantation [73, 74]. Upon implantation of the construct, an angiogenic response is
elicited by inflammatory cytokines as part of the normal healing process [75].
Neovascularization of the scaffold will occur by invading blood vessels deriving from the
surrounding host vasculature. However, the slow rate of invasion of blood vessels into the
scaffold (<1 mm per day) [76] makes it an insufficient process to timely vascularize tissues of
clinically relevant size.
To overcome this problem, several methods have been proposed to improve the
vascularization of bone constructs in experimental models: the delivery of angiogenic growth
factors, the use of EC to engineer a vascular network or a hybrid approach that combines
microsurgery approaches with bone tissue engineering concepts [75] (Figure 4).
14
Local delivery of angiogenic growth factors
The use of angiogenic growth factors such as VEGF is an appealing strategy to improve the
vascularization of a construct. Although a single bolus of VEGF improved revascularization in
animal models [77], this approach might have some limitations due to protein instability in
vivo and possible undesirable side effects. Indeed, careful dosing of VEGF is required, as
overexpression of this angiogenic growth factor leads to aberrant bone formation, bone
marrow fibrosis, abnormal blood vessels and extramedullary hematopoiesis [78]. Current
approaches are therefore more focusing on localized and sustained delivery of growth
factors, permitting prolonged exposure of regenerating tissues to lower doses [79, 80]. To
this end, numerous natural [81], synthetic [82] and composite [83] materials have been
proposed as delivery matrices. Alternatively, advances in gene therapy also facilitate the
controlled release of angiogenic growth factors, either by injection of (viral) vectors or by
using genetically modified implanted cells. Indeed, direct injection of a recombinant adenoassociated virus overexpressing VEGF induced mandibular condylar growth in mice [84].
Furthermore, genetically induced expression of VEGF in MSC has been shown to improve
bone formation in critical sized defects in mice, either when cells were used alone or in
combination with different BMPs and angiopoietin-1 [85, 86].
To conclude, stimulation of an angiogenic response aids bone regeneration in different
preclinical models. However, the timing and dosing of angiogenic growth factors have to be
very tightly regulated, as severe adverse effects might occur upon overstimulation.
Vascularization by autologous endothelial cells
Regardless of the strategy used to stimulate angiogenesis, sufficient and functional
endothelial cells (EC) are as much required as the abundance of angiogenic growth factors.
Several groups are therefore exploring the addition of autologous EC to tissue engineering
implants in order to accelerate the establishment of a functional vascular network.
15
Mature EC can be used for these vascularization strategies, although several drawbacks are
linked to the use of this cell population. First, the low availability and proliferation capacity
limit the upscaling of the constructs for clinical applications [87]. Second, EC derived from
different tissues are phenotypically different [88-90]. In contrast, endothelial progenitor cells
(EPC) might be a more interesting source of autologous EC. These cells are characterized
by the expression of CD133, CD34 and VEGFR-2 and can be isolated from human bone
marrow, fat tissue and peripheral blood. Although limited in number, EPC can be extensively
expanded in vitro and possess the capacity to differentiate into mature EC [91, 92]. EPC are
a heterogeneous cell population consisting of early and late-stage (or blood outgrowth
endothelial cells, BOEC) EPC [93, 94]. Whereas early-stage EPC are considered to be
hematopoietic cells, BOEC are considered to be true endothelial cells. Despite their relatively
late outgrowth from blood cultures, BOEC display enhanced proliferative properties, and
have typical endothelial features such as adherens junctions, in contrast to early-stage EPC.
BOEC, either alone or in combination with early-stage EPC, have already been used in a
variety of preclinical rodent models to restore the vasculature, including bone tissueengineered constructs [95, 96].
Despite the enormous angiogenic potential of these EC, a perivascular component is
required in order to engineer mature, long-lasting blood vessels. Examples of perivascular
cells that can improve the stability and functionality of blood vessels include vascular smooth
muscle cells [97] and bone marrow MSC [98].
Taken together, prevascularization strategies hold promising potential, although they are at
this stage still mostly experimental and translation to the clinic is therefore very limited.
Modulating scaffold properties and in vitro prevascularization
Scaffold properties influence the behavior of the implanted cells and it is obvious that the
porosity is important, not only for osteoblast proliferation and matrix production but also for
blood vessel ingrowth. However, conflicting data make it difficult to extrapolate in vitro
findings to the in vivo situation. In vitro, endothelial cell proliferation was enhanced in
16
scaffolds with smaller pore sizes [99], whereas osteoblast proliferation was diminished [100].
In contrast, higher porosity and pore size resulted in improved vascularization as well as
bone formation in vivo [101]. A recently developed and interesting technique is the fabrication
of a vascular network within a polymer-based scaffold, for example by photolithography [102]
or 3D fiber deposition [103]. Here, 2D and 3D structures are created, serving as template to
seed EC. An alternative way to realize in vitro prevascularization is the co-culture of EC and
osteoblast cells. Several reports indicate that these co-cultures were able to form
microcapillary-like structures within the scaffold, which remained stable during culture [104].
However, only limited evidence exists whether these in vitro pre-made networks can connect
to the host vasculature when implanted in vivo [105].
Preclinical and clinical studies using angiogenic growth factors
It is evident that an adequate vascular response is needed for normal bone healing and the
concept of therapeutic angiogenesis has been appreciated for many years. As discussed in
this review, preclinical data indicate that the local delivery of angiogenic growth factors
correlates with a positive outcome on bone healing. However, rigorous phase II and phase III
clinical trials demonstrating that angiogenic agents are beneficial are largely lacking.
Several factors might contribute to the slow or unsuccessful transition of preclinical to clinical
studies. First, the differences between animal models and the patients enrolled in clinical
trials cannot be neglected. Animals used in preclinical studies are typically young and
healthy, whereas patients are usually older with multiple comorbidities. Often, vascular
function is impaired in the elderly and the reduced angiogenic response compromises bone
repair. Therefore, preclinical experiments in animal models should not only be restricted to
proof-of-principle experiments showing the potential of angiogenic therapy, but should in
addition try to mimic better the compromised human situation and develop more predictive
preclinical models (e.g. aged, atherosclerotic, or diabetic animals).
17
In addition, optimal dosing, duration, and timing of proangiogenic therapy have still to be
defined. Preclinical data suggest that angiogenesis is a slow process requiring the presence
of angiogenic factors during a prolonged period [75, 76], but the dosing strategies have to be
well considered to avoid dose-related side effects.
We can conclude that extensive progress has been made proving the efficacy of the
stimulation of angiogenesis for bone repair in preclinical animal models, but that the
translation into the clinic still remains limited. More relevant animal models, as well as
innovations concerning delivery and dosing of angiogenic stimuli, cell-based constructs and
advances in biomaterial design might facilitate the development of a successful clinical
application.
Conclusions and future perspectives
Fracture healing is a complex process and blood vessel formation, controlled by locally
produced growth factors and cytokines, has to be tightly regulated and has to coincidence
with the activation, proliferation and differentiation of skeletal progenitor cells. Under optimal
conditions, these processes occur synchronously resulting in full restoration of the original
morphology and biomechanical properties of the fractured bones [106]. The vascular system
plays a key role in this process, not only by supplying oxygen and nutrients, but also by
delivering osteoprogenitors to the fracture site giving rise to bone-forming cells [54, 67].
However, these healing processes are likely too slow for the reconstruction of large bone
defects created by tumor resection, trauma or infection, where accelerated bone
regeneration may be required to prevent progression to a non-union. In addition, the
regenerative process might be disturbed in pathological conditions like avascular necrosis or
in diseases associated with impaired vascular function [107]. To circumvent these problems,
strategies might focus on novel techniques for synthesizing scaffolds that serve as a
template for vascularization including 3D fiber deposition. Still, one of the main problems in
18
creating a cell-seeded construct of clinically relevant size is the lag-time in blood vessel
ingrowth, seriously hampering the survival of the implanted cells in the scaffold. Therefore,
an appealing strategy might be to precondition the osteogenic cells prior to in vivo
implantation by adapting their metabolism to the environment. This way, the implanted cells
may remain viable during the early critical time frame before blood vessels arrive, hereby
hopefully improving bone formation.
Taken together, tremendous progress has been made in unraveling the importance of
angiogenesis during bone healing, but novel therapies have to be developed that favor
vascular ingrowth and at the same time promote the survival of the implanted cells,
particularly in patients with an inadequate angiogenic host response.
19
Table 1. Angiogenesis-stimulating growth factors.
Osteoblastica Angiogenicb Induction of VEGFc
Angiopoietin
+ [108]
+ [109]
ND
Fibroblast Growth Factors
+ [110]
+ [32]
+ [111]
Bone Morphogenetic Proteins
+ [112]
+ [113]
+ [113]
Insulin-like Growth Factor family
+ [114]
+ [115]
+ [116]
Platelet-Derived Growth Factor
+ [117]
+ [118]
+ [119]
Transforming Growth Factor beta family
+ [120]
+ [121]
+ [122]
Vascular Endothelial Growth Factor
+ [20]
+ [15]
NA
a
direct effect on committed (pre)osteoblasts
b
stimulation of blood vessel formation in vivo, direct or indirect
c
induction in vitro or in vivo
NA – not applicable, ND – not yet determined
20
Figure legends
Figure 1. Physiological blood vessel formation. The first step of angiogenesis is vessel
sprouting. Angiogenic growth factors trigger endothelial cells to degrade the basement
membrane (A) and select for tip and stalk cells. Tip cells then migrate into the tissue stroma
while stalk cells proliferate (B). A lumen is formed by fusion of tip cells (C-D) and flow
initiates differential branch maturation (E).
Figure 2. The angiogenic response during normal fracture healing. Due to the rupture of
blood vessels, the fracture site becomes hypoxic. Activation of the hypoxia signaling pathway
stimulates the production of VEGF and PlGF by several cell types present at the fracture site.
In addition, early blood vessel formation supports the invasion of inflammatory cells which
actively contribute to the fracture healing process and produce proangiogenic cytokines.
During the formation of the soft and hard callus, the vascular system might also mediate
progenitor migration to the fracture site and promote bone regeneration by supplying oxygen,
nutrients and ions necessary for mineralization.
Figure 3. VEGF signaling during bone regeneration. VEGF stimulates the formation of
new blood vessel, which can bring progenitor cells for bone formation to the fracture callus.
In addition, VEGF upregulates the expression of osteogenic growth factors in endothelial
cells, mediating osteoblast differentiation. Lastly, VEGF might also directly affect osteoblast
behavior including differentiation, proliferation and chemotaxis.
Figure 4. Strategies to improve vascularization in a bone tissue-engineered construct.
The healing of large bone defects using cell-seeded constructs requires adequate
vascularization. Strategies to improve the formation of a functional vascular system include
the addition of growth factors or endothelial cells, and engineering scaffold properties.
21
Acknowledgements
This work was supported by grants from the Fund for Scientific Research Flanders (FWO;
G.0835.11 and G.0A72.13) and the KU Leuven (BOF-KU Leuven GOA project 3M120209).
SS is a fellow from the Agency for Innovation by Science and Technology in Flanders (IWT).
This work is part of Prometheus, the KU Leuven R&D division for skeletal tissue engineering,
http://www.kuleuven.be/prometheus.
References
[1] Gomez-Gaviro MV, Lovell-Badge R, Fernandez-Aviles F, Lara-Pezzi E. The vascular
stem cell niche. J Cardiovasc Transl Res 2012;5:618-30.
[2] Dickson KF, Katzman S, Paiement G. The importance of the blood supply in the healing
of tibial fractures. Contemp Orthop 1995;30:489-93.
[3] Adams RH, Alitalo K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat
Rev Mol Cell Biol 2007;8:464-78.
[4] Jain RK. Molecular regulation of vessel maturation. Nat Med 2003;9:685-93.
[5] Jakobsson L, Franco CA, Bentley K, Collins RT, Ponsioen B, Aspalter IM et al.
Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting.
Nat Cell Biol 2010;12:943-53.
[6] Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell
2011;146:873-87.
[7] Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M et al. Abnormal
blood vessel development and lethality in embryos lacking a single VEGF allele. Nature
1996;380:435-9.
[8] Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nature
medicine 2003;9:669-76.
[9] Ferguson C, Alpern E, Miclau T, Helms JA. Does adult fracture repair recapitulate
embryonic skeletal formation? Mechanisms of development 1999;87:57-66.
[10] Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone
healing. Injury 2005;36:1392-404.
[11] Glowacki J. Angiogenesis in fracture repair. Clin Orthop Relat Res 1998:S82-S89.
[12] Hausman MR, Schaffler MB, Majeska RJ. Prevention of fracture healing in rats by an
inhibitor of angiogenesis. Bone 2001;29:560-4.
[13] Josko J, Gwozdz B, Jedrzejowska-Szypulka H, Hendryk S. Vascular endothelial growth
factor (VEGF) and its effect on angiogenesis. Medical science monitor : international medical
journal of experimental and clinical research 2000;6:1047-52.
22
[14] Bluteau G, Julien M, Magne D, Mallein-Gerin F, Weiss P, Daculsi G et al. VEGF and
VEGF receptors are differentially expressed in chondrocytes. Bone 2007;40:568-76.
[15] Street J, Bao M, deGuzman L, Bunting S, Peale FV, Jr., Ferrara N et al. Vascular
endothelial growth factor stimulates bone repair by promoting angiogenesis and bone
turnover. Proceedings of the National Academy of Sciences of the United States of America
2002;99:9656-61.
[16] Peng H, Wright V, Usas A, Gearhart B, Shen HC, Cummins J et al. Synergistic
enhancement of bone formation and healing by stem cell-expressed VEGF and bone
morphogenetic protein-4. The Journal of clinical investigation 2002;110:751-9.
[17] Bouletreau PJ, Warren SM, Spector JA, Peled ZM, Gerrets RP, Greenwald JA et al.
Hypoxia and VEGF up-regulate BMP-2 mRNA and protein expression in microvascular
endothelial cells: implications for fracture healing. Plastic and reconstructive surgery
2002;109:2384-97.
[18] Zelzer E, McLean W, Ng YS, Fukai N, Reginato AM, Lovejoy S et al. Skeletal defects in
VEGF(120/120) mice reveal multiple roles for VEGF in skeletogenesis. Development
2002;129:1893-904.
[19] Midy V, Plouet J. Vasculotropin/vascular endothelial growth factor induces differentiation
in cultured osteoblasts. Biochemical and biophysical research communications
1994;199:380-6.
[20] Deckers MM, Karperien M, van der Bent C, Yamashita T, Papapoulos SE, Lowik CW.
Expression of vascular endothelial growth factors and their receptors during osteoblast
differentiation. Endocrinology 2000;141:1667-74.
[21] Mayr-Wohlfart U, Waltenberger J, Hausser H, Kessler S, Gunther KP, Dehio C et al.
Vascular endothelial growth factor stimulates chemotactic migration of primary human
osteoblasts. Bone 2002;30:472-7.
[22] Harper J, Gerstenfeld LC, Klagsbrun M. Neuropilin-1 expression in osteogenic cells:
down-regulation during differentiation of osteoblasts into osteocytes. Journal of cellular
biochemistry 2001;81:82-92.
[23] Jacobsen KA, Al-Aql ZS, Wan C, Fitch JL, Stapleton SN, Mason ZD et al. Bone
formation during distraction osteogenesis is dependent on both VEGFR1 and VEGFR2
signaling. Journal of bone and mineral research : the official journal of the American Society
for Bone and Mineral Research 2008;23:596-609.
[24] Liu Y, Berendsen AD, Jia S, Lotinun S, Baron R, Ferrara N et al. Intracellular VEGF
regulates the balance between osteoblast and adipocyte differentiation. J Clin Invest
2012;122:3101-13.
[25] Dewerchin M, Carmeliet P. PlGF: a multitasking cytokine with disease-restricted activity.
Cold Spring Harb Perspect Med 2012;2.
[26] Maes C, Coenegrachts L, Stockmans I, Daci E, Luttun A, Petryk A et al. Placental
growth factor mediates mesenchymal cell development, cartilage turnover, and bone
remodeling during fracture repair. The Journal of clinical investigation 2006;116:1230-42.
[27] Javerzat S, Auguste P, Bikfalvi A. The role of fibroblast growth factors in vascular
development. Trends Mol Med 2002;8:483-9.
23
[28] Montero A, Okada Y, Tomita M, Ito M, Tsurukami H, Nakamura T et al. Disruption of the
fibroblast growth factor-2 gene results in decreased bone mass and bone formation. J Clin
Invest 2000;105:1085-93.
[29] Ohbayashi N, Shibayama M, Kurotaki Y, Imanishi M, Fujimori T, Itoh N et al. FGF18 is
required for normal cell proliferation and differentiation during osteogenesis and
chondrogenesis. Genes Dev 2002;16:870-9.
[30] Liu Z, Lavine KJ, Hung IH, Ornitz DM. FGF18 is required for early chondrocyte
proliferation, hypertrophy and vascular invasion of the growth plate. Dev Biol 2007;302:8091.
[31] Schmid GJ, Kobayashi C, Sandell LJ, Ornitz DM. Fibroblast growth factor expression
during skeletal fracture healing in mice. Dev Dyn 2009;238:766-74.
[32] Behr B, Leucht P, Longaker MT, Quarto N. Fgf-9 is required for angiogenesis and
osteogenesis in long bone repair. Proc Natl Acad Sci U S A 2010;107:11853-8.
[33] Jacob AL, Smith C, Partanen J, Ornitz DM. Fibroblast growth factor receptor 1 signaling
in the osteo-chondrogenic cell lineage regulates sequential steps of osteoblast maturation.
Dev Biol 2006;296:315-28.
[34] Shimoaka T, Ogasawara T, Yonamine A, Chikazu D, Kawano H, Nakamura K et al.
Regulation of osteoblast, chondrocyte, and osteoclast functions by fibroblast growth factor
(FGF)-18 in comparison with FGF-2 and FGF-10. J Biol Chem 2002;277:7493-500.
[35] Chikazu D, Katagiri M, Ogasawara T, Ogata N, Shimoaka T, Takato T et al. Regulation
of osteoclast differentiation by fibroblast growth factor 2: stimulation of receptor activator of
nuclear factor kappaB ligand/osteoclast differentiation factor expression in osteoblasts and
inhibition of macrophage colony-stimulating factor function in osteoclast precursors. J Bone
Miner Res 2001;16:2074-81.
[36] Holmbeck K, Bianco P, Caterina J, Yamada S, Kromer M, Kuznetsov SA et al. MT1MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease
due to inadequate collagen turnover. Cell 1999;99:81-92.
[37] Zhou Z, Apte SS, Soininen R, Cao R, Baaklini GY, Rauser RW et al. Impaired
endochondral ossification and angiogenesis in mice deficient in membrane-type matrix
metalloproteinase I. Proc Natl Acad Sci U S A 2000;97:4052-7.
[38] Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA, Hanahan D et al. MMP9/gelatinase B is a key regulator of growth plate angiogenesis and apoptosis of hypertrophic
chondrocytes. Cell 1998;93:411-22.
[39] Colnot C, Thompson Z, Miclau T, Werb Z, Helms JA. Altered fracture repair in the
absence of MMP9. Development 2003;130:4123-33.
[40] Kim JM, Shin HI, Cha SS, Lee CS, Hong BS, Lim S et al. DJ-1 promotes angiogenesis
and osteogenesis by activating FGF receptor-1 signaling. Nat Commun 2012;3:1296.
[41] Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ et al. Dicer is
essential for mouse development. Nat Genet 2003;35:215-7.
[42] Liu B, Peng XC, Zheng XL, Wang J, Qin YW. MiR-126 restoration down-regulate VEGF
and inhibit the growth of lung cancer cell lines in vitro and in vivo. Lung Cancer 2009;66:16975.
24
[43] McArthur K, Feng B, Wu Y, Chen S, Chakrabarti S. MicroRNA-200b regulates vascular
endothelial growth factor-mediated alterations in diabetic retinopathy. Diabetes
2011;60:1314-23.
[44] Dong S, Yang B, Guo H, Kang F. MicroRNAs regulate osteogenesis and
chondrogenesis. Biochem Biophys Res Commun 2012;418:587-91.
[45] Wang X, Guo B, Li Q, Peng J, Yang Z, Wang A et al. miR-214 targets ATF4 to inhibit
bone formation. Nat Med 2013;19:93-100.
[46] Sugatani T, Vacher J, Hruska KA.
osteoclastogenesis. Blood 2011;117:3648-57.
A
microRNA
expression
signature
of
[47] Murata K, Ito H, Yoshitomi H, Yamamoto K, Fukuda A, Yoshikawa J et al. Inhibition of
miR-92a Enhances Fracture Healing via Promoting Angiogenesis in a Model of Stabilized
Fracture in Young Mice. J Bone Miner Res 2014;29:316-26.
[48] Brighton CT, Krebs AG. Oxygen tension of healing fractures in the rabbit. The Journal of
bone and joint surgery American volume 1972;54:323-32.
[49] Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell 2012;148:399408.
[50] Komatsu DE, Hadjiargyrou M. Activation of the transcription factor HIF-1 and its target
genes, VEGF, HO-1, iNOS, during fracture repair. Bone 2004;34:680-8.
[51] Maes C, Carmeliet G, Schipani E. Hypoxia-driven pathways in bone development,
regeneration and disease. Nat Rev Rheumatol 2012;8:358-66.
[52] Wan C, Gilbert SR, Wang Y, Cao X, Shen X, Ramaswamy G et al. Activation of the
hypoxia-inducible factor-1alpha pathway accelerates bone regeneration. Proceedings of the
National Academy of Sciences of the United States of America 2008;105:686-91.
[53] Shen X, Wan C, Ramaswamy G, Mavalli M, Wang Y, Duvall CL et al. Prolyl hydroxylase
inhibitors increase neoangiogenesis and callus formation following femur fracture in mice.
Journal of orthopaedic research : official publication of the Orthopaedic Research Society
2009;27:1298-305.
[54] Colnot C. Cellular and molecular interactions regulating skeletogenesis. J Cell Biochem
2005;95:688-97.
[55] Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS et al. A perivascular origin
for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008;3:301-13.
[56] Sacchetti B, Funari A, Michienzi S, Di CS, Piersanti S, Saggio I et al. Self-renewing
osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment.
Cell 2007;131:324-36.
[57] Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA et al.
Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature
2010;466:829-34.
[58] Park D, Spencer JA, Koh BI, Kobayashi T, Fujisaki J, Clemens TL et al. Endogenous
bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and
regeneration. Cell Stem Cell 2012;10:259-72.
25
[59] Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A et al. Natural history of
mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci 2013.
[60] Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM et al. Multipotential
human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in
vivo. J Cell Physiol 2008;214:413-21.
[61] Ozerdem U, Grako KA, Dahlin-Huppe K, Monosov E, Stallcup WB. NG2 proteoglycan is
expressed exclusively by mural cells during vascular morphogenesis. Dev Dyn
2001;222:218-27.
[62] Skalli O, Pelte MF, Peclet MC, Gabbiani G, Gugliotta P, Bussolati G et al. Alpha-smooth
muscle actin, a differentiation marker of smooth muscle cells, is present in microfilamentous
bundles of pericytes. J Histochem Cytochem 1989;37:315-21.
[63] Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, Zhang D et al. Arteriolar niches
maintain haematopoietic stem cell quiescence. Nature 2013;502:637-43.
[64] Dellavalle A, Maroli G, Covarello D, Azzoni E, Innocenzi A, Perani L et al. Pericytes
resident in postnatal skeletal muscle differentiate into muscle fibres and generate satellite
cells. Nat Commun 2011;2:499.
[65] Krautler NJ, Kana V, Kranich J, Tian Y, Perera D, Lemm D et al. Follicular dendritic cells
emerge from ubiquitous perivascular precursors. Cell 2012;150:194-206.
[66] Diaz-Flores L, Gutierrez R, Lopez-Alonso A, Gonzalez R, Varela H. Pericytes as a
supplementary source of osteoblasts in periosteal osteogenesis. Clin Orthop Relat Res
1992:280-6.
[67] Grcevic D, Pejda S, Matthews BG, Repic D, Wang L, Li H et al. In vivo fate mapping
identifies mesenchymal progenitor cells. Stem Cells 2012;30:187-96.
[68] Zhang X, Xie C, Lin AS, Ito H, Awad H, Lieberman JR et al. Periosteal progenitor cell
fate in segmental cortical bone graft transplantations: implications for functional tissue
engineering. J Bone Miner Res 2005;20:2124-37.
[69] van Gastel N, Torrekens S, Roberts SJ, Moermans K, Schrooten J, Carmeliet P et al.
Engineering vascularized bone: osteogenic and proangiogenic potential of murine periosteal
cells. Stem Cells 2012;30:2460-71.
[70] Dickson K, Katzman S, Delgado E, Contreras D. Delayed unions and nonunions of open
tibial fractures. Correlation with arteriography results. Clin Orthop Relat Res 1994:189-93.
[71] Lammens J, Laumen A, Delport H, Vanlauwe J. The Pentaconcept in skeletal tissue
engineering. A combined approach for the repair of bone defects. Acta Orthop Belg
2012;78:569-73.
[72] Giannoudis PV, Einhorn TA, Schmidmaier G, Marsh D. The diamond concept--open
questions. Injury 2008;39 Suppl 2:S5-S8.
[73] Giannoni P, Scaglione S, Daga A, Ilengo C, Cilli M, Quarto R. Short-time survival and
engraftment of bone marrow stromal cells in an ectopic model of bone regeneration. Tissue
Eng Part A 2010;16:489-99.
[74] Muschler GF, Nakamoto C, Griffith LG. Engineering principles of clinical cell-based
tissue engineering. J Bone Joint Surg Am 2004;86-A:1541-58.
26
[75] Santos MI, Reis RL. Vascularization in bone tissue engineering: physiology, current
strategies, major hurdles and future challenges. Macromol Biosci 2010;10:12-27.
[76] Nomi M, Atala A, Coppi PD, Soker S. Principals of neovascularization for tissue
engineering. Mol Aspects Med 2002;23:463-83.
[77] Takeshita S, Zheng LP, Brogi E, Kearney M, Pu LQ, Bunting S et al. Therapeutic
angiogenesis. A single intraarterial bolus of vascular endothelial growth factor augments
revascularization in a rabbit ischemic hind limb model. J Clin Invest 1994;93:662-70.
[78] Maes C, Goossens S, Bartunkova S, Drogat B, Coenegrachts L, Stockmans I et al.
Increased skeletal VEGF enhances beta-catenin activity and results in excessively ossified
bones. EMBO J 2010;29:424-41.
[79] Chen RR, Silva EA, Yuen WW, Mooney DJ. Spatio-temporal VEGF and PDGF delivery
patterns blood vessel formation and maturation. Pharm Res 2007;24:258-64.
[80] Zisch AH, Lutolf MP, Hubbell JA. Biopolymeric delivery matrices for angiogenic growth
factors. Cardiovasc Pathol 2003;12:295-310.
[81] Pieper JS, Hafmans T, van Wachem PB, van Luyn MJ, Brouwer LA, Veerkamp JH et al.
Loading of collagen-heparan sulfate matrices with bFGF promotes angiogenesis and tissue
generation in rats. J Biomed Mater Res 2002;62:185-94.
[82] Kaigler D, Wang Z, Horger K, Mooney DJ, Krebsbach PH. VEGF scaffolds enhance
angiogenesis and bone regeneration in irradiated osseous defects. J Bone Miner Res
2006;21:735-44.
[83] Murphy WL, Simmons CA, Kaigler D, Mooney DJ. Bone regeneration via a mineral
substrate and induced angiogenesis. J Dent Res 2004;83:204-10.
[84] Rabie AB, Dai J, Xu R. Recombinant AAV-mediated VEGF gene therapy induces
mandibular condylar growth. Gene Ther 2007;14:972-80.
[85] Huang YC, Kaigler D, Rice KG, Krebsbach PH, Mooney DJ. Combined angiogenic and
osteogenic factor delivery enhances bone marrow stromal cell-driven bone regeneration. J
Bone Miner Res 2005;20:848-57.
[86] Li G, Corsi-Payne K, Zheng B, Usas A, Peng H, Huard J. The dose of growth factors
influences the synergistic effect of vascular endothelial growth factor on bone morphogenetic
protein 4-induced ectopic bone formation. Tissue Eng Part A 2009;15:2123-33.
[87] Kim S, von RH. Endothelial stem cells and precursors for tissue engineering: cell source,
differentiation, selection, and application. Tissue Eng Part B Rev 2008;14:133-47.
[88] Sumpio BE, Riley JT, Dardik A. Cells in focus: endothelial cell. Int J Biochem Cell Biol
2002;34:1508-12.
[89] Garlanda C, Dejana E. Heterogeneity of endothelial cells. Specific markers. Arterioscler
Thromb Vasc Biol 1997;17:1193-202.
[90] Chi JT, Chang HY, Haraldsen G, Jahnsen FL, Troyanskaya OG, Chang DS et al.
Endothelial cell diversity revealed by global expression profiling. Proc Natl Acad Sci U S A
2003;100:10623-8.
27
[91] Luttun A, Carmeliet G, Carmeliet P. Vascular progenitors: from biology to treatment.
Trends Cardiovasc Med 2002;12:88-96.
[92] Hristov M, Erl W, Weber PC. Endothelial progenitor cells: isolation and characterization.
Trends Cardiovasc Med 2003;13:201-6.
[93] Veleva AN, Heath DE, Cooper SL, Patterson C. Selective endothelial cell attachment to
peptide-modified terpolymers. Biomaterials 2008;29:3656-61.
[94] Fuchs S, Hermanns MI, Kirkpatrick CJ. Retention of a differentiated endothelial
phenotype by outgrowth endothelial cells isolated from human peripheral blood and
expanded in long-term cultures. Cell Tissue Res 2006;326:79-92.
[95] Ghanaati S, Fuchs S, Webber MJ, Orth C, Barbeck M, Gomes ME et al. Rapid
vascularization of starch-poly(caprolactone) in vivo by outgrowth endothelial cells in coculture with primary osteoblasts. J Tissue Eng Regen Med 2011;5:e136-e143.
[96] Yoon CH, Hur J, Park KW, Kim JH, Lee CS, Oh IY et al. Synergistic neovascularization
by mixed transplantation of early endothelial progenitor cells and late outgrowth endothelial
cells: the role of angiogenic cytokines and matrix metalloproteinases. Circulation
2005;112:1618-27.
[97] Wu HC, Wang TW, Kang PL, Tsuang YH, Sun JS, Lin FH. Coculture of endothelial and
smooth muscle cells on a collagen membrane in the development of a small-diameter
vascular graft. Biomaterials 2007;28:1385-92.
[98] Melero-Martin JM, De Obaldia ME, Kang SY, Khan ZA, Yuan L, Oettgen P et al.
Engineering robust and functional vascular networks in vivo with human adult and cord
blood-derived progenitor cells. Circ Res 2008;103:194-202.
[99] Narayan D, Venkatraman SS. Effect of pore size and interpore distance on endothelial
cell growth on polymers. J Biomed Mater Res A 2008;87:710-8.
[100] Karageorgiou V, Kaplan D. Porosity of 3D biomaterial scaffolds and osteogenesis.
Biomaterials 2005;26:5474-91.
[101] Bonfield W. Designing porous scaffolds for tissue engineering. Philos Trans A Math
Phys Eng Sci 2006;364:227-32.
[102] Tsuda Y, Shimizu T, Yamato M, Kikuchi A, Sasagawa T, Sekiya S et al. Cellular
control of tissue architectures using a three-dimensional tissue fabrication technique.
Biomaterials 2007;28:4939-46.
[103] Moroni L, de Wijn JR, van Blitterswijk CA. 3D fiber-deposited scaffolds for tissue
engineering: influence of pores geometry and architecture on dynamic mechanical
properties. Biomaterials 2006;27:974-85.
[104] Santos MI, Unger RE, Sousa RA, Reis RL, Kirkpatrick CJ. Crosstalk between
osteoblasts and endothelial cells co-cultured on a polycaprolactone-starch scaffold and the in
vitro development of vascularization. Biomaterials 2009;30:4407-15.
[105] Unger RE, Ghanaati S, Orth C, Sartoris A, Barbeck M, Halstenberg S et al. The rapid
anastomosis between prevascularized networks on silk fibroin scaffolds generated in vitro
with cocultures of human microvascular endothelial and osteoblast cells and the host
vasculature. Biomaterials 2010;31:6959-67.
28
[106]
Einhorn T. Mechanisms of fracture healing. Hosp Pract 1991;26 Suppl 1:41-5.
[107] Dimitriou R, Jones E, McGonagle D, Giannoudis PV. Bone regeneration: current
concepts and future directions. BMC Med 2011;9:66.
[108] Kasama T, Isozaki T, Odai T, Matsunawa M, Wakabayashi K, Takeuchi HT et al.
Expression of angiopoietin-1 in osteoblasts and its inhibition by tumor necrosis factor-alpha
and interferon-gamma. Transl Res 2007;149:265-73.
[109] Suzuki T, Miyamoto T, Fujita N, Ninomiya K, Iwasaki R, Toyama Y et al. Osteoblastspecific Angiopoietin 1 overexpression increases bone mass. Biochemical and biophysical
research communications 2007;362:1019-25.
[110] Marie PJ. Fibroblast growth factor signaling controlling bone formation: an update.
Gene 2012;498:1-4.
[111] Saadeh PB, Mehrara BJ, Steinbrech DS, Spector JA, Greenwald JA, Chin GS et al.
Mechanisms of fibroblast growth factor-2 modulation of vascular endothelial growth factor
expression by osteoblastic cells. Endocrinology 2000;141:2075-83.
[112] Wan M, Cao X. BMP signaling in skeletal development. Biochem Biophys Res
Commun 2005;328:651-7.
[113] Deckers MM, van Bezooijen RL, van der Horst G, Hoogendam J, van der Bent C,
Papapoulos SE et al. Bone morphogenetic proteins stimulate angiogenesis through
osteoblast-derived vascular endothelial growth factor A. Endocrinology 2002;143:1545-53.
[114] Minuto F, Palermo C, Arvigo M, Barreca AM. The IGF system and bone. J Endocrinol
Invest 2005;28:8-10.
[115] Hansson HA, Brandsten C, Lossing C, Petruson K. Transient expression of insulinlike growth factor I immunoreactivity by vascular cells during angiogenesis. Exp Mol Pathol
1989;50:125-38.
[116] Goad DL, Rubin J, Wang H, Tashjian AH, Jr., Patterson C. Enhanced expression of
vascular endothelial growth factor in human SaOS-2 osteoblast-like cells and murine
osteoblasts induced by insulin-like growth factor I. Endocrinology 1996;137:2262-8.
[117] Colciago A, Celotti F, Casati L, Giancola R, Castano SM, Antonini G et al. In Vitro
Effects of PDGF Isoforms (AA, BB, AB and CC) on Migration and Proliferation of SaOS-2
Osteoblasts and on Migration of Human Osteoblasts. Int J Biomed Sci 2009;5:380-9.
[118] Caplan AI, Correa D. PDGF in bone formation and regeneration: new insights into a
novel mechanism involving MSCs. J Orthop Res 2011;29:1795-803.
[119] Bouletreau PJ, Warren SM, Spector JA, Steinbrech DS, Mehrara BJ, Longaker MT.
Factors in the fracture microenvironment induce primary osteoblast angiogenic cytokine
production. Plastic and reconstructive surgery 2002;110:139-48.
[120] Chen G, Deng C, Li YP. TGF-beta and BMP signaling in osteoblast differentiation and
bone formation. Int J Biol Sci 2012;8:272-88.
[121] Yang EY, Moses HL. Transforming growth factor beta 1-induced changes in cell
migration, proliferation, and angiogenesis in the chicken chorioallantoic membrane. J Cell
Biol 1990;111:731-41.
29
[122] Saadeh PB, Mehrara BJ, Steinbrech DS, Dudziak ME, Greenwald JA, Luchs JS et al.
Transforming growth factor-beta1 modulates the expression of vascular endothelial growth
factor by osteoblasts. The American journal of physiology 1999;277:C628-C637.
30
Download