You are on page 1of 16

JPT-06665; No of Pages 16

Pharmacology & Therapeutics xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Pharmacology & Therapeutics


journal homepage: www.elsevier.com/locate/pharmthera

Mesenchymal stem/stromal cells as a pharmacological and therapeutic


approach to accelerate angiogenesis
Annelies Bronckaers ⁎, Petra Hilkens, Wendy Martens, Pascal Gervois, Jessica Ratajczak,
Tom Struys, Ivo Lambrichts
Group of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium

a r t i c l e i n f o a b s t r a c t

Keywords: Mesenchymal stem cells or multipotent stromal cells (MSCs) have initially captured attention in the scientific
Mesenchymal stem/stromal cells world because of their differentiation potential into osteoblasts, chondroblasts and adipocytes and possible
Angiogenesis transdifferentiation into neurons, glial cells and endothelial cells. This broad plasticity was originally hypothe-
Endothelial differentiation sized as the key mechanism of their demonstrated efficacy in numerous animal models of disease as well as in
Secretome clinical settings. However, there is accumulating evidence suggesting that the beneficial effects of MSCs are pre-
Gene therapy
dominantly caused by the multitude of bioactive molecules secreted by these remarkable cells. Numerous angio-
genic factors, growth factors and cytokines have been discovered in the MSC secretome, all have been
demonstrated to alter endothelial cell behavior in vitro and induce angiogenesis in vivo. As a consequence,
MSCs have been widely explored as a promising treatment strategy in disorders caused by insufficient angiogen-
esis such as chronic wounds, stroke and myocardial infarction. In this review, we will summarize into detail the
angiogenic factors found in the MSC secretome and their therapeutic mode of action in pathologies caused by
limited blood vessel formation. Also the application of MSC as a vehicle to deliver drugs and/or genes in (anti-)
angiogenesis will be discussed. Furthermore, the literature describing MSC transdifferentiation into endothelial
cells will be evaluated critically.
© 2014 Elsevier Inc. All rights reserved.

Contents

1. Identification and characterization of mesenchymal stromal/stem cells . . . . . . . . . . . . . . . . . . . . . 0


2. Current concepts of angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Role of MSCs in angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. MSC as vehicles for targeted (anti-)angiogenic drug and gene delivery . . . . . . . . . . . . . . . . . . . . . 0
5. Therapeutic expectations and challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

Abbreviations: AD-MSCs, adipose-derived mesenchymal stem cells; Ang-1, angiopoietin-1; BM, bone marrow; BM-MSC, bone marrow mesenchymal stem cell; CAM, chorioallantoic
membrane; CM, conditioned medium; Cyr61, cysteine-rich, angiogenic inducer 61; EC, endothelial cell; ECM, extracellular matrix; EGF, epidermal growth factor; EGM-2, endothelial
growth medium-2; FGF-2, fibroblast growth factor-2; HGF, hepatocyte growth factor; IGF-1, insulin-like growth factor 1; IL-6, interleukin-6; ISCT, International Society for Cell
Therapy; MCP-1, monocyte chemoattractant protein-1; MI, myocardial infarction; MMP, matrix metalloprotease; MSC, mesenchymal stromal/stem cell; PAD, peripheral artery disease;
PDGF, platelet-derived growth factor; PLGF, placental growth factor; SDF-1, stromal cell derived factor-1; TGF-α, transforming growth factor-α; tMCAO, transient middle cerebral artery
occlusion; TNF-α, tumor necrosis factor α; tPA, tissue plasminogen activator; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor; vWF, von Willebrand Factor; WHO, World
Health Organization.
⁎ Corresponding author at: Department of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium. Tel.: +32 11 26 92 77; fax: +32 11 26 85 99.
E-mail address: annelies.bronckaers@uhasselt.be (A. Bronckaers).

http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
0163-7258/© 2014 Elsevier Inc. All rights reserved.

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
2 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

1. Identification and characterization of mesenchymal stromal/ used to enrich BM-derived MSCs, the fact that to date there is no single
stem cells specific unambiguous determinant to designate MSCs in situ, remains a
particular struggle (Bianco et al., 2008; Caplan & Correa, 2011; Keating,
Since their discovery in the 1960s, the fascinating nature of mes- 2012). On the other hand, it is possible that the ISCT marker profile def-
enchymal stromal/stem cells (MSCs) attracted many scientists and inition of MSCs is too simplistic and unsuitable for MSCs isolated from
this is reflected by the 26,968 PubMed citations on MSCs at the mo- other tissues than the BM, as for example MSCs derived from adipose
ment of this writing. tissue in fact do express CD34 (Traktuev et al., 2008; Keating, 2012). An-
Almost 5 decades ago, the pioneer Friedenstein and his coworkers other drawback is that the expression of these markers may be deter-
identified within the bone marrow (BM) a minor subpopulation of mined or may even be induced by culture conditions rather than
cells that had osteogenic potential in vivo (Friedenstein et al., 1966). being intrinsic characteristics of MSCs in situ. In mice, even strain differ-
These cells could be distinguished from other BM cells (such as the ences influence the expression of surface epitopes on BM-MSCs (Peister
hematopoietic cells) by both their rapid adherence to plastic and et al., 2004). As there is still no consensus on the surface marker expres-
their fibroblast-like morphology. Since these cells showed to be sion profile of MSCs among leading investigators within the field, new
highly clonogenic as they could efficiently generate single-cell and more advanced molecular tools that help unravel the MSC prote-
derived colonies, they were designated as colony-forming unit fibro- ome and secretome could provide a solution to uniformly define MSCs
blasts (Friedenstein et al., 1970). The true physiological and biologi- (Keating, 2012).
cal role for these cells appeared to be support of the hematopoiesis in Traditionally, MSCs are considered to be of ‘pure’ mesodermal origin
the bone marrow (Clark & Keating, 1995). In analogy to the hematopoi- as most skeletal bones are derived from the axial and lateral mesoderm
etic stem cell system, Caplan introduced the popular name ‘mesen- (Bianco et al., 2008; Caplan & Correa, 2011). However, in the embryonic
chymal stem cell (MSC)’ referring to the multipotent differentiation development of craniofacial bones and their marrow, the ectodermal
potential of these cells (Goshima et al., 1991). Later on, it was confirmed neural crest cells are also involved (Hall, 2008). Furthermore, there is
that the progeny of such a single BM-derived cell was able to differenti- accumulating evidence that for almost all organs in the body, an MSC
ate into multiple mesenchymal tissues such as bone, cartilage, adipose population is present in the perivascular niche associated with blood
and fibrous tissues (Pittenger et al., 1999). Besides this mesodermal dif- vessels in vivo. These perivascular cells, principally called pericytes,
ferentiation potential, MSCs are also suggested to form myocardial cells are located at the abluminal side of blood vessels and communicate
(Toma et al., 2002; Kawada et al., 2004), hepatocytes (K. D. Lee et al., with the endothelial cells. Based on immunoselection for CD146, NG2
2004), neuron-like and neuronal cells (Kopen et al., 1999; Wislet- and the pericyte marker PDGF-Rβ, Crisan et al. purified SC from various
Gendebien et al., 2005) under the appropriate induction conditions. In tissues such as skeletal muscle, pancreas, adipose tissue and placenta. In
addition to the bone marrow, MSC populations can be isolated from a addition to their osteogenic, chondrogenic and adipogenic differentia-
wide variety of tissues such as (and not limited to) adipose tissue tion potentials, all cells appeared to be myogenic in vitro and in vivo, re-
(Zuk et al., 2002), dental pulp (Gronthos et al., 2000), amniotic fluid gardless of their origin of skeletal muscle or non-muscle tissues (Crisan
(Nadri & Soleimani, 2007), umbilical cord blood (Erices et al., 2000) et al., 2008). Therefore, it is now speculated that all MSCs indeed are
and even breast milk (Patki et al., 2010). A great contribution to the pericytes and that their biological role is to fuel local tissue growth
popularity of the MSCs can be attributed to their immune-suppressive and repair (Caplan & Correa, 2011).
profile: Le Blanc et al. (2003) reported that MSCs do not express MHC As MSCs display a broad differentiation capacity in vitro, it was orig-
class II surface markers and no co-stimulatory molecules such as inally hypothesized that MSC transplantation would induce tissue re-
CD40, CD80 and CD86, suggesting that MSCs can be “invisible” for the pair by replacing the damaged host tissue. Despite their long-lasting
host immune system and therefore could be clinically applied in allo- therapeutic efficacy in a wide variety of in vivo models and clinical set-
genic settings. tings (such as bone and cartilage repair, cardiovascular and neurological
In spite of their promising and remarkable features, it remains diseases), the incidence of MSC engraftment remained to be surprising-
disputable whether MSCs really fulfill all hallmark ‘stem cell’ charac- ly low. For example, children with osteogenesis imperfecta who re-
teristics, such as self-renewal and differentiation in vivo, as adequate ceived MSC transplantation showed a significant improvement in
test systems for these two properties are still lacking (Bianco et al., bone mineral density and growth velocity, while the population of
2008). Therefore, it is generally considered that the name ‘mesen- engrafted MSCs is less than 1% (Horwitz et al., 2002). Another striking
chymal stromal cell’ (also abbreviated as MSC) is more appropriate example of this low incorporation into the host was demonstrated by
than the widely used historical description ‘mesenchymal stem cell’ Wu et al., who studied the rate of mouse BM-MSC engraftment in a mu-
(Bianco et al., 2008; Prockop & Oh, 2012). rine wound healing model. By using sex-mismatched GFP + mBM-
Another bottleneck in MSC research in the years 1990–2000 was MSCs, the number of cells engrafted into the wounded skin was found
the fact that due to differences and inconsistencies in isolation, ex- to be 27% at 7 days post-injection and even decreased to only 2.5% at
pansion and identification methodology between various research 28 days after administration (Y. Wu et al., 2007). This unexpected low
groups, it was difficult to compare the results of published MSC studies engraftment efficacy implied a major challenge for the field to explain
(Caplan & Correa, 2011; Keating, 2012; Prockop & Oh, 2012). In order to the beneficial effects of the MSCs (as they in most cases only temporar-
clearly define MSCs, the International Society for Cell Therapy (ISCT) ily resided in the host) which was translated into a ‘back to the bench’
proposed the following minimal criteria for MSCs: MSCs have a effect. There is now accumulating evidence that the general therapeutic
fibroblastoid phenotype, adhere to plastic and possess a trilineage effects of MSC treatment are due to their ability to alter the host micro-
mesodermal differentiation capacity towards chondrocytes, adipo- environment rather than their capacity to (trans)differentiate and in-
cytes and osteocytes in vitro. Additional requirements include the corporate into the host tissue. The basic mechanisms, other than
expression of the cell surface molecules CD105 (endoglin), CD73 differentiation, by which MSCs are suggested to affect the host include:
(ecto 5′ nucleotidase) and CD90 (Thy-1) and the absence of the he-
matopoietic markers CD34, CD45, CD14 (or CD11b), CD79α and the (1) the modulation of the immune system (the ‘immunomodulatory
MHC II class cellular receptor HLA-DR (Horwitz et al., 2005). While effect’),
it is assumed that expression of this broad set of markers defines (2) the secretion of factors that induce tissue repair (the trophic or
cells as MSCs, most of these markers are also found on fibroblastic paracrine effect),
cells from any tissue (Bianco et al., 2008). Although various surface (3) recruitment of endogenous MSCs to the site of injury
molecules such as CD146 (Sacchetti et al., 2007), STRO-1 (Simmons (4) possibly transfer of mitochondria or vesicular components con-
& Torok-Storb, 1991) and CD271 (Jones et al., 2002) have been taining mRNA, microRNA and proteins (Fig. 1) (Spees et al.,

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 3

Paracrine/Trophic effects Immunomodulatory effects


MSC CD8+ T cell
Neutrophil

Treg cell NK cell B cell

Angiogenesis
CD4+ T cell

Plasma cell

Monocyte Dendritic cell

Proliferation and differentiation of host cells Differentiation


MSC

Neuron
Anti-apoptosis Cardiomyocyte
Chondrocyte Adipocyte
Osteocyte

Transfer of vesicular components


Recruitment of endogenous stem cells
MSC

Fig. 1. Proposed mechanisms of action of MSC transplantation. MSCs secrete a wide variety of proteins that affect host cells, a phenomenon that is called the trophic or paracrine effect. This
includes the induction of angiogenesis, protection of the host cells against apoptosis, stimulation of host cell proliferation and local stem cell or precursor cell differentiation and the re-
cruitment of endogenous stem cells to the site of engraftment. The immunomodulatory effects of MSC consist of inhibiting the proliferation and activity of neutrophils, NK cells, B cells,
CD4+ cells and CD8+ T cells, preventing the maturation of monocytes into dendritic cells, suppressing plasma cell immunoglobulin production but stimulating proliferation of regulatory
T cells. In some physiological settings, MSCs are able to differentiate themselves into multiple cell types and to transfer vesicles containing mRNA, proteins, microRNA and perhaps mito-
chondria to the host cells.

2006; Meirelles Lda et al., 2009; Hocking & Gibran, 2010; Ben- and where MSCs can (functionally) improve blood vessel formation
Ami et al., 2011; Caplan & Correa, 2011; Bieback et al., 2012; will be essential for unmasking new contributions by these cells in
Prockop & Oh, 2012). a whole array of diseases.

The local immunosuppressive actions of MSCs have fuelled hope


of allogeneic MSC treatment in settings where the use of autologous 2. Current concepts of angiogenesis
cells is limited or even impossible. The trophic and immunomodula-
tory activities of the MSCs are considered to account for the majority Within the human body, a complex labyrinth of blood vessels
of the beneficial effects of MSC and inspired Caplan to launch the provides all cells and tissues with vital nutrients and oxygen needed
concept of MSCs as a ‘drugstore’, secreting specialized bioactive mol- for survival and proliferation. This network includes large, stable ar-
ecules which prevent unfavorable immune reactions and support the teries and veins as well as small, dynamic microvascular structures
injured tissue to repair (Caplan & Correa, 2011). This spectrum of such as capillaries, arterioles and venules.
bioactive molecules is generally defined as the MSC secretome and In the embryo, new blood vessels arise through vasculogenesis, mean-
includes growth factors, chemokines and cytokines (Ranganath ing the de novo formation of a basic vasculature out of mesoderm-derived
et al., 2012). endothelial precursors, termed angioblasts (Friedenstein et al., 1970;
Since MSC research is now predominantly focused on the thera- Herbert & Stainier, 2011). Subsequently, this primordial vascular bed is
peutic advantages of the secretome, more than on the potential dif- enlarged by a process defined as angiogenesis: the branching of new vas-
ferentiation of engrafted MSCs, we will summarize the paracrine cular sprouts out of pre-existing ‘parental’ blood vessels (Friedenstein
effects of MSCs particularly in diseases that are caused by limited et al., 1970; Folkman, 1971; Ribatti & Crivellato, 2012). Other mechanisms
or insufficient angiogenesis. First, the process of angiogenesis and of vascular development include splitting of pre-existing blood vessels
the pro-angiogenic role of MSCs in vitro and in vivo are described. (intussusception) or stimulation of vessel expansion by circulating pre-
In addition, the transdifferentiation potential of MSCs towards endo- cursor cells. However, the majority of the blood vessel network is consid-
thelial cells is overviewed in detail. A greater understanding of how ered to be built through angiogenic processes (Friedenstein et al., 1970).

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
4 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

A B C

D E

F G

Figure legend:
Endothelial cell (EC) Basement membrane

Pericyte Matrix metalloproteinase


Chemokine
Tip cell
Chemokine receptor
Stalk cell Growth factor

Extracellular matrix (ECM) Growth factor receptor

Fig. 2. Key steps of angiogenesis. A) Small blood vessels consist of hollow tubes aligned with endothelial cells (EC) surrounded by pericytes, a basement membrane (BM) and extracellular
matrix (ECM). Angiogenesis is a multistep process involving the following steps: (B) pericyte detachment and breakdown of the extracellular matrix and basement membrane by, for ex-
ample matrix metalloproteinases, (C) EC activation by angiogenic factors such as growth factors and cytokines, (D–E) formation of a tip cell which guides the migration and a stack cell
which proliferates and forms a primitive tube (F), and (G) finally pericyte attachment, deposition of BM and EC followed by maturation into fully functional blood vessels.

Normal physiological angiogenesis is a fundamental event during et al., 2011; Carmeliet & Jain, 2011b). This type of blood vessel forma-
embryonic development, wound and fracture healing and for the tion is tightly regulated in a spatial and temporal manner by the ex-
growth and function of the female reproductive organs (Bhadada tensive interplay of a wide variety of molecules such as matrix

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 5

metalloproteases, growth factors, chemokines, adhesion molecules, 3. Role of MSCs in angiogenesis


enzymes and angiogenic inhibitors (Carmeliet, 2005; Ferrara &
Kerbel, 2005; Carmeliet & Jain, 2011a). 3.1. The MSC secretome as a powerful inducer of angiogenesis
Fig. 2 visualizes the key steps of angiogenesis. Small blood capil-
laries consist of hollow tubes composed of endothelial cells (EC) sup- 3.1.1. Angiogenic factors in the MSC secretome
ported by pericytes and surrounded by a basement membrane and Table 1 summarizes the angiogenic mediators and their functions
extracellular matrix (ECM) (Eble & Niland, 2009) (Fig. 2A). In the detected in MSCs of different tissue sources. Using ELISA, antibody
adult, blood vessels and EC remain in a quiescent state and are rarely arrays and immunohistochemistry a broad repertoire of angiogenic
activated to form new branches. The basement membrane and factors have been detected in the secretome of BM-MSCs including
pericytes surrounding the EC prevent the EC from leaving their posi- VEGF, FGF-2, angiopoietin-1 (Ang-1), monocyte chemoattractant
tions. Only in response to specific stimuli such as hypoxia and inflam- protein-1 (MCP-1), interleukin-6 (IL-6) and placental growth factor
mation, the angiogenic process is initiated (Carmeliet & Jain, 2011b). (PLGF) (Kinnaird et al., 2004a; Hung et al., 2007; Wu et al., 2007; L.
First, pericytes detach and both the ECM and basement membrane are Chen et al., 2008; Boomsma & Geenen, 2012). Recently, Estrada et al.
degraded by proteases such as the matrix metalloproteases (MMPs) identified the presence of the protein Cyr61 (cysteine-rich, angiogenic
(Fig. 2B). This leads to destabilization of the blood vessels and an in- inducer 61) in the mBM-MSCs with advanced mass spectrometry.
crease in vascular permeability. In addition, the proteolytic breakdown Addition of an antibody against Cyr61 inhibited BM-MSC-induced
of ECM releases sequestered proangiogenic growth factors and EC tube formation in vitro and blood vessel formation in vivo in the
chemokines, such as vascular endothelial growth factor (VEGF), fibro- mouse matrigel assay, indicating that Cyr61 plays a key role in BM-
blast growth factor-2 (FGF-2) and stromal cell derived factor-1 (SDF- MSC-mediated angiogenesis (Estrada et al., 2009). Concerning
1) (Arroyo & Iruela-Arispe, 2010). Once liberated, these proteins are angiogenic receptors, it has been shown that BM-MSCs lack VEGF-
able to bind to the receptors of the EC, thereby activating the EC and in- receptors, but they do express their co-receptor neuropilin-1,
ducing the start of branch formation (Fig. 2C). This initiation of which co-localizes with platelet-derived growth factor (PDGF)-re-
sprouting requires the specification of EC into ‘tip’ and ‘stalk’ cells ceptor. This cross-talk is important in MSC proliferation, migration
with different morphologies and functional properties, a process and network formation (Ball et al., 2010). Placental MSCs produce
which is tightly regulated by VEGF/Notch signaling (Phng & Gerhardt, IL-6 (S. H. Liu et al., 2010), while adipose-derived MSCs (AD-MSCs)
2009; Potente et al., 2011; Ribatti & Crivellato, 2012) (Fig. 2D). EC tip secrete high amounts of VEGF, hepatocyte growth factor (HGF) and
cells have numerous filopodia and primarily migrate and coordinate transforming growth factor β (TGF-β) (Rehman et al., 2004). Elevat-
the direction of the branch formation, while stalk cells follow the tip ed levels of VEGF and MCP-1 have also been found in human dental
cells and proliferate during sprout elongation. These stalk cells also pulp stem cells (hDPSCs) (Kandel & Pittenger, 1999; Aranha et al.,
form the lumen of the primitive blood vessel, either by the fusion of in- 2010).
tracellular pinocytic vesicles or by rearrangement of their shape and The secretion of angiogenic factors by MSCs can be upregulated
junctions (Potente et al., 2011; Ribatti & Crivellato, 2012) (Fig. 2E). by a range of chemokines and hypoxic conditions. De Luca et al. dem-
Subsequently, the stable sprout begins to transform into a mature onstrated that transforming growth factor α (TGF-α) significantly
blood vessel and fuses with an existing capillary (Fig. 2F–G). New induces the secretion of various growth factors such as VEGF, HGF,
ECM is deposited around the new tube, recruited pericytes cover IL-6, IL-8, PDGF-BB and Ang-2 in BM-MSCs. Both the MEK/MAPK
the EC and finally, a stable blood vessel with a functional blood and PI3K/AKT pathways were involved in the ability of TGF-α to in-
flow is established (Fig. 2G). Without pericyte attachment, blood crease the levels of these factors in the BM-MSC secretome. Conse-
vessels are considered to be unstable and can regress rapidly quently, conditioned medium (CM) from TGF-α treated MSCs
(Potente et al., 2011). induced more blood vessel growth compared to CM from untreated
The equilibrium between angiogenic stimulators and inhibitors BM-MSCs in the in vivo chorioallantoic membrane (CAM) assay
in the microenvironment, the so-called angiogenic balance, tightly (De Luca et al., 2011). In addition, Crisostomo et al. (2008) demon-
controls the rate of blood vessel formation (Bergers & Benjamin, strated that hypoxia, tumor necrosis factor α (TNF-α) and LPS stim-
2003). Numerous angiogenic stimulators exist, including FGF-2, ulated the production of VEGF, FGF-2, HGF and insulin-like growth
SDF-1, placental growth factor (PLGF) and epidermal growth factor factor 1 (IGF-1) via induction of the transcription factor NF-κB. In
(EGF). VEGF, one of the most studied angiogenic proteins, is consid- AD-MSCs, LPS increased the secretion of several chemokines includ-
ered to be the most potent mediator of neovascularization in health ing IL-6 and TNF-α, whereas FGF-2 and epidermal growth factor
and disease (Carmeliet & Jain, 2011a; Potente et al., 2011). However, (EGF) upregulated HGF production (Kilroy et al., 2007). BM-MSCs
recent studies reveal the existence of alternative splicing variants of preconditioned with interferon-β (IFN-β) produced higher levels of
VEGF (the VEGF-xxxb isoforms) which may have anti-angiogenic and MCP-1 and interferon gamma-induced protein 10 than untreated MSC
anti-tumoral properties (Woolard et al., 2009; Harris et al., 2012). (Croitoru-Lamoury et al., 2007). Under hypoxic conditions, the CM of
Thrombospondin, endostatin, angiostatin and angiopoietin-2 are other BM-MSCs contained higher amounts of for instance VEGF, IGF-1, SDF-
examples of endogenous angiogenic inhibitors (Carmeliet & Jain, 1 and erythropoietin compared to CM of dermal fibroblasts (L. Chen
2011a; Potente et al., 2011). Disturbance of angiogenic balance et al., 2008). It has also been shown that hypoxia elevates secretion of
can lead to a growing list of life-threatening pathological disorders. VEGF but not of FGF-2 in hDPSC cultures due to upregulation of the
Extensive angiogenesis, for instance, contributes to rheumatoid transcription factor HIF-1α (Aranha et al., 2010). Accordingly, also in
arthritis, psoriasis, blinding eye diseases and cancer growth and AD-MSCs, hypoxia selectively increased VEGF but not FGF-2, HGF and
metastases. On the other hand, inadequate stimulation of the an- TGF-β mRNA and protein levels (Rehman et al., 2004). Interestingly,
giogenic process is one of the main causes of insufficient recovery serum-deprivation during 30 days elevated the levels of VEGF, HGF,
following ischemic insults as seen in atherosclerosis, stroke, myo- IGF-1 and Ang in the secretome of human BM-MSCs (Bianco et al.,
cardial infarction and chronic wounds (Bhadada et al., 2011; 2008). CM of these serum-deprived cells generated longer neovascular
Carmeliet & Jain, 2011a). In addition, in the field of tissue engineer- sprouts in the ex vivo aortic ring assay compared to CM of control BM-
ing, the main reason of tissue transplantation failure is the lack of MSCs (Bianco et al., 2008).
proper angiogenesis, causing necrosis of the engrafted cells
(Laschke et al., 2006). It is in these clinical settings, where angio- 3.1.2. MSCs alter the behavior of EC in vitro
genesis is limited, that MSC transplantation might be promising As angiogenesis is a multi-step process involving EC survival, prolif-
as an efficient therapy. eration, migration, tube formation and maturation, several in vitro

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
6 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

Table 1
Angiogenic factors secreted by cultured MSCs.

Angiogenic factor Full-name Function MSC source REF

Angiogenin Angiogenin Cell migration, invasion, proliferation and tube formation hBM-MSC Hung et al., 2007
Ang-1 Angiopoietin-1 Vessel stabilization, EC survival, recruitment of pericytes mBM-MSC Wu et al., 2007
Ang-2 Angiopoietin-2 EC migration and sprouting mBM-MSC Wu et al., 2007
Cyr61 Cysteine-rich 61 Cell adhesion and EC migration mBM-MSC Estrada et al., 2009
FGF-2 Fibroblast growth factor-2 EC proliferation, migration, remodeling of the extracellular matrix mBM-MSC Kinnaird et al., 2004a,
HGF Hepatocyte growth factor EC and SMC proliferation, migration rBM-MSC Tögel et al., 2007
hAD-MSC Rehman et al., 2004
IGF-1 Insulin-like-growth factor-1 EC proliferation, survival, induction VEGF and plasminogen activators rBM-MSC Tögel et al., 2007
IL6 Interleukin-6 EC proliferation and migration hBM-MSC Hung et al., 2007
hPl-MSC Liu et al., 2010
IL8 Interleukin-8 EC proliferation, survival, migration and tube formation hBM-MSC Majumdar et al., 1998;
De Luca et al., 2011
MCP-1 Monocyte chemoattractant protein-1 EC migration mBM-MSC Kinnaird et al., 2004a;
Boomsma & Geenen, 2012
hBM-MSC Hung et al., 2007
MIG Monokine induced by interferon-gamma Chemoattractant of T-cells, inhibition of angiogenesis mBM-MSC Boomsma & Geenen, 2012
MIP-1α Macrophage inflammatory protein-1alpha Proinflammation, chemoattractants of immune cells mBM-MSC Boomsma & Geenen, 2012
MIP-1β Macrophage inflammatory protein-1beta Proinflammation, chemoattractants of immune cells mBM-MSC Boomsma & Geenen, 2012
NAP-2 (CXCL7) Neutrophil activating protein 2 Neutrophil recruitment, EC migration, release of VEGF and MMP hBM-MSC Hung et al., 2007
PLGF Placental growth factor (PLGF) Induction of vessel formation mBM-MSC Kinnaird et al., 2004a,
TGF-beta Tumor growth factor-beta Tube formation, vessel stabilization, ECM synthesis hAD-MSC Rehman et al., 2004
TIMP-1 Tissue inhibitor of metalloproteinase-1 Inhibitor of metalloproteinases hBM-MSC Hung et al., 2007
TIMP-2 Tissue inhibitor of metalloproteinase-2 Inhibitor of metalloproteinases hBM-MSC Hung et al., 2007
VEGF Vascular endothelial growth factor Increase vessel permeability, ECM degradation, EC proliferation, mBM-MSC Kinnaird et al., 2004a,
migration, tube formation and survival Wu et al., 2007
hBM-MSC Hung et al., 2007
hDPSC Bronckaers et al., 2013,
Tran-Hung et al., 2008
hAD-MSC Rehman et al., 2004

models exist that mimic each of these essential events. There is detailed described that BM-MSCs co-aligned with tubular EC structures in a
and elegant literature that describes the effect of MSCs on each of these pericyte-like manner and some BM-MSC populations enhanced the
phases in vitro. complexity and extent of these tubes in a 3D matrigel co-culture sys-
AD-MSCs have been shown to induce EC proliferation (Rehman tem (Sorrell et al., 2009). Accordingly, Au et al. (2008) have demon-
et al., 2004), while MSCs derived from pulp tissue had no effect on strated that hBM-MSCs efficiently stabilized nascent blood vessels
EC cell growth (Kandel & Pittenger, 1999; Bronckaers et al., 2013). in vivo by functioning as perivascular precursor cells in an in vivo tis-
For BM-MSCs, contradictory results have been reported: Two studies sue engineered blood vessel construct. These findings are in concor-
indicated that the CM of BM-MSCs had no effect (Gruber et al., 2005; dance with the current hypothesis that MSCs are indeed pericytes
Potapova et al., 2007), whereas others demonstrated that BM-MSCs (Caplan & Correa, 2011). On the other hand, Otsu et al. (2009)
do increase EC proliferation (Kinnaird et al., 2004b; Potapova et al., demonstrated that addition of high numbers of BM-MSCs caused an
2007; Duffy et al., 2009). These conflicting results might be ex- inhibition of tube formation in a matrigel assay due to the production
plained by variations in isolation methods, species (mouse versus of cytotoxic reactive oxygen species. The observed anti-angiogenic
human) and EC sources used. effect was considered to be caused by the large amount of administered
In addition, EC migration in vitro was induced by various MSC MSCs, as the EC cytotoxicity was significantly reduced when the num-
populations, including Wharton Jelly-derived umbilical vein MSCs ber of added BM-MSCs also decreased. These results strongly indicate
(Choi et al., 2013), amniotic MSCs (Kim et al., 2013), AD-MSCs that administration of high MSC concentrations should be avoided in
(Takahashi et al., 2010), human dental pulp stem cells (DPSCs) MSC-based treatments as it can potentially induce apoptosis of newly
(Bronckaers et al., 2013) and BM-MSCs (Gruber et al., 2005; formed blood vessels (Otsu et al., 2009).
Potapova et al., 2007; Burlacu et al., 2013). Furthermore, the ability MSCs also have the capacity to protect EC from apoptosis in vitro. The
of EC to degrade the surrounding ECM in vitro (in the so-called inva- results published by Hung et al. (2007) demonstrated that the secretome
sion assay) is stimulated by MSC. AD-MSCs as well as BM-MSCs have of BM-MSCs inhibited hypoxia-induced apoptosis of EC, probably
been shown to induce EC invasion, albeit through upregulation of through activation of the PI3K/Akt and not the ERK pathway. In coculture
distinct proteases (Kinnaird et al., 2004b; Kachgal & Putnam, with BM-MSCs, apoptosis of UV-irradiated fibroblasts and also of lung
2011). Potapova et al. (2007) demonstrated that CM of BM-MSCs epithelial cells incubated under hypoxic and low pH conditions was de-
cultured in special three-dimensional aggregates had a more pro- creased through the secretion of the peptide hormone stanniocalcin-1
nounced effect on EC proliferation, migration and invasion than CM (Block et al., 2009). Placental MSCs significantly reduced oxidative
of monolayer BM-MSCs, probably due to a higher concentration of stress-related apoptosis (induced by the chemical tert-butyl hydroperox-
VEGF, FGF-2, angiogenin and IL-11. ide) via the STAT3 signal transduction cascade (S. H. Liu et al., 2010). CM
BM-MSC and CM of BM-MSC induce EC tube formation on colla- of hypoxic AD-MSCs abrogated EC cell death induced by a cocktail of cy-
gen and matrigel in vitro (Wu et al., 2007; Duffy et al., 2009; cloheximide and TNF-α (Rehman et al., 2004).
Estrada et al., 2009; Boomsma & Geenen, 2012). In addition, it was In conclusion, MSCs are involved in all stages of angiogenesis, not
demonstrated that human BM-MSCs in direct co-culture with EC on only in the early steps such as proliferation or migration of EC but also
matrigel increased the persistence of pre-existing blood vessels, sug- in the later phases which involve blood vessel maturation.
gesting that BM-MSCs not only promote tube formation, but also
play an active role in stabilization and maturation of newly formed 3.1.3. MSCs have angiogenic properties in vivo
vessels. This effect was considered to be caused by direct cell contact In this section, we will first discuss the in vivo angiogenic capacities in
between the BM-MSCs and EC (Duffy et al., 2009). Another study animal models that allow one to exclusively investigate neoangiogenesis

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 7

in the absence of any pathology, such as the ‘chicken chorioallantoic low (less than 1%). In addition, no integration of the transplanted
membrane (CAM) assay’ and the ‘mouse matrigel plug assay’. Next, we GFP + BM-MSCs into the host vasculature was observed (Y. Wu et al.,
discuss the benefits of MSC-treatment in animal models and human clin- 2007). Some investigators state that besides induction of angiogenesis,
ical trials that display a wide variety of disorders caused by limited angio- MSCs contribute to wound repair by differentiation into keratinocytes
genesis. A summary of all published scientific data is beyond the scope of (Sasaki et al., 2008), although evidence of this transdifferentiation is
this review, and hence, only key observations that exemplify the pro- not provided in all studies (Hocking & Gibran, 2010). Moreover, the ap-
angiogenic impact of MSC administration will be highlighted. In addition, plication of concentrated CM of BM-MSCs or CM of hypoxia-pretreated
contradictions in the literature will also be discussed critically. AD-MSCs to murine excisional wounds significantly enhanced wound
healing (L. Chen et al., 2008; E. Y. Lee et al., 2009). Addition of neutral-
3.1.3.1. CAM assay. The chicken chorioallantoic membrane (CAM) is an izing antibodies against VEGF and FGF-2 to the wounds treated with
extra-embryonic, highly vascularized tissue that serves as a transient the AD-MSC CM reduced the wound repair, indicating that upregulation
gas exchange surface for the embryo (comparable to the lungs in of angiogenesis caused the observed amelioration (E. Y. Lee et al., 2009).
adult vertebrates). The main cellular components of the CAM are In contrast, another study addresses the beneficial effect of the BM-MSC
EC and mural cells such as pericytes. Traditionally, this model has CM to the induced recruitment of macrophages and endothelial progen-
been successfully used to investigate angiogenic factors in developmen- itor cells to the site of injury (L. Chen et al., 2008). Interestingly, a small
tal biology and to select anti-angiogenic compounds for anticancer pilot study in patients with acute or severe chronic wounds showed
treatments. As the early chicken embryo lacks a complete immune sys- that topical administration of autologous BM-MSCs with a fibrin spray
tem, mammalian cells or tissues can be placed onto the CAM without re- significantly reduced wound size (Falanga et al., 2007).
jection allowing a simple assessment of the angiogenic potential of the
transplant (Laschke et al., 2006). BM-MSCs, CM of BM-MSCs, hDPSCs 3.1.4. MSCs as a therapy for diseases caused by limited angiogenesis
and placental MSCs have been shown to stimulate blood vessel growth
in this small and accessible in vivo model (Kandel & Pittenger, 1999; 3.1.4.1. Peripheral artery disease (ischemia). Ischemia is a restriction of
Gruber et al., 2005; M. Y. Lee et al., 2009). Serum-deprived BM-MSCs blood supply to certain organs or tissues, caused by pathological
proved to cause significantly more chicken blood vessel growth than changes in the vascular system, such as vasoconstriction or thrombo-
normal BM-MSCs (Bianco et al., 2008). In addition, CAM angiogenesis sis (obstruction of a blood vessel). A common pathology associated
induced by EC-differentiated placental MSCs was inhibited by addition with ischemia is peripheral artery disease (PAD), referring to ob-
of antibodies against integrins α5 and β1 (M. Y. Lee et al., 2009). struction of the large limb arteries caused by atherosclerosis and in-
flammation. PAD can cause a wide variety of symptoms including
3.1.3.2. Mouse matrigel plug assay. The matrigel plug angiogenesis assay weakness and cramping in the muscles, chronic wounds and ulcers,
is an in vivo technique to detect newly formed blood vessels in a gel changes in color and temperature of the tissue, muscle atrophy and
plug which is transplanted into immunocompromised mice. Matrigel gangrene. Since effective pharmacological treatments do not exist
is a reconstituted basement membrane preparation that is extracted yet, amputation of the affected limb is needed to solve the unbear-
from the Engelbreth–Holm–Swarm mouse sarcoma, a tumor rich in able symptoms in a third of the cases. Owing to their angiogenic po-
ECM proteins such as laminin, collagen IV and entactin. Usually, a mix- tential, MSCs can be considered as an effective treatment option
ture of stem cells and matrigel is subcutaneously injected into mice and since restoration of the blood flow might eventually restore the func-
directly after injection, this matrigel solidifies and forms a plug. Several tionality of the affected extremity. PAD is mimicked in animal
weeks post injection, these plugs are removed and the number of blood models by blocking the blood flow by (temporary) ligation or resec-
vessels is assessed. tion of important hind limb arteries, such as the femoral artery,
mBM-MSCs dose-dependently induced angiogenesis in the resulting in ischemia of this extremity (Iwase et al., 2005).
mouse matrigel assay and the blood vessel sprouting was more pro- In a murine model of hindlimb ischemia induced by distal femoral
nounced than in matrigels incubated with recombinant FGF-2 or ligation, local intramuscular injection of GFP + BM-MSCs improved
VEGF (Al-Khaldi et al., 2003b; Estrada et al., 2009). A study by the limb perfusion, increased blood vessel density, reduced the inci-
research group of Verfaillie indicated that BM-MSCs are indeed able to dence of auto-amputation, attenuated muscle atrophy and signifi-
attract host blood vessels to subcutaneous matrigel transplants (mea- cantly improved limb function. These actions occurred without
sured 3 weeks post injection), but these tubes are leaky as evidenced observable MSC incorporation into the blood vessels, but were probably
by the presence of red blood cell pools in the matrigel (Roobrouck et al., caused by the paracrine actions of the MSCs as VEGF and FGF-2 levels
2011). Recently, subcutaneous DPSC-matrigel transplants in mice were were increased in the MSC-treated muscles and VEGF colocalized with
shown to induce more vessel formation than BM-MSC plugs. This the administered GFP + MSC (Kinnaird et al., 2004b). In a rat model
DPSC-induced neovascularization was inhibited by addition of soluble of hindlimb ischemia, treatment with BM-MSCs was found to be supe-
VEGF-receptor 1, which neutralizes VEGF-A (Janebodin et al., 2013). rior compared to administration of BM mononuclear cells. In contrast
to the aforementioned mouse study of Kinnaird et al., transplanted
3.1.3.3. Wound healing. Wound healing is a very complex multifactorial MSCs were considered to differentiate into EC as von Willebrand Factor
process involving the balanced interaction of inflammation, re- (vWF)-positive transplanted cells were found in the tissue 3 weeks
epithelialization, angiogenesis and deposition and remodeling of after transplantation (Iwase et al., 2005). Also, Al-Khaldi et al. (2003a)
the ECM. Poor wound healing is one of the most serious complications found that administered BM-MSCs expressed EC markers (such as
in patients who suffer from diabetes. As the incidence of diabetes CD34 and the EC marker factor VIII) 4 weeks after transplantation in a
mellitus is reaching pandemic proportions worldwide and current rat model of ischemia. Recently, a phase II clinical trial demonstrated
treatments only result in partial and temporal healing, there is a grow- that intra-arterial infusion of autologous bone marrow significantly in-
ing need for an effective therapy (Hocking & Gibran, 2010). creased both peripheral blood perfusion and the percentage of
Numerous researchers already described the beneficial effects of amputation-free patients with critical limb ischemia one year after the
BM-MSCs in wound healing (reviewed in Hocking & Gibran, 2010). treatment (Schiavetta et al., 2012). Moreover, intravenous injection of
For example, BM-MSC treatment of wounds in diabetic mice resulted hAD-MSCs improved blood perfusion in a mouse hind limb ischemia
in accelerated wound closure, increased re-epithelialization and model (Rehman et al., 2004; Moon et al., 2006). Implanted DiI-labeled
wound vascularity probably through the release of angiopoietin-1 hAD-MSCs also seemed to differentiate into EC as vWF/DiI double pos-
and VEGF (Y. Wu et al., 2007). In this case, the rate of MSC engraft- itive cells were detected within the muscle tissue. Nevertheless, the ac-
ment 28 days after injection was demonstrated to be extremely tual incorporation rate of these stem cells into the host vascular

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
8 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

structures was less than 1%, indicating that the paracrine effects of AD- 3.1.4.3. Cerebral ischemia/stroke. Stroke, also referred to as encephalic
MSCs principally account for the beneficial effect on hindlimb ischemia vascular accident, is a focal neurologic deficit caused by a disturbance
(Moon et al., 2006). In addition, intramuscular injection of a side popu- in the blood supply of the brain. The most recurrent type of stroke is is-
lation of porcine DPSCs (CD31−/CD146− DPSCs) increased blood flow chemic stroke, caused by a blockage of the blood circulation by a throm-
and capillary density in a murine hindlimb ischemia model. These bosis or an arterial embolism leading to a decrease in oxygen and
transplanted cells were present in close proximity to the vessel and in nutrient delivery to the brain. Depending on the affected brain area, is-
situ hybridization revealed that these cells expressed several angiogenic chemic stroke is associated with temporary or permanent loss of brain
proteins such as VEGF and MMP-3 (Iohara et al., 2008). tissue and function, resulting in a wide variety of devastating symptoms
comprising motor, sensory, balance and cognitive dysfunction. Follow-
ing MI, stroke is the most common cause of death worldwide and the
3.1.4.2. Myocardial infarction. According to the World Health Organiza- most common cause of adult disability, with 250,000 people in Europe
tion (WHO), ischemic heart disease is the leading cause of mortality in becoming disabled after their first stroke each year.
western societies. Myocardial infarction (MI) leads to heart dilatation Currently, tissue plasminogen activator (tPA) is the only available
or cardiac failure often resulting in significant disability or death. The ra- intervention to reduce the infarct size (Tissue plasminogen activator
tionale behind MSC-treatment for MI is to repair the damaged heart tis- for acute ischemic stroke. The National Institute of Neurological
sue by cardiomyocyte differentiation and by supplying growth factors Disorders and Stroke rt-PA Stroke Study Group, 1995). However, op-
that induce angiogenesis or stimulate resident cardiac stem cell migra- timal results are only observed if tPA is given within 90 min follow-
tion and commitment to cardiomyocytes (Ranganath et al., 2012). In an- ing the primary insult (Hacke et al., 2004), thereby strongly
imal models, MI is caused by a ligation of one of the coronary arteries, reducing its therapeutic capacities. This narrow therapeutic window
such as the left artery descending and left circumflex artery. in combination with many contraindications makes tPA therapy only
Abundant evidence demonstrates the therapeutic benefits of available to about 5% of stroke victims (Brown, 2005). Therefore,
bone marrow (Dai et al., 2005), adipose (Miyahara et al., 2006), there is still a high demand for novel treatment options that aim at
pulp (Gandia et al., 2008) and even amniotic-derived MSCs (Kim long-term recovery of the affected brain area. MSC therapy might
et al., 2013) in various mouse or rat models of MI. Transplantation offer an alternative approach to induce functional recovery by
of these cells significantly improved ventricular function, which is means of improving revascularization, affecting neural plasticity or
associated with the induction of myogenesis and angiogenesis. AD- inducing cell replacement near the stroke area. Over the years,
MSCs transplanted as a monolayer, improved cardiac function after many animal stroke models have been developed in order to simu-
MI through the secretion of HGF and VEGF (Miyahara et al., 2006), late what happens during stroke in humans. Among them, the tran-
while intramyocardial injection of amniotic MSCs did increase the sient middle cerebral artery occlusion (tMCAO) model is by far the
levels not only of VEGF but also of Ang-1 in the affected heart tissue most frequently used. The main advantage of the tMCAO model lies
(Kim et al., 2013). In a hamster model of cardiomyopathy, injection in its physiological relevance being the occlusion of one of the main
of porcine BM-MSCs into the hamstring skeletal muscles improved cerebral arteries. Other models include a photochemical, autologous
cardiac repair. In this model, MSCs secreted multiple IL-6-like cyto- thromboembolic and the more recently developed endothelin-1
kines, which induced activation of the JAK/STAT pathway and in- model (Kudo et al., 1982; De Ryck et al., 1989; Windle et al., 2006).
creased the levels of VEGF and HGF in the cardiac tissue. The Numerous reports demonstrated that delivery of BM-MSCs by a
reparative impact of MSCs on heart failure was reversed by addition wide variety of routes (intravenous, intra-arterial and intracerebral)
of the JAK/STAT pathway inhibitor WP1066 (Shabbir et al., 2010). reduced infarct size and improved the functional outcome in ische-
Furthermore, a recent meta-analysis of large animal models (such mic stroke (Banerjee et al., 2012; Ma et al., 2013). Although the ini-
as pigs, dogs and sheep) of MI indicated that the transplantation of tial idea was that MSCs would enhance recovery after stroke by
BM-MSCs leads to a significant improvement of left ventricle ejection transdifferentiation into neurons and glial cells, it is now suggested
fraction. In addition, a trend towards greater advantage in the case of that MSCs have a beneficial impact primarily through the inhibition
administration of MSCs when compared with BM-mononuclear cells of apoptosis, neuroprotection and enhancement of endogenous
was reported (van der Spoel et al., 2011). In all available documenta- neurogenesis and angiogenesis (Zhang et al., 2009; Banerjee et al.,
tions, the ability of the engrafted MSCs to transdifferentiate/fuse into 2012). Indeed, multiple studies describe the production of angiogenic
cardiomyocytes was found to be extremely scarce (Dai et al., 2005; factors by MSCs indicating a key role for these factors in the improved
Gandia et al., 2008; van der Spoel et al., 2011). In contrast to the re- recovery seen in transplanted stroke animals (Kurozumi et al., 2005).
ports of Otsu et al., who claimed that high numbers of administrated Chen et al. (2003) demonstrated in a tMCAO rat model of ischemic
MSC can cause toxicity, the meta-analysis of MI models of large ani- stroke, that intravenous injection of human BM-MSCs enhanced angio-
mals indicated that the best results were obtained with the highest genesis in the ischemic brain, probably by increasing the endogenous
amount of injected cells (Otsu et al., 2009; van der Spoel et al., VEGF and VEGFR2 expression. Another research group reported a sig-
2011). This contradiction might be explained by the differences in nificant upregulation of Ang-1 and Ang-2 mRNAs in BM-MSC treated
animal model or by alternative ways of administration. Indeed, as in rats using the same animal model (Ma et al., 2013). Moreover, MSCs iso-
MI, cells are mostly injected intramyocardially or in the intracoronary lated from the peripheral blood reduced lesion volume, improved re-
artery (and immediately diluted into the blood flow or a larger tissue gional cerebral blood flow and stimulated functional improvement
area), whereas in the study of Otsu et al., MSCs were locally applied. after transplantation (Ukai et al., 2007). A recent study compared the
Currently, the safety, dose and impact associated with MSC- beneficial effect of intravenous administration of BM-MSCs and AD-
treatment for MI in humans are intensively studied and already a few MSCs in a rat stroke model. AD-MSCs were found to be as effective as
clinical trials have been published (Tongers et al., 2011; Ranganath BM-MSCs in promoting functional recovery, reducing cell death as
et al., 2012). For example, a double-blinded, randomized clinical trial well as increasing cellular proliferation, neurogenesis, and the expres-
by Hare et al. (2009) indicated that intravenous allogenic BM-MSC sion of angiogenesis markers (such as VEGF) at 14 days post-
treatment is safe and that cell-treated patients had improved outcomes infarction (Gutierrez-Fernandez et al., 2013). Even the intraperitoneal
with regard to cardiac arrhythmias, pulmonary function and left injection of a cell-free extract derived from MSCs significantly de-
ventricular function compared to the placebo group. In addition, S. L. creased the ischemic volume and improved motor function (D. Jeon
Chen et al. (2004) showed that intracoronary application of BM-MSCs et al., 2013).
also resulted in improved left ventricular function and exercise This extensive preclinical work on MSC treatment in stroke, encour-
capability. aged the study of these promising cells in phase I/II clinical trials

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 9

(Banerjee et al., 2012). A small study with 5 patients that intravenously Table 2). MSC commitment to EC is mostly demonstrated by the up-
received autologous BM-MSCs, showed that the MSC-treated group had regulation of typical EC surface molecules such as CD31,CD34, VEGF-
the tendency to have a better functional outcome, although this differ- receptor1 (VEGFR1), VEGFR2 and vWF. As change in surface marker
ence was not statistically significant (Bang et al., 2005). The same profile is insufficient to evidence differentiation, functional tests are
authors published the results of a long-term follow-up study of intrave- also performed, including in vitro tube formation on matrigel and up-
nous autologous BM-MSC transplantation in patients with ischemic take of acetylated-low density lipoproteins (although macrophages
stroke. These experiments revealed that the BM-MSC treatment im- and pericytes also internalize these molecules) (Voyta et al., 1984).
proved functional outcome and survival, even 5 years after MSC admin- Based on these commonly used in vitro assays, it might be inaccurate
istration (J. S. Lee et al., 2010). A recent non-blinded trial (in 12 patients) to designate these differentiated MSCs as fully mature and functional
with autoserum-expanded autologous BM-MSCs, indicated that intra- EC. It is therefore more correct to define these cells as EC-like cells.
venous administration of the cells 36–133 days after stroke was not Although the in vitro differentiation approaches listed in Table 2
only safe and feasible, but also could reduce infarct size by 20% at one demonstrate indications for EC-like characteristics such as EC mark-
week post-infusion (Honmou et al., 2011). Although these clinical trials er expression, matrigel tube formation and Ac-LDL uptake, there is
had a small sample size and the study of Honmou et al. was non-blinded almost no literature available which shows morphological and ultra-
and did not exclude placebo effects, all investigations demonstrated the structural evidence of EC transdifferentiation. For example, the pres-
potential benefit of MSC-treatment in stroke and justified additional ence of typical EC characteristics such as Weibel–Palade bodies and
blinded, placebo-controlled studies. tight junctions, which could be demonstrated by transmission elec-
tron microscopy, have never been reported for EC-directed MSCs
3.1.4.4. Ambiguous role of MSC in tumor formation. As MSCs are able to in- (except in Jazayeri et al., 2008).
duce blood vessel formation in vivo and since tumor growth and metas- Skewing MSCs to EC differentiation has been commonly achieved
tases are angiogenesis-dependent, the contribution of MSC to cancer by using VEGF. In one of the earliest attempts of EC differentiation, it
development has been the subject of intense investigations. Although was demonstrated that confluent hBM-MSCs grown in medium with
several studies suggest that MSCs have a tumor-suppressive function 2% FCS and 50 ng/ml VEGF for 7 days, displayed a substantial increase in
(Khakoo et al., 2006; Otsu et al., 2009), other reports indicate that EC surface markers such as VEGFR1, VEGFR2, VE-Cadherin, VCAM-1 and
MSCs promote tumor growth in various cancer models such as adeno- vWF. Moreover, when incubated on matrigel in vitro, treated BM-MSCs
carcinoma (E. S. Jeon et al., 2010), melanoma (Djouad et al., 2003) and formed characteristic capillary-like structures. These differentiation ex-
breast cancer (Galie et al., 2008) (reviewed in Klopp et al., 2011 and periments were only performed with cells at low passage number
Cuiffo & Karnoub, 2012). These contradictory results might be explained (below 5 passages) (Oswald et al., 2004). Furthermore, hBM-MSCs in-
by variations in MSC source, isolation method of MSCs, tumor cell type, cubated in medium with 5% FCS, IGF and VEGF were immunopositive
contamination of the applied MSCs with tumor cells, and differences in for CD31, vWF, Tie2, VCAM1 and VE-cadherin. Electron microscopic
timing, dose and mode (systemic injection versus local application into analysis showed the presence of typical EC morphological features
the tumor) of MSC administration (Klopp et al., 2011). Although the including Weibel–Palade bodies, tight junctions and caveolae
molecular mechanisms behind the tumor-promoting capacities of (Jazayeri et al., 2008). In addition, multipotent stem cells isolated
MSCs are still not fully understood, several papers indicate that MSCs in- from the placenta (hPMSCs) showed an increased expression of EC
deed stimulate tumor growth by enhancing angiogenic processes (Jeon markers CD31, CD34, VEGFR1 and VEGFR2 after incubation with
et al., 2010; Liu et al., 2011; Suzuki et al., 2011; Cuiffo & Karnoub, 2012). 50 ng/ml VEGF (M. Y. Lee et al., 2009). Recently, Bento et al. de-
For example, Y. Liu et al. (2011) demonstrated that VEGF or HIF-1α scribed that MSCs derived from exfoliated teeth (the so-called milk
siRNA effectively reduced MSC-favored colon tumor growth in vivo. teeth), differentiated into VEGFR2/CD31 positive EC-like cells. Inhibi-
Other proposed mechanisms of promoting tumor progression include tion of ERK signaling with siRNA against MEK1 or with the chemical
malignant transformation of MSCs, protection against apoptosis, vascu- compound U0126 completely abrogated this differentiation, unveiling
lar support and immunomodulation. The tumor-suppressing capacities that the VEGF/MEK1/ERK signaling transduction cascade is key in EC
of MSCs, on the other hand, are attributed to induction of apoptosis and commitment of this type of MSC (Bento et al., 2013). Furthermore,
inflammation, inhibition of angiogenesis and alteration of the cell cycle hBM-MSCs cultured for 3 weeks in ‘endothelial growth medium-2’
progression (Klopp et al., 2011). (EGM-2, which contains VEGF, EGF, FGF-2, IGF-1, hydrocortisone, hepa-
Another intriguing capacity of MSCs is that they are able to home to rin, ascorbic acid and 2% FCS), displayed significantly elevated expres-
developing tumors with great affinity. They do selectively home not sion of CD31, CD144, VEGFR2, CD105 and CD34 and were also able to
only to solid tumors, but also to metastases far removed from the prima- form tubes on matrigel in vitro (J. W. Liu et al., 2007). In contrast,
ry site of the tumor (Nakamizo et al., 2005; Dwyer et al., 2010). Accord- Roobrouck et al. (2011) demonstrated that incubation of hBM-MSC in
ingly, they have been explored as delivery vehicles of anticancer (gene) basal medium containing VEGF significantly increased mRNA expres-
therapy (see Section 4). Before MSCs find their way to the clinic as a sion of CD34, VEGFR1, and VEGFR2 but not of Tie-2 and vWF and the
therapeutic tool in the fight against cancer, the exact conditions under mRNA level of CD31 was even decreased. These hBM-MSC did not
which MSCs are able to promote tumor growth have yet to be fully elu- form a tubular network when plated onto matrigel in vitro, thereby
cidated. Nevertheless, a recent systemic meta-analysis of clinical trials failing the functional test of full EC differentiation (Roobrouck
with MSCs showed that, so far, no de novo malignancies in patients et al., 2011).
have been reported in over 1000 patients treated with MSCs for a vari- Several other recent reports question the role of VEGF in EC com-
ety of disorders (Lalu et al., 2012). mitment of MSCs. For example, Fan et al. (2011) demonstrated that
hBM-MSCs cultured in the presence of 20, 40 or 80 ng/ml of VEGF
3.2. MSC transdifferentiation towards endothelial cells (EC) had no increase in CD31, vWF or VEGFR2. Furthermore, Galas and
Liu suggested that VEGF treatment does not accelerate EC differentia-
3.2.1. EC differentiation in vitro tion of MSCs: confluent BM-MSCs incubated with EGM-2 had increased
As previously mentioned, the differentiation of MSCs towards EC, PECAM and vWF mRNA levels, VE-cadherin, VEGFR2 and vWF protein
both in vitro and after transplantation, remains a controversial as- expression and Ac-LDL uptake compared to untreated (and proliferat-
pect. Supported by the idea of the tremendous plasticity of MSCs, ing) BM-MSCs. Addition of VEGF (at 50 or 100 ng/ml) to this EGM-2 me-
as they were proposed to have the ability to transdifferentiate into dium did not further accelerate EC marker expression. These data
other cell types than mesodermal lineage cells, numerous efforts suggest that either a component of EGM-2 or the confluent MSC cell–
have been made to differentiate MSC to EC in vitro (summarized in cell contacts induced the EC-specification of BM-MSC (Galas & Liu,

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
10
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate

Table 2
Media used to induce EC transdifferentiation of MSCs in vitro.

Reference MSC type Incubation Differentiation medium Resulting marker expression Functional evidence of EC
time transformation

Al-Kaldi et al., 2003b mBM-MSC 14 days 10% FCS, 50 ng/ml VEGF CD31 Matrigel tube formation
Alviano et al., 2007 hAM-MSC 7 days 2% FCS and 50 ng/ml VEGF VEGFR1,VEGFR2, vWF, CD34, ICAM-1 Matrigel tube formation

A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx


Bai et al., 2010 rBM-MSC 7 days VEGF alone CD31, Factor VII, VEGFR2, tPA ND
Bai et al., 2010 rBM-MSC 8 days Shear stress (24 h) + VEGF (7 days) CD31, Factor VII, VEGFR2, tPA ND
Bento et al., 2013 hSHED 28 days EGM-2 + 50 ng/ml VEGF VEGFR2, CD31 Elongated morphology
Chen et al., 2009 BM-MSC 12 days 5% FCS + 100 ng/ml VEGF + 50 ng/ml EGF + 1 μg/ml VEGFR2, vWF, VE cadherin Matrigel tube formation and LDL uptake
hydrocortisone
Chen et al., 2009 WJ-MSC 12 days 5% FCS + 100 ng/ml VEGF + 50 ng/ml EGF + 1 μg/ml VEGFR2, vWF, VE cadherin Matrigel tube formation and LDL uptake
hydrocortisone
Chung et al., 2009 hBM-MSC 21 days EGF + VEGF + hydrocortisone VEGF, vWF, VE-cadherin Matrigel tube formation
Fischer et al., 2009 hAD-MSC 18 days ECGS (14 days) + shear stress (4 days) CD31 LDL uptake
Gang et al., 2006 UM-BMSC 21 days EGF + VEGF + hydrocortisone VEGFR1, VEGFR2, VE-Cadherin, vWF, VCAM-1, Tie-1 and Matrigel tube formation and LDL uptake
Tie-2
Iohara et al., 2008 DPSC 10 days On matrigel, EGM-2 CEACAM-1, occludin, CD146 (mRNA) Matrigel tube formation
Iohara et al., 2008 DPSC 3 days EBM-2 + 2% serum + 10 ng/ml VEGF + 10 ng/ml FGF-2 vWF, VE-cadherin, CD31 Matrigel tube formation and LDL uptake
Jazayeri et al., 2008 BM-MSC 5 days 5% FCS, 50 ng/ml VEGF and 20 ng/ml IGF-1 CD31, vWF, VEGFR2, VEGFR1, Tie2, VCAM1 and VE-cadherin Matrigel tube formation
Karbanova et al., 2011 hDPSC 10 days 10% FCS + 1% ITS + 20 ng/ml VEGF 10% of cells, upregulation of CD31, CD34, vWF ND
Konno et al., 2010 mAD-MSC 12 days EGM-2 BulletKit (without the FGF-2) + ITS + 10 ng/ml FGF-2 CD34, Tie-2, VEGFR2, VE-cadherin Matrigel tube formation and LDL uptake
M.Y. Lee et al., 2009 Placental MSC 21 days EGM-2 + 2% FCS + 50 ng/ml VEGF CD31, CD34, VEGFR-1 and VEGFR2, vWF and VE-cadherin ND
Liu et al., 2007 hBM-MSC 21 days EGM-2 + 2% FCS CD31, CD144, VEGFR2, CD105 and CD34 Matrigel tube formation and LDL uptake
Marchionni et al., 2009 hDPSC 7 days 2% FCS and 50 ng/ml VEGF vWF,ICAM-1,CD34 Matrigel tube formation
Moon et al., 2006 hAD-MSC 2 days On matrigel, EGM-2 + 50 ng/ml VEGF + 10 ng/ml FGF-2 vWF ND
Oswald et al., 2004 hBM-MSC 7 days 2% FCS + 50 ng/ml VEGF VEGFR1, VEGFR2, VE-Cadherin, VCAM-1 and vWF Matrigel tube formation
Pankajakshan et al., 2013 Porcine BM-MSC 10 days EGM-2 + 50 ng/ml VEGF vWF, VE-cadherin, CD31 Matrigel tube formation and LDL uptake
Portalska et al., 2013 hBM-MSC 11 days EGM-2 + shear stress (10 days) + 1 day on matrigel VEGFR1, vWF, CD31 LDL uptake
Roobrouck et al., 2011 hBM-MSC 14 days Basal medium + 100 ng/ml VEGF CD34, VEGFR1, and VEGFR2 but not of Tie-2 and vWF Failure matrigel tube formation
Takahashi et al., 2010 AD-MSC 7 days EGM-2 + 5% FCS VEGFR1 and Ang-1 not VEGFR2, VE-cadherin nor CD31 ND
Whyte et al., 2011 hBM-MSC 14 or 28 days Basal medium VE-cadherin, PECAM-1, vWF, and VEGFR1 Matrigel tube formation and LDL uptake
Wu et al., 2005 rBM-MSC 5 days Co-culture with rabbit EC VEGFR1, no vWF ND
Xu et al., 2008 rBM-MSC 7 days 2% FCS + 0.5 μM simvastatin CD31, vWF, VE-cadherin, VEGFR1, VEGFR2 Matrigel tube formation
Zhang et al., 2008 hBM-MSC 21 days 5% FCS + 50 ng/ml VEGF vWF, VEGFR1,VEGFR2, Tie-1, Tie-2 Matrigel tube formation and LDL uptake
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 11

2014). This is in agreement with a study of Whyte et al., who could include mechanical stimuli (Maul et al., 2011), shear stress (Fischer
prove that culture density of BM-MSC regulates EC commitment. et al., 2009; Bai et al., 2010) and seeding the cells on elastic nanofiber
Human BM-MSCs incubated at high densities (10,000 cells/cm2) for hydrogels (Wingate et al., 2012) or on 3-dimensional matrices
14 or 28 days obtained an EC-like phenotype expressing VE-cadherin, (Valarmathi et al., 2009; Zhang et al., 2010). For example, placenta-
PECAM-1, vWF, and VEGFR1, displaying Ac-LDL-uptake and forming derived MSC subjected to shear stress for 24 h showed a significant
tubes in the matrigel assay. These high density plated hBM-MSCs had increase in vWF and PECAM-1 expression, LDL-uptake and matrigel
an increased expression of VEGF. This autocrine VEGF secretion tube formation (Wu et al., 2008). Despite the characterization of sev-
plays a role in the EC differentiation of hBM-MSCs as inhibition of eral key actors for EC commitment such as the Notch signal cascade,
VEGF with neutralizing antibodies resulted in a decrease of VEGFR1 additional cues for functional and homogenous EC differentiation
or VE-cadherin. Nevertheless, incubation of low density plated BM- must be identified to fully understand the underlying molecular
MSC with VEGF did not induce EC marker expression. Taken togeth- mechanisms. Successful homogenous EC differentiation of MSCs
er, these data indicate that VEGF alone is not sufficient to initiate EC could be of great use for the development of functional blood vessels
differentiation, but this growth factor enhances EC commitment in in the field of tissue engineering.
high cell density cultures (Whyte et al., 2011).
These contradictory findings on the role of VEGF in EC differentiation 3.2.2. EC differentiation in vivo
of MSCs might be caused by the heterogeneity of MSC, differences in As described above, the rate of engraftment of MSCs remains very
MSC isolation, tissue sources and species, differences in assays used to low in most settings. Therefore, it is assumed that the angiogenic effect
demonstrate EC characteristics and artifacts associated with culturing of MSCs is predominantly caused by their paracrine actions rather than
techniques. It might also be worth to mention that not in all cases and their EC transdifferentiation potential. For example, Al-Khaldi et al.
reports MSCs have VEGF-receptors (Whyte et al., 2011). In addition showed that by using LacZ transgenic BM-MSCs in the murine matrigel
to VEGFR1 and VEGFR2, VEGF is able to bind various co-receptors in- plug assay over 99% of the generated blood vessels originated from
cluding neuropilins and heparan sulfate proteoglycans (Ruiz de host-derived EC and that transdifferentiation of BM-MSCs towards EC
Almodovar et al., 2009). It is possible that, depending on the receptor in that setting was very rare. Only 10% of injected BM-MSCs were
profile, MSCs respond differently to soluble VEGF in the medium. found in the close proximity of- or in blood vessels (Al-Khaldi et al.,
Xu et al. demonstrated that the Notch-signaling pathway plays a key 2003b). Another striking example is a study with GFP + BM-MSC in
role in MSC commitment to EC. Rat BM-MSCs incubated for 7 days in 2% the treatment of wounds in diabetic mice. The rate of MSC engraftment
FCS and 0.5 μM simvastatin (a commonly used cholesterol-lowering 28 days post-injection was extremely low (less than 1%) and no partic-
agent), had increased surface expression of CD31, vWF, VE-cadherin, ipation of the transplanted GFP + BM-MSCs into the host vasculature
VEGFR1 and VEGFR2 and showed tube formation on the matrigel was observed (Wu et al., 2007). Although the low engraftment of
assay. Targeted inhibition of Notch signaling with Notch 1 siRNA MSCs in most settings and the lack of incorporation into host blood
suppressed this EC differentiation (Xu et al., 2009). Moreover, the vessels is abundantly described, it may not be ruled out that in par-
aforementioned study of Whyte et al. also showed the fundamental ticular settings or time frames, the EC differentiation of MSC exists.
role of Notch signaling in EC differentiation. High density plated For example, a recent study by Yue, where rBM-MSCs were injected
hBM-MSCs were immunopositive for Notch transcription factor HES-1 in a rat vein grafting model, showed that the injected DAPI labeled
and the Notch receptors 1, 2 and 3 during EC specification. Knockdown BM-MSCs differentiated into EC 2 weeks after transplantation as
of the Notch receptors with specific siRNA or addition of the chemical they were capable of adhering to the injured vascular wall and
Notch inhibitor DAPT abrogated the EC commitment of these hBM- showed immunopositivity for CD31 and endothelial nitric oxide syn-
MSCs. In addition, incubation of hBM-MSCs seeded at low density thase (Yue et al., 2008).
with EDTA (which activates Notch) for 24 h resulted in an upregulation Although the evidence of full EC differentiation of MSCs in vivo is
of vWF and VEGFR1 (Whyte et al., 2011). In addition, it was shown that debatable, preconditioning MSCs in the so-called EC-differentiation
the Notch-pathway in cross-talk with VEGF signaling, is also involved in medium could be useful as this pretreatment might transform the
the arterial-venous fate of EC-differentiated BM-MSCs. Treatment of MSCs towards a more ‘angiogenic’ cell type. For example, placental
confluent BM-MSC with 50 ng/ml VEGF for 14 days increased expres- MSCs incubated with 50 ng/ml VEGF displayed higher angiogenic activ-
sion of the venous marker EphB4, while at the concentration of ity than normal placental MSCs in the CAM assay (M. Y. Lee et al., 2009).
100 ng/ml, venous ephrin B4 expression decreased while the arterial Also Whyte et al. (2011) demonstrated that high density plated BM-
markers ephrin B2, Dll4 and Notch4 were strongly upregulated. This MSCs (which differentiate into EC-like cells) were found to induce sig-
VEGF-dependent arteriogenesis was blocked by inhibition of the nificantly more blood vessels in this in vivo assay. In addition, AD-
Notch pathway, resulting in a shift from arterial to venous EC fate MSCs incubated with EGM-2 for 7 days, secreted higher amounts of
(G. Zhang et al., 2008). A recent study by Chung et al. showed that angiopoietin-1, promoted HUVEC migration and significantly sup-
during endothelial differentiation, the expression pattern of 4 HOX pressed neointimal formation in a wire injury model of the rat femoral
genes changed significantly: incubation of hBM-MSCs with EGF and artery compared to control AD-MSCs (Takahashi et al., 2010).
VEGF for 21 days resulted in a dramatic increase of HOXA7 and
HOXB3 while HOXA3 and HOXB13 decreased. Although the exact 4. MSC as vehicles for targeted
role of each HOX gene in EC differentiation remains to be elucidated, (anti-)angiogenic drug and gene delivery
these data indicate that these HOX genes might be involved in EC
commitment of MSCs (Chung et al., 2009). Although MSCs naturally possess an enormous therapeutic poten-
Furthermore, Lozito et al. described that hBM-MSCs expressed EC tial, gene therapy is applied to modify MSCs to further improve their ef-
marker CD31 when directly co-cultured with EC. As this effect was not ficacy and even extend the spectrum of diseases for which MSCs could
seen in the indirect co-culture systems but could be mimicked by grow- provide a successful cure. MSCs can be readily transduced with all the
ing the BM-MSC on decellularized EC matrices, this indicated that not clinically available viral vector systems including those based upon ret-
the soluble factors but EC-matrix acts as a critical regulator of BM-MSC rovirus, lentivirus, adenovirus and adeno-associated virus. Viral trans-
commitment to EC (Lozito et al., 2009). duction of MSCs is relatively easy and results in efficient production of
Classically, the control of stem cell fate has been mainly assigned cytoplasmic, membrane-bound and secreted protein products. As viral
to molecular mediators such as growth factors. New data indicate vectors are able to integrate into the host genome, this method could re-
that in addition to soluble factors, physical cues can also provide a sult in a long-term gene expression in vivo (Porada & Almeida-Porada,
successful method to induce MSC EC differentiation. Such tools 2010). Numerous animal studies have reported the success of MSCs as

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
12 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

a gene delivery vehicle in a wide variety of pathologies in vivo. How- 5. Therapeutic expectations and challenges
ever, many clinical trials in which viral vectors were used, have been
terminated since application of these vectors had induced adverse ef- Although MSCs originally captured the attention as a therapeutic
fects including toxicities, immune- and oncogenicity (Marshall, 1999; agent for tissue repair due to their presumed in vivo plasticity, there is
Hacein-Bey-Abina et al., 2003; Santos et al., 2011). Therefore, an in- now emerging evidence suggesting that immunomodulatory and para-
creasing number of non-viral vectors are currently being developed crine actions predominantly contribute to their broad therapeutic effi-
such as calcium phosphate, liposomes, niosomes and nanoparticles. As cacy. The last few years, intensive research has led to the identification
the gene expression by such a non-viral system is transient and the of a multitude of bioactive angiogenic molecules such as VEGF, FGF-2
transfection efficiency is relatively low, current studies on these gene and MCP-1 in the conditioned medium of MSCs. Additionally, MSCs
delivery methods are still limited to in vitro settings (Hu et al., 2010). and the broad repertoire of angiogenic mediators they secrete, have
been shown to induce angiogenesis and thereby enhancing tissue repair
4.1. MSCs designed as carriers for pro-angiogenic factors and functional outcome in a wide variety of pathologies associated with
insufficient angiogenesis.
MSCs genetically engineered to secrete cytokines and other angio- Some unresolved issues, however, still impede these promising cells
genic factors have been used in a wide variety of animal models. For ex- to become a realistic treatment option for the future. The absence of a
ample, using a lentiviral vector, Xu and coworkers designed BM-MSCs unique marker of MSCs as well as the heterogeneity/inconsistency of
overexpressing Ang-1, an angiogenic protein inducing endothelial sur- MSCs isolation and expansion techniques are still major hurdles,
vival and vascular stabilization. In an in vivo mouse model, the LPS- which makes it a huge struggle to compare preclinical and clinical re-
induced lung injury was significantly alleviated in the group treated sults from different research groups. Furthermore, as in most settings
with BM-MSCs carrying Ang1, compared with groups treated with the rate of engraftment of MSCs is extremely low, the question arises
BM-MSCs or Ang1 alone (Xu et al., 2008). In a MCAO-induced rat stroke concerning the fate of the administered MSCs. There is the possibility
model, animals receiving FGF-2-modified BM-MSCs demonstrated sig- that they home to other tissues or undergo necrosis or apoptosis. Ad-
nificant functional recovery compared with rats treated with normal vances in cell labeling and imaging techniques are essential to clarify
BM-MSCs (Ikeda et al., 2005). In addition, both HGF- and VEGF- this issue. In addition, recent preclinical and clinical studies have now
overexpressing mBM-MSC treatments resulted in smaller scar sizes, in- begun to demonstrate that MSC treatment is safe and feasible, although
creased angiogenesis and better preserved left ventricular function the long-term biosafety is still an essential concern and warrants further
when compared with MSCs transfected with empty vector in a mouse long-term follow-up of the patients. Another key obstacle before MSC-
model of MI (Deuse et al., 2009). based treatment can enter the clinic as a standard therapy is the current
inefficient protocol of MSC expansion. Since the existing culture tech-
4.2. MSCs engineered for anti-cancer treatments nology (serial passaging in plastic flasks) is still time-consuming and
costly, there is an urgent need to develop large scale-up production of
As MSCs are able to migrate to the tumor side (cf. Section 3.1.4.4), MSCs in serum-free conditions (for example in bioreactors).
they represent a very powerful and unique vehicle to selectively de- Despite the increasing number of studies indicating the beneficial
liver anti-cancer gene products. The delivery of interleukins via effects of MSC treatment, optimization of these therapies remains a
MSCs has been explored in order to improve the anti-cancer surveil- huge challenge. Transduction of angiogenic factors in MSCs or pre-
lance by activating immune cells such as cytotoxic lymphocytes and conditioning of MSCs by hypoxia or certain chemical agents has
natural killer cells (Shah, 2012). For example, interleukin-12 secret- been shown to boost the angiogenic effects of MSCs, which might
ing MSCs reduced tumor growth in renal cell and cervical tumor mice be a valuable strategy to maximize their clinical potential (although
models and prevented metastasis in mice bearing pre-established there might be some biosafety concerns, which also warrant in-
metastases of melanoma, hepatoma and breast tumors (X. Chen depth examination). Also, besides adapting the treatment protocol
et al., 2008; Shah, 2012). Also treatment of MSCs containing anti- to multiple administrations, the identification and use of a certain su-
proliferating and proapoptotic genes such as IFN-α, IFN-β and perior subpopulation of MSCs could ameliorate existing MSC therapies.
TRAIL displayed a significant antitumor activity in several mouse Since the evidence arises that the paracrine mechanisms mostly ac-
models. In addition, gene therapy is applied to specific target the count for the beneficial effects of MSCs, the use of the MSC secretome
tumor-associated angiogenesis. In a recent study, it was shown that instead of cellular therapy requires further exploration in clinical set-
single administration of stem cells overexpressing thrombospondin tings. Since the application of autologous (or allogeneic) MSCs requires
markedly reduced tumor vessel-density and inhibited tumor- the time-consuming expansion to sufficient numbers, the use of an allo-
progression in mice bearing human gliomas (van Eekelen et al., geneic MSC secretome could overcome this problem. It could be pre-
2010). Systemic administration of MSCs designed to express NK4, pared in large amounts in advance and is then directly available as an
an antagonist of HGF, efficiently decreased metastases by blocking an- ‘off-the-shelf’ product. This may be especially helpful in clinical settings
giogenesis and lymphangiogenesis in mice bearing lung metastases where immediate treatment is desired, for example after MI or acute
(Kanehira et al., 2007). Another strategy in the fight against cancer is stroke. This secretome-approach could lead to the development of spe-
the so-called prodrug therapy, which involves delivery of ‘suicide cialized stem cell products, designed to contain specific trophic factors
genes’ encoding enzymes that are able to convert nontoxic prodrugs to suit the specific subtype of disorder. Nevertheless, clear understand-
into active antitumoral antimetabolites. Examples of such genes that ing of the in vivo MSC secretome and its potential effect on the microen-
are currently investigated in clinical trials are cytosine deaminase and vironment remain crucial before further (pre)clinical studies can be
herpes simplex virus thymidine kinase which respectively activate 5- performed.
fluorocytosine and Ganciclovir (Menon et al., 2008; Shah, 2012). Com- In conclusion, over the last decade, tremendous advances have been
bined therapy of cytosine deaminase expressing AD-MSCs and the made in the understanding of the angiogenic role of MSCs. This is evi-
prodrug 5-fluorocytosine resulted in a significant production of the che- denced by the successful application and neovascularization in a wide
motherapeutic 5-fluorouracil and inhibited tumor formation in mouse variety of in vivo settings of MI, stroke, ischemia and wound healing,
models of melanoma (Kucerova et al., 2008). In addition, BM-MSCs resulting in an increasing optimism of both basic scientists and clini-
overexpressing herpes simplex virus thymidine kinase injected into cians. Though the road remains long and hard before these puzzling
the tumor or the vicinity of the tumor were shown to successfully re- cells can become a realistic everyday therapeutic option, routine clinical
duce tumor volume in rats bearing glioma and treated with ganciclovir application of MSCs for a wide variety of pathologies associated with
(Miletic et al., 2007). limited angiogenesis is an exciting prospect.

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 13

Conflicts of interest Chen, S. L., Fang, W. W., Ye, F., Liu, Y. H., Qian, J., Shan, S. J., et al. (2004). Effect on left ven-
tricular function of intracoronary transplantation of autologous bone marrow mesen-
chymal stem cell in patients with acute myocardial infarction. Am J Cardiol 94, 92–95.
The authors declare that there are no conflicts of interest Chen, X., Lin, X., Zhao, J., Shi, W., Zhang, H., Wang, Y., et al. (2008). A tumor-selective
biotherapy with prolonged impact on established metastases based on cytokine
gene-engineered MSCs. Mol Ther 16, 749–756.
Acknowledgments Chen, L., Tredget, E. E., Wu, P. Y., & Wu, Y. (2008). Paracrine factors of mesenchymal stem
cells recruit macrophages and endothelial lineage cells and enhance wound healing.
PLoS One 3, e1886.
The preparation of the manuscript was supported by the Re- Chen, J., Zhang, Z. G., Li, Y., Wang, L., Xu, Y. X., Gautam, S.C., et al. (2003). Intravenous ad-
search Foundation—Flanders (‘Fonds Wetenschappelijk onderzoek ministration of human bone marrow stromal cells induces angiogenesis in the ische-
Vlaanderen—FWO’, grant Nr. 1.5.060.13N). Petra Hilkens benefits mic boundary zone after stroke in rats. Circ Res 92, 692–699.
Chen, M. Y., Lie, P. C., Li, Z. L., & Wei, X. (2009). Endothelial differentiation of Wharton's
from a PhD scholarship of the FWO and Annelies Bronckaers is a
jelly-derived mesenchymal stem cells in comparison with bone marrow-derived
postdoctoral fellow of the FWO. The FWO was not involved in mesenchymal stem cells. Exp Hematol 37, 629–640.
study design collection, analysis and interpretation of the data, in Choi, M., Lee, H. S., Naidansaren, P., Kim, H. K., O, E., Cha, J. H., et al. (2013). Proangiogenic
features of Wharton's jelly-derived mesenchymal stromal/stem cells and their ability
writing the report and in the decision to submit the paper for pub-
to form functional vessels. Int J Biochem Cell Biol 45, 560–570.
lication. We would also like to thank Nathalie Geurts for critical re- Chung, N., Jee, B. K., Chae, S. W., Jeon, Y. W., Lee, K. H., & Rha, H. K. (2009). HOX gene anal-
vision of the manuscript. ysis of endothelial cell differentiation in human bone marrow-derived mesenchymal
stem cells. Mol Biol Rep 36, 227–235.
Clark, B. R., & Keating, A. (1995). Biology of bone marrow stroma. Ann N Y Acad Sci 770,
References 70–78.
Crisan, M., Yap, S., Casteilla, L., Chen, C. W., Corselli, M., Park, T. S., et al. (2008). A
Al-Khaldi, A., Al-Sabti, H., Galipeau, J., & Lachapelle, K. (2003). Therapeutic angiogenesis perivascular origin for mesenchymal stem cells in multiple human organs. Cell
using autologous bone marrow stromal cells: improved blood flow in a chronic Stem Cell 3, 301–313.
limb ischemia model. Ann Thorac Surg 75, 204–209. Crisostomo, P. R., Wang, Y., Markel, T. A., Wang, M., Lahm, T., & Meldrum, D. R. (2008).
Al-Khaldi, A., Eliopoulos, N., Martineau, D., Lejeune, L., Lachapelle, K., & Galipeau, J. (2003). Human mesenchymal stem cells stimulated by TNF-alpha, LPS, or hypoxia produce
Postnatal bone marrow stromal cells elicit a potent VEGF-dependent neoangiogenic growth factors by an NF kappa B- but not JNK-dependent mechanism. Am J Physiol
response in vivo. Gene Ther 10, 621–629. Cell Physiol 294, C675–C682.
Alviano, F., Fossati, V., Marchionni, C., Arpinati, M., Bonsi, L., Franchina, M., Lanzoni, G., Croitoru-Lamoury, J., Lamoury, F. M., Zaunders, J. J., Veas, L. A., & Brew, B. J. (2007). Human
Cantoni, S., Cavallini, C., Bianchi, F., Tazzari, P. L., Pasquinelli, G., Foroni, L., Ventura, mesenchymal stem cells constitutively express chemokines and chemokine receptors
C., Grossi, A., & Bagnara, G. P. (2007). Term Amniotic membrane is a high throughput that can be upregulated by cytokines, IFN-beta, and Copaxone. J Interferon Cytokine
source for multipotent Mesenchymal Stem Cells with the ability to differentiate into Res 27, 53–64.
endothelial cells in vitro. BMC Dev Biol 7, 11. Cuiffo, B. G., & Karnoub, A. E. (2012). Mesenchymal stem cells in tumor development:
Anonymous (1995). Tissue plasminogen activator for acute ischemic stroke. The National emerging roles and concepts. Cell Adh Migr 6, 220–230.
Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl J Med Dai, W., Hale, S. L., Martin, B. J., Kuang, J. Q., Dow, J. S., Wold, L. E., et al. (2005). Allogeneic
333, 1581–1587. mesenchymal stem cell transplantation in postinfarcted rat myocardium: short- and
Aranha, A.M., Zhang, Z., Neiva, K. G., Costa, C. A., Hebling, J., & Nor, J. E. (2010). Hypoxia long-term effects. Circulation 112, 214–223.
enhances the angiogenic potential of human dental pulp cells. J Endod 36, 1633–1637. De Luca, A., Gallo, M., Aldinucci, D., Ribatti, D., Lamura, L., D'Alessio, A., et al. (2011). Role
Arroyo, A. G., & Iruela-Arispe, M. L. (2010). Extracellular matrix, inflammation, and the an- of the EGFR ligand/receptor system in the secretion of angiogenic factors in mesen-
giogenic response. Cardiovasc Res 86, 226–235. chymal stem cells. J Cell Physiol 226, 2131–2138.
Au, P., Tam, J., Fukumura, D., & Jain, R. K. (2008). Bone marrow-derived mesenchymal De Ryck, M., Van Reempts, J., Borgers, M., Wauquier, A., & Janssen, P. A. (1989). Photo-
stem cells facilitate engineering of long-lasting functional vasculature. Blood 111, chemical stroke model: flunarizine prevents sensorimotor deficits after neocortical
4551–4558. infarcts in rats. Stroke 20, 1383–1390.
Bai, K., Huang, Y., Jia, X., Fan, Y., & Wang, W. (2010). Endothelium oriented differentiation Deuse, T., Peter, C., Fedak, P. W., Doyle, T., Reichenspurner, H., Zimmermann, W. H., et al.
of bone marrow mesenchymal stem cells under chemical and mechanical stimula- (2009). Hepatocyte growth factor or vascular endothelial growth factor gene transfer
tions. J Biomech 43, 1176–1181. maximizes mesenchymal stem cell-based myocardial salvage after acute myocardial
Ball, S. G., Bayley, C., Shuttleworth, C. A., & Kielty, C. M. (2010). Neuropilin-1 regulates infarction. Circulation 120, S247–S254.
platelet-derived growth factor receptor signalling in mesenchymal stem cells. Djouad, F., Plence, P., Bony, C., Tropel, P., Apparailly, F., Sany, J., et al. (2003). Immunosup-
Biochem J 427, 29–40. pressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals.
Banerjee, S., Williamson, D. A., Habib, N., & Chataway, J. (2012). The potential benefit of Blood 102, 3837–3844.
stem cell therapy after stroke: an update. Vasc Health Risk Manag 8, 569–580. Duffy, G. P., Ahsan, T., O'Brien, T., Barry, F., & Nerem, R. M. (2009). Bone marrow-derived
Bang, O. Y., Lee, J. S., Lee, P. H., & Lee, G. (2005). Autologous mesenchymal stem cell trans- mesenchymal stem cells promote angiogenic processes in a time- and dose-
plantation in stroke patients. Ann Neurol 57, 874–882. dependent manner in vitro. Tissue Eng Part A 15, 2459–2470.
Ben-Ami, E., Berrih-Aknin, S., & Miller, A. (2011). Mesenchymal stem cells as an immuno- Dwyer, R. M., Khan, S., Barry, F. P., O'Brien, T., & Kerin, M. J. (2010). Advances in mesen-
modulatory therapeutic strategy for autoimmune diseases. Autoimmun Rev 10, chymal stem cell-mediated gene therapy for cancer. Stem Cell Res Ther 1, 25.
410–415. Eble, J. A., & Niland, S. (2009). The extracellular matrix of blood vessels. Curr Pharm Des 15,
Bento, L. W., Zhang, Z., Imai, A., Nor, F., Dong, Z., Shi, S., et al. (2013). Endothelial differen- 1385–1400.
tiation of SHED requires MEK1/ERK signaling. J Dent Res 92, 51–57. Erices, A., Conget, P., & Minguell, J. J. (2000). Mesenchymal progenitor cells in human um-
Bergers, G., & Benjamin, L. E. (2003). Tumorigenesis and the angiogenic switch. Nat Rev bilical cord blood. Br J Haematol 109, 235–242.
Cancer 3, 401–410. Estrada, R., Li, N., Sarojini, H., An, J., Lee, M. J., & Wang, E. (2009). Secretome from mesen-
Bhadada, S. V., Goyal, B. R., & Patel, M. M. (2011). Angiogenic targets for potential disor- chymal stem cells induces angiogenesis via Cyr61. J Cell Physiol 219, 563–571.
ders. Fundam Clin Pharmacol 25, 29–47. Falanga, V., Iwamoto, S., Chartier, M., Yufit, T., Butmarc, J., Kouttab, N., et al. (2007). Autolo-
Bianco, P., Robey, P. G., & Simmons, P. J. (2008). Mesenchymal stem cells: revisiting histo- gous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray
ry, concepts, and assays. Cell Stem Cell 2, 313–319. accelerate healing in murine and human cutaneous wounds. Tissue Eng 13, 1299–1312.
Bieback, K., Wuchter, P., Besser, D., Franke, W., Becker, M., Ott, M., et al. (2012). Mesen- Fan, W., Crawford, R., & Xiao, Y. (2011). The ratio of VEGF/PEDF expression in bone marrow
chymal stromal cells (MSCs): science and f(r)iction. J Mol Med (Berl) 90, 773–782. mesenchymal stem cells regulates neovascularization. Differentiation 81, 181–191.
Block, G. J., Ohkouchi, S., Fung, F., Frenkel, J., Gregory, C., Pochampally, R., et al. (2009). Ferrara, N., & Kerbel, R. S. (2005). Angiogenesis as a therapeutic target. Nature 438,
Multipotent stromal cells are activated to reduce apoptosis in part by upregulation 967–974.
and secretion of stanniocalcin-1. Stem Cells 27, 670–681. Fischer, L. J., McIlhenny, S., Tulenko, T., Golesorkhi, N., Zhang, P., Larson, R., et al. (2009).
Boomsma, R. A., & Geenen, D. L. (2012). Mesenchymal stem cells secrete multiple cyto- Endothelial differentiation of adipose-derived stem cells: effects of endothelial cell
kines that promote angiogenesis and have contrasting effects on chemotaxis and ap- growth supplement and shear force. J Surg Res 152, 157–166.
optosis. PLoS One 7, e35685. Folkman, J. (1971). Tumor angiogenesis: therapeutic implications. N Engl J Med 285,
Bronckaers, A., Hilkens, P., Fanton, Y., Struys, T., Gervois, P., Politis, C., et al. (2013). Angio- 1182–1186.
genic properties of human dental pulp stem cells. PLoS One 8, e71104. Friedenstein, A. J., Chailakhjan, R. K., & Lalykina, K. S. (1970). The development of fibro-
Brown, R. (2005). Adam and Victor's Principles of Neurology (8th ed.). McGraw Hill. blast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells.
Burlacu, A., Grigorescu, G., Rosca, A.M., Preda, M. B., & Simionescu, M. (2013). Factors se- Cell Tissue Kinet 3, 393–403.
creted by mesenchymal stem cells and endothelial progenitor cells have complemen- Friedenstein, A. J., Piatetzky, S., II, & Petrakova, K. V. (1966). Osteogenesis in transplants of
tary effects on angiogenesis in vitro. Stem Cells Dev 22, 643–653. bone marrow cells. J Embryol Exp Morphol 16, 381–390.
Caplan, A. I., & Correa, D. (2011). The MSC: an injury drugstore. Cell Stem Cell 9, 11–15. Galas, R. J., Jr., & Liu, J. C. (2014). Vascular endothelial growth factor does not accelerate
Carmeliet, P. (2005). Angiogenesis in life, disease and medicine. Nature 438, 932–936. endothelial differentiation of human mesenchymal stem cells. J Cell Physiol 229(1),
Carmeliet, P., & Jain, R. K. (2011). Molecular mechanisms and clinical applications of an- 90–96, http://dx.doi.org/10.1002/jcp.24421.
giogenesis. Nature 473, 298–307. Galie, M., Konstantinidou, G., Peroni, D., Scambi, I., Marchini, C., Lisi, V., et al. (2008). Mes-
Carmeliet, P., & Jain, R. K. (2011). Principles and mechanisms of vessel normalization for enchymal stem cells share molecular signature with mesenchymal tumor cells and
cancer and other angiogenic diseases. Nat Rev Drug Discov 10, 417–427. favor early tumor growth in syngeneic mice. Oncogene 27, 2542–2551.

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
14 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

Gandia, C., Arminan, A., Garcia-Verdugo, J. M., Lledo, E., Ruiz, A., Minana, M.D., et al. Kanehira, M., Xin, H., Hoshino, K., Maemondo, M., Mizuguchi, H., Hayakawa, T., et al.
(2008). Human dental pulp stem cells improve left ventricular function, induce an- (2007). Targeted delivery of NK4 to multiple lung tumors by bone marrow-derived
giogenesis, and reduce infarct size in rats with acute myocardial infarction. Stem mesenchymal stem cells. Cancer Gene Ther 14, 894–903.
Cells 26, 638–645. Karbanova, J., Soukup, T., Suchanek, J., Pytlik, R., Corbeil, D., & Mokry, J. (2011). Character-
Gang, E. J., Jeong, J. A., Han, S., Yan, Q., Jeon, C. J., & Kim, H. (2006). In vitro endothelial po- ization of dental pulp stem cells from impacted third molars cultured in low serum-
tential of human UC blood-derived mesenchymal stem cells. Cytotherapy 8, 215–227. containing medium. Cells Tissues Organs 193, 344–365.
Goshima, J., Goldberg, V. M., & Caplan, A. I. (1991). The osteogenic potential of culture- Kawada, H., Fujita, J., Kinjo, K., Matsuzaki, Y., Tsuma, M., Miyatake, H., et al. (2004).
expanded rat marrow mesenchymal cells assayed in vivo in calcium phosphate ce- Nonhematopoietic mesenchymal stem cells can be mobilized and differentiate into
ramic blocks. Clin Orthop Relat Res, 298–311. cardiomyocytes after myocardial infarction. Blood 104, 3581–3587.
Gronthos, S., Mankani, M., Brahim, J., Robey, P. G., & Shi, S. (2000). Postnatal human dental Keating, A. (2012). Mesenchymal stromal cells: new directions. Cell Stem Cell 10,
pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A 97, 13625–13630. 709–716.
Gruber, R., Kandler, B., Holzmann, P., Vogele-Kadletz, M., Losert, U., Fischer, M. B., et al. Khakoo, A. Y., Pati, S., Anderson, S. A., Reid, W., Elshal, M. F., Rovira, I. I., et al. (2006).
(2005). Bone marrow stromal cells can provide a local environment that favors mi- Human mesenchymal stem cells exert potent antitumorigenic effects in a model of
gration and formation of tubular structures of endothelial cells. Tissue Eng 11, Kaposi's sarcoma. J Exp Med 203, 1235–1247.
896–903. Kilroy, G. E., Foster, S. J., Wu, X., Ruiz, J., Sherwood, S., Heifetz, A., et al. (2007). Cyto-
Gutierrez-Fernandez, M., Rodriguez-Frutos, B., Ramos-Cejudo, J., Teresa Vallejo-Cremades, kine profile of human adipose-derived stem cells: expression of angiogenic, he-
M., Fuentes, B., Cerdan, S., et al. (2013). Effects of intravenous administration of allo- matopoietic, and pro-inflammatory factors. J Cell Physiol 212, 702–709.
genic bone marrow- and adipose tissue-derived mesenchymal stem cells on func- Kim, S. W., Zhang, H. Z., Kim, C. E., Kim, J. M., & Kim, M. H. (2013). Amniotic mesenchymal
tional recovery and brain repair markers in experimental ischemic stroke. Stem Cell stem cells with robust chemotactic properties are effective in the treatment of a myo-
Res Ther 4, 11. cardial infarction model. Int J Cardiol 168(2), 1062–1069, http://dx.doi.org/10.1016/j.
Hacein-Bey-Abina, S., von Kalle, C., Schmidt, M., Le Deist, F., Wulffraat, N., McIntyre, E., ijcard.2012.11.003 (Epub 2012 Dec 4).
et al. (2003). A serious adverse event after successful gene therapy for X-linked se- Kinnaird, T., Stabile, E., Burnett, M. S., Lee, C. W., Barr, S., Fuchs, S., et al. (2004). Marrow-
vere combined immunodeficiency. N Engl J Med 348, 255–256. derived stromal cells express genes encoding a broad spectrum of arteriogenic cyto-
Hacke, W., Donnan, G., Fieschi, C., Kaste, M., von Kummer, R., Broderick, J. P., et al. (2004). kines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms.
Association of outcome with early stroke treatment: pooled analysis of ATLANTIS, Circ Res 94, 678–685.
ECASS, and NINDS rt-PA stroke trials. Lancet 363, 768–774. Kinnaird, T., Stabile, E., Burnett, M. S., Shou, M., Lee, C. W., Barr, S., et al. (2004). Local de-
Hall, B. K. (2008). The neural crest and neural crest cells: discovery and significance for livery of marrow-derived stromal cells augments collateral perfusion through para-
theories of embryonic organization. J Biosci 33, 781–793. crine mechanisms. Circulation 109, 1543–1549.
Hare, J. M., Traverse, J. H., Henry, T. D., Dib, N., Strumpf, R. K., Schulman, S. P., et al. (2009). Klopp, A. H., Gupta, A., Spaeth, E., Andreeff, M., & Marini, F., III (2011). Concise review: dis-
A randomized, double-blind, placebo-controlled, dose-escalation study of intrave- secting a discrepancy in the literature: do mesenchymal stem cells support or sup-
nous adult human mesenchymal stem cells (prochymal) after acute myocardial in- press tumor growth? Stem Cells 29, 11–19.
farction. J Am Coll Cardiol 54, 2277–2286. Konno, M., Hamazaki, T. S., Fukuda, S., Tokuhara, M., Uchiyama, H., Okazawa, H., Okochi,
Harris, S., Craze, M., Newton, J., Fisher, M., Shima, D. T., Tozer, G. M., et al. (2012). Do H., & Asashima, M. (2010). Efficiently differentiating vascular endothelial cells from
anti-angiogenic VEGF (VEGFxxxb) isoforms exist? A cautionary tale. PLoS One 7, adipose tissue-derived mesenchymal stem cells in serum-free culture. Biochem
e35231. Biophys Res Commun 400.
Herbert, S. P., & Stainier, D. Y. (2011). Molecular control of endothelial cell behaviour dur- Kopen, G. C., Prockop, D. J., & Phinney, D.G. (1999). Marrow stromal cells migrate
ing blood vessel morphogenesis. Nat Rev Mol Cell Biol 12, 551–564. throughout forebrain and cerebellum, and they differentiate into astrocytes
Hocking, A.M., & Gibran, N. S. (2010). Mesenchymal stem cells: paracrine signaling and after injection into neonatal mouse brains. Proc Natl Acad Sci U S A 96,
differentiation during cutaneous wound repair. Exp Cell Res 316, 2213–2219. 10711–10716.
Honmou, O., Houkin, K., Matsunaga, T., Niitsu, Y., Ishiai, S., Onodera, R., et al. (2011). Intra- Kucerova, L., Matuskova, M., Pastorakova, A., Tyciakova, S., Jakubikova, J., Bohovic,
venous administration of auto serum-expanded autologous mesenchymal stem cells R., et al. (2008). Cytosine deaminase expressing human mesenchymal stem
in stroke. Brain 134, 1790–1807. cells mediated tumour regression in melanoma bearing mice. J Gene Med 10,
Horwitz, E. M., Gordon, P. L., Koo, W. K., Marx, J. C., Neel, M.D., McNall, R. Y., et al. (2002). 1071–1082.
Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate Kudo, M., Aoyama, A., Ichimori, S., & Fukunaga, N. (1982). An animal model of cerebral in-
growth in children with osteogenesis imperfecta: implications for cell therapy of farction. Homologous blood clot emboli in rats. Stroke 13, 505–508.
bone. Proc Natl Acad Sci U S A 99, 8932–8937. Kurozumi, K., Nakamura, K., Tamiya, T., Kawano, Y., Ishii, K., Kobune, M., et al. (2005).
Horwitz, E. M., Le Blanc, K., Dominici, M., Mueller, I., Slaper-Cortenbach, I., Marini, F. C., Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage
et al. (2005). Clarification of the nomenclature for MSC: the International Society in the rat middle cerebral artery occlusion model. Mol Ther 11, 96–104.
for Cellular Therapy position statement. Cytotherapy 7, 393–395. Lalu, M. M., McIntyre, L., Pugliese, C., Fergusson, D., Winston, B. W., Marshall, J. C., et al.
Hu, Y. L., Fu, Y. H., Tabata, Y., & Gao, J. Q. (2010). Mesenchymal stem cells: a promising (2012). Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systemat-
targeted-delivery vehicle in cancer gene therapy. J Control Release 147, 154–162. ic review and meta-analysis of clinical trials. PLoS One 7, e47559.
Hung, S.C., Pochampally, R. R., Chen, S.C., Hsu, S.C., & Prockop, D. J. (2007). Angiogenic ef- Laschke, M. W., Harder, Y., Amon, M., Martin, I., Farhadi, J., Ring, A., et al. (2006). Angio-
fects of human multipotent stromal cell conditioned medium activate the PI3K–Akt genesis in tissue engineering: breathing life into constructed tissue substitutes.
pathway in hypoxic endothelial cells to inhibit apoptosis, increase survival, and stim- Tissue Eng 12, 2093–2104.
ulate angiogenesis. Stem Cells 25, 2363–2370. Le Blanc, K., Tammik, C., Rosendahl, K., Zetterberg, E., & Ringden, O. (2003). HLA expres-
Ikeda, N., Nonoguchi, N., Zhao, M. Z., Watanabe, T., Kajimoto, Y., Furutama, D., et al. sion and immunologic properties of differentiated and undifferentiated mesenchy-
(2005). Bone marrow stromal cells that enhanced fibroblast growth factor-2 secre- mal stem cells. Exp Hematol 31, 890–896.
tion by herpes simplex virus vector improve neurological outcome after transient Lee, J. S., Hong, J. M., Moon, G. J., Lee, P. H., Ahn, Y. H., Bang, O. Y., et al. (2010). A long-term
focal cerebral ischemia in rats. Stroke 36, 2725–2730. follow-up study of intravenous autologous mesenchymal stem cell transplantation in
Iohara, K., Zheng, L., Wake, H., Ito, M., Nabekura, J., Wakita, H., et al. (2008). A novel stem patients with ischemic stroke. Stem Cells 28, 1099–1106.
cell source for vasculogenesis in ischemia: subfraction of side population cells from Lee, M. Y., Huang, J. P., Chen, Y. Y., Aplin, J.D., Wu, Y. H., Chen, C. Y., et al. (2009). Angiogen-
dental pulp. Stem Cells 26, 2408–2418. esis in differentiated placental multipotent mesenchymal stromal cells is dependent
Iwase, T., Nagaya, N., Fujii, T., Itoh, T., Murakami, S., Matsumoto, T., et al. (2005). Compar- on integrin alpha5beta1. PLoS One 4, e6913.
ison of angiogenic potency between mesenchymal stem cells and mononuclear cells Lee, K. D., Kuo, T. K., Whang-Peng, J., Chung, Y. F., Lin, C. T., Chou, S. H., et al. (2004). In
in a rat model of hindlimb ischemia. Cardiovasc Res 66, 543–551. vitro hepatic differentiation of human mesenchymal stem cells. Hepatology 40,
Janebodin, K., Zeng, Y., Buranaphatthana, W., Ieronimakis, N., & Reyes, M. (2013). 1275–1284.
VEGFR2-dependent angiogenic capacity of pericyte-like dental pulp stem cells. J Lee, E. Y., Xia, Y., Kim, W. S., Kim, M. H., Kim, T. H., Kim, K. J., et al. (2009). Hypoxia-
Dent Res 92, 524–531. enhanced wound-healing function of adipose-derived stem cells: increase in stem
Jazayeri, M., Allameh, A., Soleimani, M., Jazayeri, S. H., Piryaei, A., & Kazemnejad, S. (2008). cell proliferation and up-regulation of VEGF and bFGF. Wound Repair Regen 17,
Molecular and ultrastructural characterization of endothelial cells differentiated from 540–547.
human bone marrow mesenchymal stem cells. Cell Biol Int 32, 1183–1192. Liu, J. W., Dunoyer-Geindre, S., Serre-Beinier, V., Mai, G., Lambert, J. F., Fish, R. J., et al.
Jeon, D., Chu, K., Lee, S. T., Jung, K. H., Ban, J. J., Park, D. K., et al. (2013). Neuroprotective (2007). Characterization of endothelial-like cells derived from human mesenchymal
effect of a cell-free extract derived from human adipose stem cells in experimental stem cells. J Thromb Haemost 5, 826–834.
stroke models. Neurobiol Dis 54, 414–420. Liu, Y., Han, Z. P., Zhang, S. S., Jing, Y. Y., Bu, X. X., Wang, C. Y., et al. (2011). Effects of in-
Jeon, E. S., Lee, I. H., Heo, S.C., Shin, S. H., Choi, Y. J., Park, J. H., et al. (2010). Mesenchymal flammatory factors on mesenchymal stem cells and their role in the promotion of
stem cells stimulate angiogenesis in a murine xenograft model of A549 human ade- tumor angiogenesis in colon cancer. J Biol Chem 286, 25007–25015.
nocarcinoma through an LPA1 receptor-dependent mechanism. Biochim Biophys Acta Liu, S. H., Huang, J. P., Lee, R. K., Huang, M. C., Wu, Y. H., Chen, C. Y., et al. (2010). Paracrine
1801, 1205–1213. factors from human placental multipotent mesenchymal stromal cells protect endo-
Jones, E. A., Kinsey, S. E., English, A., Jones, R. A., Straszynski, L., Meredith, D.M., et al. thelium from oxidative injury via STAT3 and manganese superoxide dismutase acti-
(2002). Isolation and characterization of bone marrow multipotential mesenchymal vation. Biol Reprod 82, 905–913.
progenitor cells. Arthritis Rheum 46, 3349–3360. Lozito, T. P., Kuo, C. K., Taboas, J. M., & Tuan, R. S. (2009). Human mesenchymal stem cells
Kachgal, S., & Putnam, A. J. (2011). Mesenchymal stem cells from adipose and bone mar- express vascular cell phenotypes upon interaction with endothelial cell matrix. J Cell
row promote angiogenesis via distinct cytokine and protease expression mecha- Biochem 107, 714–722.
nisms. Angiogenesis 14, 47–59. Ma, X. L., Liu, K. D., Li, F. C., Jiang, X. M., Jiang, L., & Li, H. L. (2013). Human mesenchymal
Kandel, E. R., & Pittenger, C. (1999). The past, the future and the biology of memory stor- stem cells increases expression of alpha-tubulin and angiopoietin 1 and 2 in focal ce-
age. Philos Trans R Soc Lond B Biol Sci 354, 2027–2052. rebral ischemia and reperfusion. Curr Neurovasc Res 10, 103–111.

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx 15

Majumdar, M. K., Thiede, M. A., Mosca, J. D., Moorman, M., & Gerson, S. L. (1998). Pheno- Santos, J. L., Pandita, D., Rodrigues, J., Pego, A. P., Granja, P. L., & Tomas, H. (2011). Non-
typic and functional comparison of cultures of marrow-derived mesenchymal stem viral gene delivery to mesenchymal stem cells: methods, strategies and application
cells (MSCs) and stromal cells. J Cell Physiol 176, 57–66. in bone tissue engineering and regeneration. Curr Gene Ther 11, 46–57.
Marchionni, C., Bonsi, L., Alviano, F., Lanzoni, G., Di Tullio, A., Costa, R., Montanari, M., Sasaki, M., Abe, R., Fujita, Y., Ando, S., Inokuma, D., & Shimizu, H. (2008). Mesenchymal
Tazzari, P. L., Ricci, F., Pasquinelli, G., Orrico, C., Grossi, A., Prati, C., & Bagnara, G. P. stem cells are recruited into wounded skin and contribute to wound repair by
(2009). Angiogenic potential of human dental pulp stromal (stem) cells. Int J transdifferentiation into multiple skin cell type. J Immunol 180, 2581–2587.
Immunopathol Pharmacol 22, 699–706. Schiavetta, A., Maione, C., Botti, C., Marino, G., Lillo, S., Garrone, A., et al. (2012). A
Marshall, E. (1999). Gene therapy death prompts review of adenovirus vector. Science phase II trial of autologous transplantation of bone marrow stem cells for critical
286, 2244–2245. limb ischemia: results of the Naples and Pietra Ligure Evaluation of Stem Cells
Maul, T. M., Chew, D. W., Nieponice, A., & Vorp, D. A. (2011). Mechanical stimuli differen- study. Stem Cells Transl Med 1, 572–578.
tially control stem cell behavior: morphology, proliferation, and differentiation. Shabbir, A., Zisa, D., Lin, H., Mastri, M., Roloff, G., Suzuki, G., et al. (2010). Activation of host
Biomech Model Mechanobiol 10, 939–953. tissue trophic factors through JAK–STAT3 signaling: a mechanism of mesenchymal
Meirelles Lda, S., Fontes, A.M., Covas, D. T., & Caplan, A. I. (2009). Mechanisms involved in stem cell-mediated cardiac repair. Am J Physiol Heart Circ Physiol 299, H1428–H1438.
the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev 20, Shah, K. (2012). Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv
419–427. Rev 64, 739–748.
Menon, L. G., Shi, V. J., & Carroll, R. S. (2008). Mesenchymal Stromal Cells as a Drug Delivery Simmons, P. J., & Torok-Storb, B. (1991). Identification of stromal cell precursors in human
System. StemBook. (Cambridge (MA)): Doug Melton (Harvard Stem Cell Institute, bone marrow by a novel monoclonal antibody, STRO-1. Blood 78, 55–62.
Harvard University). Sorrell, J. M., Baber, M.A., & Caplan, A. I. (2009). Influence of adult mesenchymal stem cells
Miletic, H., Fischer, Y., Litwak, S., Giroglou, T., Waerzeggers, Y., Winkeler, A., et al. (2007). on in vitro vascular formation. Tissue Eng Part A 15, 1751–1761.
Bystander killing of malignant glioma by bone marrow-derived tumor-infiltrating Spees, J. L., Olson, S. D., Whitney, M. J., & Prockop, D. J. (2006). Mitochondrial transfer
progenitor cells expressing a suicide gene. Mol Ther 15, 1373–1381. between cells can rescue aerobic respiration. Proc Natl Acad Sci U S A 103,
Miyahara, Y., Nagaya, N., Kataoka, M., Yanagawa, B., Tanaka, K., Hao, H., et al. (2006). 1283–1288.
Monolayered mesenchymal stem cells repair scarred myocardium after myocardial Suzuki, K., Sun, R., Origuchi, M., Kanehira, M., Takahata, T., Itoh, J., et al. (2011). Mesenchy-
infarction. Nat Med 12, 459–465. mal stromal cells promote tumor growth through the enhancement of neovasculari-
Moon, M. H., Kim, S. Y., Kim, Y. J., Kim, S. J., Lee, J. B., Bae, Y. C., et al. (2006). Human zation. Mol Med 17, 579–587.
adipose tissue-derived mesenchymal stem cells improve postnatal neovascular- Takahashi, M., Suzuki, E., Oba, S., Nishimatsu, H., Kimura, K., Nagano, T., et al. (2010).
ization in a mouse model of hindlimb ischemia. Cell Physiol Biochem 17, Adipose tissue-derived stem cells inhibit neointimal formation in a paracrine
279–290. fashion in rat femoral artery. Am J Physiol Heart Circ Physiol 298, H415–H423.
Nadri, S., & Soleimani, M. (2007). Comparative analysis of mesenchymal stromal cells Tögel, F., Weiss, K., Yang, Y., Hu, Z., Zhang, P., & Westenfelder, C. (2007). Vasculotropic,
from murine bone marrow and amniotic fluid. Cytotherapy 9, 729–737. paracrine actions of infused mesenchymal stem cells are important to the recovery
Nakamizo, A., Marini, F., Amano, T., Khan, A., Studeny, M., Gumin, J., et al. (2005). Human from acute kidney injury. Am J Physiol Renal Physiol 292, F1626–1635.
bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Toma, C., Pittenger, M. F., Cahill, K. S., Byrne, B. J., & Kessler, P. D. (2002). Human mesen-
Res 65, 3307–3318. chymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine
Oswald, J., Boxberger, S., Jorgensen, B., Feldmann, S., Ehninger, G., Bornhauser, M., et al. heart. Circulation 105, 93–98.
(2004). Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Tongers, J., Losordo, D. W., & Landmesser, U. (2011). Stem and progenitor cell-based ther-
Stem Cells 22, 377–384. apy in ischaemic heart disease: promise, uncertainties, and challenges. Eur Heart J 32,
Otsu, K., Das, S., Houser, S. D., Quadri, S. K., Bhattacharya, S., & Bhattacharya, J. (2009). 1197–1206.
Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells. Traktuev, D. O., Merfeld-Clauss, S., Li, J., Kolonin, M., Arap, W., Pasqualini, R., et al. (2008).
Blood 113, 4197–4205. A population of multipotent CD34-positive adipose stromal cells share pericyte and
Pankajakshan, D., Kansal, V., & Agrawal, D. K. (2013). In vitro differentiation of bone mar- mesenchymal surface markers, reside in a periendothelial location, and stabilize en-
row derived porcine mesenchymal stem cells to endothelial cells. J Tissue Eng Regen dothelial networks. Circ Res 102, 77–85.
Med 7, 911–920. Tran-Hung, L., Laurent, P., Camps, J., & About, I. (2008). Quantification of angiogenic
Patki, S., Kadam, S., Chandra, V., & Bhonde, R. (2010). Human breast milk is a rich source growth factors released by human dental cells after injury. Arch Oral Biol 53,
of multipotent mesenchymal stem cells. Hum Cell 23, 35–40. 9–13.
Peister, A., Mellad, J. A., Larson, B.L., Hall, B.M., Gibson, L. F., & Prockop, D. J. (2004). Adult Ukai, R., Honmou, O., Harada, K., Houkin, K., Hamada, H., & Kocsis, J.D. (2007). Mes-
stem cells from bone marrow (MSCs) isolated from different strains of inbred mice enchymal stem cells derived from peripheral blood protects against ischemia.
vary in surface epitopes, rates of proliferation, and differentiation potential. Blood 103, J Neurotrauma 24, 508–520.
1662–1668. Valarmathi, M. T., Davis, J. M., Yost, M. J., Goodwin, R. L., & Potts, J.D. (2009). A three-
Phng, L. K., & Gerhardt, H. (2009). Angiogenesis: a team effort coordinated by notch. Dev dimensional model of vasculogenesis. Biomaterials 30, 1098–1112.
Cell 16, 196–208. van der Spoel, T. I., Jansen of Lorkeers, S. J., Agostoni, P., van Belle, E., Gyongyosi, M.,
Pittenger, M. F., Mackay, A.M., Beck, S.C., Jaiswal, R. K., Douglas, R., Mosca, J.D., et al. Sluijter, J. P., et al. (2011). Human relevance of pre-clinical studies in stem cell
(1999). Multilineage potential of adult human mesenchymal stem cells. Science therapy: systematic review and meta-analysis of large animal models of ischae-
284, 143–147. mic heart disease. Cardiovasc Res 91, 649–658.
Porada, C. D., & Almeida-Porada, G. (2010). Mesenchymal stem cells as therapeutics and van Eekelen, M., Sasportas, L. S., Kasmieh, R., Yip, S., Figueiredo, J. L., Louis, D. N., et al.
vehicles for gene and drug delivery. Adv Drug Deliv Rev 62, 1156–1166. (2010). Human stem cells expressing novel TSP-1 variant have anti-angiogenic effect
Portalska, K. J., Groen, N., Krenning, G., Georgi, N., Mentink, A., Harmsen, M. C., van on brain tumors. Oncogene 29, 3185–3195.
Blitterswijk, C., & de Boer, J. (2013). The effect of donor variation and senescence Voyta, J. C., Via, D. P., Butterfield, C. E., & Zetter, B. R. (1984). Identification and isolation of
on endothelial differentiation of human mesenchymal stromal cells. Tissue Eng Part endothelial cells based on their increased uptake of acetylated-low density lipopro-
A 19, 2318–2329. tein. J Cell Biol 99, 2034–2040.
Potapova, I. A., Gaudette, G. R., Brink, P. R., Robinson, R. B., Rosen, M. R., Cohen, I. S., et al. Whyte, J. L., Ball, S. G., Shuttleworth, C. A., Brennan, K., & Kielty, C. M. (2011). Density of
(2007). Mesenchymal stem cells support migration, extracellular matrix invasion, human bone marrow stromal cells regulates commitment to vascular lineages.
proliferation, and survival of endothelial cells in vitro. Stem Cells 25, 1761–1768. Stem Cell Res 6, 238–250.
Potente, M., Gerhardt, H., & Carmeliet, P. (2011). Basic and therapeutic aspects of angio- Windle, V., Szymanska, A., Granter-Button, S., White, C., Buist, R., Peeling, J., et al. (2006).
genesis. Cell 146, 873–887. An analysis of four different methods of producing focal cerebral ischemia with
Prockop, D. J., & Oh, J. Y. (2012). Medical therapies with adult stem/progenitor cells endothelin-1 in the rat. Exp Neurol 201, 324–334.
(MSCs): a backward journey from dramatic results in vivo to the cellular and molec- Wingate, K., Bonani, W., Tan, Y., Bryant, S. J., & Tan, W. (2012). Compressive elasticity of
ular explanations. J Cell Biochem 113, 1460–1469. three-dimensional nanofiber matrix directs mesenchymal stem cell differentiation
Ranganath, S. H., Levy, O., Inamdar, M. S., & Karp, J. M. (2012). Harnessing the mesenchy- to vascular cells with endothelial or smooth muscle cell markers. Acta Biomater 8,
mal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell 10, 1440–1449.
244–258. Wislet-Gendebien, S., Hans, G., Leprince, P., Rigo, J. M., Moonen, G., & Rogister, B. (2005).
Rehman, J., Traktuev, D., Li, J., Merfeld-Clauss, S., Temm-Grove, C. J., Bovenkerk, J. E., et al. Plasticity of cultured mesenchymal stem cells: switch from nestin-positive to excit-
(2004). Secretion of angiogenic and antiapoptotic factors by human adipose stromal able neuron-like phenotype. Stem Cells 23, 392–402.
cells. Circulation 109, 1292–1298. Woolard, J., Bevan, H. S., Harper, S. J., & Bates, D. O. (2009). Molecular diversity of VEGF-A
Ribatti, D., & Crivellato, E. (2012). “Sprouting angiogenesis”, a reappraisal. Dev Biol 372, as a regulator of its biological activity. Microcirculation 16, 572–592.
157–165. Wu, X., Huang, L., Zhou, Q., Song, Y., Li, A., Jin, J., & Cui, B. (2005). Mesenchymal stem cells
Roobrouck, V. D., Clavel, C., Jacobs, S. A., Ulloa-Montoya, F., Crippa, S., Sohni, A., et al. participating in ex vivo endothelium repair and its effect on vascular smooth muscle
(2011). Differentiation potential of human postnatal mesenchymal stem cells, cells growth. International journal of cardiology 105, 274–282.
mesoangioblasts, and multipotent adult progenitor cells reflected in their tran- Wu, C. C., Chao, Y. C., Chen, C. N., Chien, S., Chen, Y. C., Chien, C. C., et al. (2008). Synergism
scriptome and partially influenced by the culture conditions. Stem Cells 29, of biochemical and mechanical stimuli in the differentiation of human placenta-
871–882. derived multipotent cells into endothelial cells. J Biomech 41, 813–821.
Ruiz de Almodovar, C., Lambrechts, D., Mazzone, M., & Carmeliet, P. (2009). Role Wu, Y., Chen, L., Scott, P. G., & Tredget, E. E. (2007). Mesenchymal stem cells enhance
and therapeutic potential of VEGF in the nervous system. Physiol Rev 89, wound healing through differentiation and angiogenesis. Stem Cells 25,
607–648. 2648–2659.
Sacchetti, B., Funari, A., Michienzi, S., Di Cesare, S., Piersanti, S., Saggio, I., et al. (2007). Self- Xu, J., Liu, X., Chen, J., Zacharek, A., Cui, X., Savant-Bhonsale, S., et al. (2009). Simvastatin
renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic enhances bone marrow stromal cell differentiation into endothelial cells via notch
microenvironment. Cell 131, 324–336. signaling pathway. Am J Physiol Cell Physiol 296, C535–C543.

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013
16 A. Bronckaers et al. / Pharmacology & Therapeutics xxx (2014) xxx–xxx

Xu, J., Qu, J., Cao, L., Sai, Y., Chen, C., He, L., et al. (2008). Mesenchymal stem cell-based Zhang, J., Li, Y., Zhang, Z. G., Lu, M., Borneman, J., Buller, B., et al. (2009). Bone marrow
angiopoietin-1 gene therapy for acute lung injury induced by lipopolysaccharide in stromal cells increase oligodendrogenesis after stroke. J Cereb Blood Flow Metab 29,
mice. J Pathol 214, 472–481. 1166–1174.
Yue, W. M., Liu, W., Bi, Y. W., He, X. P., Sun, W. Y., Pang, X. Y., et al. (2008). Mesenchymal Zhang, G., Zhou, J., Fan, Q., Zheng, Z., Zhang, F., Liu, X., et al. (2008). Arterial-venous
stem cells differentiate into an endothelial phenotype, reduce neointimal formation, endothelial cell fate is related to vascular endothelial growth factor and Notch
and enhance endothelial function in a rat vein grafting model. Stem Cells Dev 17, status during human bone mesenchymal stem cell differentiation. FEBS Lett
785–793. 582, 2957–2964.
Zhang, G., Drinnan, C. T., Geuss, L. R., & Suggs, L. J. (2010). Vascular differentiation of bone Zuk, P. A., Zhu, M., Ashjian, P., De Ugarte, D. A., Huang, J. I., Mizuno, H., et al. (2002).
marrow stem cells is directed by a tunable three-dimensional matrix. Acta Biomater 6, Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13,
3395–3403. 4279–4295.

Please cite this article as: Bronckaers, A., et al., Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate
angiogenesis, Pharmacology & Therapeutics (2014), http://dx.doi.org/10.1016/j.pharmthera.2014.02.013

You might also like