Advances in Bioscience and Biotechnology, 2013, 4, 31-46 ABB
http://dx.doi.org/10.4236/abb.2013.410A4004 Published Online October 2013 (http://www.scirp.org/journal/abb/)
From skeletal to non skeletal: The intriguing roles of
BMP-9: A literature review
E. Leblanc1,2,3, G. Drouin3,4, G. Grenier3,4, N. Faucheux5, R. Hamdy1,2
1Department of Surgery, Orthopedic Surgery Division, McGill University, Montreal, Canada
2Shriners Hospital for Children, Montreal, Canada
3Department of Surgery, Orthopedic Surgery Division, Université de Sherbrooke, Sherbrooke, Canada
4Étienne-Lebel Clinical Research Centre, Université de Sherbrooke, Sherbrooke, Canada
5Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, Canada
Email: elisabeth.leblanc2@usherbrooke.ca
Received 13 August 2013; revised 14 September 2013; accepted 1 October 2013
Copyright © 2013 E. Leblanc et al. This is an open access article distributed under the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
In the well-known superfamily of transforming growth
factors beta (TGF-
), bone morphogenetic proteins
(BMPs) are one of the most compelling cytokines for
their major role in regulation of cell growth and dif-
ferentiation in both embryonic and adult tissues. This
subfamily was first described for its ability of poten-
tiating bone formation, but nowadays, the power of
BMPs is well beyond the bone healing scope. Some of
the BMPs have been well studied and described in the
literature, but the BMP9 is still worthy of attention. It
has been shown by many authors that it is the most
potent osteogenic BMP. Moreover, it has been de-
scribed as one of the rare circulating BMPs. In this
paper, we will review the recent literature on BMP9
and the different avenues for future research in that
field. Our primary scope is to review its relation to
bone formation and to elaborate on the available lit-
erature on other systems.
Keywords: BMP9; Review; Osteogenesis;
Chondrogenesis; Angiogenesis; Tumorigenesis;
Neurogenesis; Glucose Metabolism
1. INTRODUCTION
Fifty years ago, Urist et al. first reported that demineral-
ized bone matrix could promote ectopic bone formation
by stimulating transformation of primitive mesenchymal
cells of soft tissues into osteogenic cells [1]. Among the
growth factors with osteoinductive potential within the
bone matrix, the most important seem to be the bone
morphogenetic proteins (BMPs). BMPs belong to the
transforming growth factor beta (TGF-
) superfamily.
This group of proteins is characterized by a polybasic
proteolytic processing site which is cleaved to produce a
mature protein containing seven conserved cysteine re-
sidues [2]. More than 20 BMPs have been identified to
date [3], and most of them homodimerize via disulphide
bridges to be active, although heterodimers have been
described and are prop o sed to be m or e powerful [ 4,5].
Classically, BMPs are subdivided in four classes based
on their sequence homology: BMP 2/4, BMP 5/6/7/8,
BMP 9/10 and BMP 12/13/14 [6,7]. The sequence simi-
larity of the members of these subclasses is more than
50% and is also quite conserved across species [8,9].
BMPs signal through serine/threonine kinase receptors
composed of type I and II subtypes that activate the Smad
pathway and some mitogen activated protein kinases
(MAPKs) (Miyazono, 2005 #94; Bandyopadhyay, 2013
#320). Genetic disru ption of BMP pathways can result in
skeletal and extraskeletal abnormalities by their major role
in cell proliferation and differentiation during develop-
ment [10,11].
BMPs can also be classified by their osteogenic poten-
tial. To date, BMP2/4/6/7/9 have been identified as hav-
ing osteogenic properties, notably for their involvement
in the regulation of osteoblast differentiation [12-16 ] and
bone formation [14,17]. From this BMP group, BMP9
has been shown to be the most osteogenic [14]. This might
be due to the lack of inhibition of BMP9 by noggin, an
extracellular antagonist of BMP-2 or BMP-7 [12,18].
While some BMPs such as BMP 2/3/4/6/7/8 can be pro-
duced by preosteoblasts [19], BMP9 is mainly produced
in the liver [20,21]. BMP9 is also circulating at active
concentrations and is known to be a vascular quiescence
factor [22]. BMP9 is the physiological ligand of the en-
dothelial receptor ALK1 (activin receptor-like kinase 1,
also known as Acvrl1) in association with BMPRII [23-
Published Online October 2013 in SciRes. http://www.scirp.org/journal/abb
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
32
25]. ALK1 is one of the seven type I recaptors for TGF-
fa mily memb e rs [26] .
Review articles on BMP9 are needed to better under-
stand its potential. Lamplot et al. have recently reviewed
BMP9 signaling in depth. In this study, the osteogenic
pathways including the Smad and different mediators of
BMP9 were reviewed. They also point out crosstalk with
other factors such as TGF-β1, Wnt/β-catenin signaling,
Growth hormone, Mitogen activated protein kinases
(MAPKs), Hypoxia inducible factor 1 alpha (HIF1α),
Notch, Peroxisome proliferator-activated receptor gamma
(PPAR-γ), insulin-like growth factor (IGF) and retinoid
acid [27]. Here, we propose an overview of the multiple
roles of BMP9 in cell proliferation and differentiation
and its relation to current clinical issues. The compelling
osteogenic potential o f BMP9 will be discussed, but also
its role bone resorption, chondrog enesis, cancer develop-
ment, angiogenesis, neurogenesis, hepatocyte physiology,
glucose metabolism, and myogenesis (Figure 1).
2. MECHANISM OF ACTION
BMPs transduce signal through serine/threonine kinase
heterotetramer receptors composed of two type I and two
type II receptors. Once the BMP binds to the receptor
complex, the type II receptors phosphorylate the type I
receptors. Signal will then transduce via Smad (canonical)
or non-Smad pathways. In the canonical Smad pathway,
type I receptors phosphorylate the receptor regulated-
Smads (R-Smad), i.e. Smad1/5/8 which then recruit the
common mediator-Smad4 (coSmad). The R-Smad/co-
Smad complex translocates to the nucleus and regulates
target gene transcription by binding to Smad elements in
their promoters [6,28]. The Smad-independent pathway
[28,29].
In mice and humans, seven TGF-β family type I re-
ceptors (activin receptor-like kinase; ALK1 to ALK7)
have been identified. Luo et al. have demonstrated by
dominant negative mutations and RNAi studies that on ly
ALK1 and ALK2 mutants inhibited BMP9-induced
Smad activity in bone marrow mesenchymal stem cells
as well as osteogenic differentiation and ectopic bone
formation [30]. Brown et al. found that BMP9 has a
strong relative affinity for ALK1 and for BMPRII, one of
the five different TGF-β family type II receptors that
have been identified (ActRIIA, ActRIIB, BMPRII,
Figure 1. The multiple effects of BMP9. (): downregulation; (+): upregulation, MPC: mesenchymal progenitor cells.
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 33
TGFBRII and AMHRII) [23 ]. BMP9 also has a good af-
finity for Activin RIIA, another type II receptor [23].
More recently, Townson et al. analyzed the kinetic and
thermodynamic properties of BMP9 with the different
type II receptors and they noticed that ActRIIB had the
strongest affinity for BMP9 followed by BMPRII. The
affinity of BMP9 for ActRIIA was 300-fold weaker than
for ActRIIB [31].
To date, amongst all BMPs, only BMP2/4/6/7 and 9
have been identified to have osteogenic properties and
BMP9 seems to be the one with the strongest osteogenic
potential [14{Li, 2003 #7]}. Few studies tried to under-
stand why BMP9 has this strong osteogenic potential.
First, Bergeron et al. have shown that the Smad phos-
phorylation induced by BMP9 was not prevented by
noggin, an extracellular antagonist of the BMP pathway
[12]. These data were recently confirmed by two studies
by Wang et al. and Luther et al. and reinforced the hy-
pothesis that the high osteogenic capacity of BMP9 is
probably augmented by its noggin resistance [18,32].
Kang et al. also showed that BMP3 inhibited the osteo-
genic potential of BMP2 and BMP7 but had no effect on
BMP9 [14]. To date, no BMP9 inhibitor has been identi-
fied. Moreover, BMP9 is a secreted protein that has a
pro-region but this pro-region does not seem to function-
ally inhibit BMP9; quite opposite, the pro-region may
stabilize and protect BMP9 in vivo [23]. Finally, BMP9
has synergistic partners that would enhance its effects.
Growth hormones have been shown to be upregulated by
BMP9 and act synergistically to promote endochondral
ossification in mesenchymal progenitor cells (MPC) [33].
EGF also demonstrated an increase in BMP9-induced
bone formation in cultured mouse fœtal limb explants. In
MPCs, BMP9 expression was up-regulated when EGF
was present. Furthermore, EGF receptor expression was
directly up-regulated by BMP9 [34]. These specific fea-
tures could, in part, explain the strong osteogenic poten-
tial of BMP9 as compared to other osteogenic BMPs.
Production of endogenous BMP9 is mainly located in
the liver [20,21]. Indeed , BMP9 transcripts were lower in
the human lung (400 fold less), brain (470 fold less) and
bone marrow (3400 fold less) than in the liver [21]. Us-
ing Harlan Sprague Dawley rats, Miller et al. have first
identified the non-parenchymal liver cells (Kupffer and
endothelial cells) as BMP9 producers, no BMP9 being
detectable in hepatocytes [20]. In contrast, Bidart et al.
(2012) have recently found, using human liver biopsies,
that BMP9 is present in hepatocytes and intrahepatic
biliary epithelial cells [21]. In their experimental condi-
tions, BMP9 was not detected in blood vessel walls and
mesenchyme [21]. BMP9 mRNA was also detected by
semi quantitative reverse transcription polymerase chain
reaction (RT-PCR) in both fresh human endochondral
(iliac bone) and intramembranous (mandible) bones [35].
Primary cultures of osteoblastic cells extracted from hu-
man intramembranous and endochondral bones seem to
be able to synthesize BMP9 [35 ]. BMP9 is also found in
human circulation (2 - 12 ng/mL) under two forms: an
active (60%) and inactive form (40%) that can be further
activated by a serine endoprotease to a mature and fully
active form [21]. A mouse development study revealed
that BMP9 circulating levels peak within 3 weeks after
birth (with a peak at about 6 ng/mL) and decrease to 2
ng/mL in adulthood [21].
3. BMP9 AND BONE FORMATION
The first report about BMP9 as an osteogenic factor is in
the mid 90’s by Celeste et al. [36]. In this study, the au-
thors describe the molecular cloning and biological ac-
tivity of this new member of the TGF-
superfamily [36].
The osteogenic potential of BMP9 was then mostly test-
ed through viral vectors to promote bone formation in
many clinical settings.
Hence, Alden et al. [37] showed that they could fill a
cranio-facial critical bone defect in rats with an adenovi-
ral human BMP9 vector. Radiologic and histologic ana-
lyses of the mandibular bones demonstrated significant
bony healing compared to adenoviral
-galactosidase
controls. Also, the mechanism through which bone was
formed during the critical defect healing was proven to
be endochondral ossification [37].
In the spinal region, BMPs have been shown to pro-
mote spinal arthrodesis at a higher rate than autologous
bone alone, and to BMP2 and 7, which are actually ap-
proved for this specific indication [38]. Another group
has evaluated the efficacy of the use of a direct percuta-
neous injection of BMP9 expressing adenovirus to pro-
mote spinal fusion in rats [39]. The animals were directly
injected with viral v ectors in the lumbar paraspinal mus-
culature and were sacrificed after 16 weeks. Computer-
ized tomography studies and histological analysis dem-
onstrated bone induction at the injection sites, leading to
solid spinal arthrodesis. The authors did not have any
evidence of pseudarthroses, nerve root compression, or
systemic side effects. This study, while strongly support-
ing the advancement of BMP gene therapy techniques
toward clinical use, has also shown that the use of BMP9
through gene therapy could be an interesting avenue to
explore [39].
In a similar experimental design, ex vivo BMP9 gene
therapy on human mesenchymal stem cells (hMSCs) has
been proven to be efficacious for spinal fusion in rats.
Indeed, Dumont et al. infected ex vivo hMSCs by using
adenovirus (Ad) containing BMP9’s chimeric sequence
composed of the murine BMP9 proregion and human
mature region under the control of cytomegalovirus
(CMV) promoter [40]. The infected hMSCs by AdBMP9
or Ad β-galactosidase used as control were then injected
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
34
in paraspinal muscles of nude athymic rats. After only 8
weeks, rats treated with BMP9 expressing cells, showed
radiographically and histologically massive ectopic bone
formation leading to spinal fusion [40]. The results of
this study suggested that gene therapy on hMSCs ex vivo
using CMV driven BMP9 expression was possible and
may be useful to induce sp inal f usion . While show ing the
effect of BMP9 on hMSCs, this study also brought a
more attractive way of using BMP gene therapy. The
year after, the same team showed that a similar injection
of AdBMP9 ex vivo treated hMSCs was able to produce
ectopic bone formation in the hindlimbs musculature of
athymic nude rats [41].
Heterotopic bone formation by itself can tu rn out to be
useful if the muscle is used as a vascularised bone graft.
Hence, the latissimus dorsi muscle from nude rat was
infected with an adenoviral vector of BMP9 to make a
vascularised bone flap using heterotopic ossification as
an advantage [42]. With weekly biopsies and using RT-
PCR and immunohistochemistry, they found that the mu-
scle expressed chondrogenic and then osteogenic mark-
ers through the 3 weeks following the injection. They
estimated that the flap harvest sh ould be after 2 weeks to
be used as a vascularised moldable bone graft [42].
Gene therapy has been shown to be efficient, but the
transfer in humans is problematic because of its viral na-
ture and the existing lack of control over that kind of
technology. To work around that scientific and ethical
issue, nucleofecting hMSCs with nonviral plasmid ex-
pressing hBMP9 was developed [43]. Nonviral gene
transfer is safer, can be controlled and is reproducible.
hBMP9-nucleofected hMSCs in culture demonstrated an
increase in calcium deposition compared to control
EGFP (enhanced green fluorescent protein) nucleofected
hMSCs. Moreover, hBMP9-nucleofected hMSCs trans-
planted in ectopic sites in mice induced bone formation
28 days post-injection [43]. This safe and attractive al-
ternative was a breakthrough in the field of regenerative
medicine at the time, however this technique is demand-
ing.
In the late 70’s, Boyer’s team synthetized the first
functional polypeptide product from a gene of chemi-
cally synthesized origin [44]. This turning point in bio-
technology opened the door to the use of recombinant
proteins for clinical purpose. The use of recombinant
proteins is safer than viral gene transfer and yet more
accessible than nucleofection. Furthermore, the avail-
ability of recombinant BMP9 has liberalized its use and
led to more accessible research on its osteogenic poten-
tial. Even if recombinant human BMP9 (rhBMP9) was
used by Song and Celeste in the mid 90’s, reports on its
use for bone formation assays appear 10 years later in the
mid 2000’s [12,36,45,46]. The interest of using rhBMP9
is also related to the controlled dosag e and an easier cor-
relation to its effect. Thus rhBMP9 produced by E. coli at
500 ng/mL was shown to induce after 5 days th e expres-
sion of early markers of bone differentiation such as
smad-1/5, run × 2, dl × 5, osterix, osteopontin, bone sia-
loprotein and alkaline phosphatase in C2C12 cells [47].
In the same manner, Bergeron et al. have shown that a
peptide derived from BMP9, pBMP9 is able to induce
osteoblastic differentiation in pre-osteoblasts [12,48].
The osteoinductive potential of rhBMP9 was especial-
ly determined in the context of muscular heterotopic os-
sification (HO). Indeed, HO is a common model to study
the mechanism of ossification process in animals [49,50].
On the other hand, this model can be useful in order to
obtain a better understanding of the ossification process-
es occuring in muscle and to treat that condition [50].
Some papers have shown that adult muscle regeneration
could be impaired by BMPs [51,52]. Myogenesis was re-
ported to be inhibited by BMPs by their down-regulation
on satellite cells fusion [53]. Besides, mesenchymal pro-
genitor cells contained into the muscle are also respon-
sive to BMPs [54,55]. Overall, it was shown that both
myogenic and mesenchymal cell populations could be al-
tered by BMP and that tends to go from myogenesis to
osteogenesis [16].
Since the most common traumatic form of HO occurs
generally at sites of damaged muscles, we hypothesized a
relation between damaged muscle microenvironment and
BMP9 induced HO [56]. In this paper, we show for the
first times that BMP9 induces HO only in damaged mus-
cle. This paper highlighted the importance of cell micro-
environment for HO. We also showed that muscle resi-
dent stromal cells (mrSCs) are sensitive to BMP9 sig-
naling and express bone specific markers when exposed
to BMP9. Interestingly, mrSCs isolated from damaged
muscle are more responsive to BMP9. During a trans-
plantation experiment using labeled cells, we demon-
strated that mrSCs contribute to HO. Importantly, the
addition of a fusion protein containing the receptor
ALK1 significantly in hibited the osteoinductiv e potential
of BMP9 in mrSCs in vitro as well as HO in damaged
muscles [56].
Our experimental results show that a better under-
standing of the ossification processes and BMP implica-
tion is essential for developing new bone healing and HO
treatments. They also revealed that the osteoinductive
potential of rhBMP9 is highly dependent on the micro-
environment [56]. Therefore, delivery systems have to be
developed to protect the protein against breakdown. The
type of carrier, its concentration as well as the BMP dose
used in these delivery systems greatly influence the
growth factor release over time and its osteoinductive
potential [57]. In vitro and in vivo studies using different
delivery systems have been therefore studied throughout
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 35
the years for rhBMP9 and/or its less expensive derived
peptide (pBMP9) [48,58,59].
Delivery systems slow-down degradation and enhance
the effect of growth factors like BMP9. Bessa et al. has
recently developed BMP loaded silk fibroin microparti-
cles as delivery systems of BMP2, 9, or 14 [59]. The
encapsulation efficiency of the BMPs loaded at 0.5 or 5
µg varied from 68% to 98% and BMP14 was better re-
tained over time than BMP4 and BMP9 in this silk fib-
roin carrier [59].
The combination of collagen Type I gel, BMP (100
ng/mL) and bioactive glass microspheres (45S5 Bio-
glass®, GM) has been shown to in crease osteoblastic dif-
ferentiation by Bergeron et al. [48]. For this assay, a pep-
tide derived from BMP9 (pBMP9) to treat MC3T3-E1
preosteoblasts was used. Alkaline phosphatase staining
showed that pBMP9 induced osteoblast differentiation
and that bioactive GM increased this effect. Since pre-
osteoblasts secrete matrix metalloproteinases (MMPs),
the influence of the delivery system on MMPs expres-
sion was then studied. pBMP9 with bioactive GM gener-
ated less MMP than did rhBMP-2 [48]. Thus, this deliv-
ery system using a combination of bioactive GM and
BMP9 derived peptide seems to be promising.
In vivo experiments also highlighted the importance of
the carrier choice. For example, rhBMP9 (169.2 ng/ml)
embedded in chitosan can induce endochondral ossifica-
tion 24 days after injection in quadriceps of mice, while
BMP9 entrapped into collagen seems less efficient [58].
pBMP9, a peptide derived from the knuckle epitope of
BMP9, is also able to induce bone formation in quadric-
ceps when injected embedded in chitosan [58]. This un-
derlines the importance to optimize carrier and the in-
fluence of microenvironment, to improve BMP9 in vivo
bone induction. Together, these delivery systems can
offer promising approaches for the sustained delivery of
BMPs, such as BMP9 for tissue engineering applications
and for improving bone formation.
Another alternative to improve bone formation is to
develop biomimetic materials that favor cell adhesion
and promote their response to rhBMP9 or pBMP9. For
example, the functionalization of material by adhesive
peptides that target specific integrins can strongly influ-
ence the bone cell response to BMP9 and pBMP9 [60,
61]. The peptide derived from the bone sialoprotein Ac-
CGGNGERPRGDTYRAY-NH2 that is recognized by the
avb3 integrins promote the early differentiation of
MC3T3-E1 preosteoblasts induced by pBMP9. In con-
trast, pBMP9 had no effect on cells attached through the
a2b1 integrins to the peptides Ac-CGGDGEA-NH2 de-
rived from type I collagen [61]. The selection of adhesive
peptides to develop biomimetic materials promoting
bone cell response to BMPs is therefore critical.
4. BMP9 AND CHONDROGENESIS
Chondrogenesis is a multistep process that leads to the
formation of cartilage [62]. Understanding chondrogene-
sis is essential when addressing bone formation since
cartilage is part of bone development and repair [62].
The first step of chondrogenesis is the aggregation and
condensation of mensenchymal progenitor cells (MPCs)
to form chondroprogenitors [63,64]. Then, chondropro-
genitors differentiate into chondrocytes that proliferate
and produce matrix containing collagen II, IX, XI and
aggrecan [63,65]. The n ewly formed cartilage serves as a
template for the formation of long bone, a process called
endochondral ossification [63,66]. Indeed, chondrocytes
undergo a terminal differentiation to hypertrophic phe-
notype, followed by calcification of the cartilage matrix,
which leads to its vascularisation and its ossification [63].
Both intramembranous and endochondral bone formation
are associated to BMPs, but BMP9 was proposed to be
more related to endochondral bone formation [37]. Its
proposed implication in endochondral ossification, is
highlighting its major role in skeleton development,
maintenance and repair [35].
BMP signaling is essential during chon drogenesis; it is
required for the condensation of MPCs and their differ-
entiation into chondrocytes [64-66]. Kramer et al. have
shown that BMP2 (2 ng/ml; 10 ng/ml) and BMP-4 (10
ng/ml) are able to induce cho ndrogenic differentiation of
embryonic stem cells via embryonic bodies [67]. More
interesting ly, an increased mRNA expression of BMP1/2/
5/6 have been reported in human chondrocytes through-
out chondrogenic differentiation [68,69] and that adult
articular chondrocytes was able to express BMP7 [70].
BMP9 has been shown to induce ectopic bone formation
into skeletal muscle [56,71]. This occurs via an endo-
chondral bone process as shown by Varady et al. [71].
They noticed that six days after an intramuscular injec-
tion of AdBMP9 vector, MPCs had begun a differentia-
tion into primitive chondroblasts that secreted loose ex-
tracellular matrix [71]. Thirteen days post-injection, they
also observed hypertrophied chondrocytes. Three weeks
post-injection, woven bone appeared and turned into
lamellar bone within three months [71]. Majumdar et al.
showed that Sox9, a nuclear transcription factor required
to undergo chondrogenesis, was upregulated in BMP9
treated MPCs [72]. Furthermore, BMP9 treatment (100
ng/ml) also induced overexpression of Col2a1 and ag-
grecan after 2 weeks, two important components of the
cartilage matrix [72]. Finally, Blunk et al. have shown
that BMP9 was able to stimulate the proliferation of pri-
mary bovine chondrocytes and extracellular matrix de-
position at a concentration of more than 1 ng/ml and to
induce chondrocytes hypertrophy and mineralization
when the concentration was above 10 ng/ml [73]. All
these results strongly suggest that BMP9 is a modulator
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
36
of chondrocytes activity.
5. BMP9 AND BONE RESORPTION
BMP9, as described above, is involved in chondrogene-
sis and can strongly promote bone formation. In addition,
Fong et al. recently demonstrated in vitro that rhBMP9
can also augment bone resorption [74]. This increase was
shown to be functional and not related to osteoclast for-
mation. Furthermore, rhBMP9 could alter the intrinsic
apoptosis pathway and increase survival of osteoclasts.
The effect of rhBMP9 on osteoclast was explained by the
presence of ALK1 and BMPRII co-receptors and their
activation of the Smad 1/5/8 and non-smad MAPK/ERK
pathways. These results show for the first time that
BMP9 can directly affect human osteoclasts, acting on
their function and th eir survival [74].
6. BMP9 AND CANCER DEVELOPMENT
There are still discrepancies in studies concerning BMP9
and cancer development. Although some studies suggest
that BMP9 promoted differentiation and decreased pro-
liferation, whereas some others have shown the opposite.
Indeed, different studies have found that BMP9 restricts
tumor growth including prostate cancer, osteosarcoma
and colon adenocarcinoma [75-77]. The relation between
cancer and BMP9 seems to be mostly through two sepa-
rate mechanisms: cancer cell migration and invasion as
well as angiogenesis process [75-79].
Recent studies analyzed the effect of BMP9 (rhBMP9
or AdBMP9) in different human osteosarcoma cell lines
such as MG-63 cells [80,81]. For example, BMP9 ex-
pression was obtained through infection of human os-
teosarcoma MG-63 and 143B cells by a BMP9 express-
ing adenovirus [82]. Migration and invasion in vitro po-
tential were significantly lower (about 50% - 55%) in the
infected cells [82]. In a sub-cutaneous nude mice model,
the BMP9 overexpressing tumors were found to be sig-
nificantly smaller [77]. Then, BMP9 was found to induce
apoptosis in osteosarcoma cells [77]. Moreover matrix
metalloproteinases (MMP), specifically MMP-9, were
shown to be decreased significantly by the overexpres-
sion of BMP9 in osteosarcoma cells [82]. The loss of
migration and invasion potential was suggested to be
through a Smad-dependent pathway by downregulating
the expression and activity of MMP9 [77,82].
Furthermore, BMPs have been implicated in the pro-
duction of bone metastases in prostate cancer [83]. How-
ever, based on the observation that the expression of
BMP9 was decreased or absent in higher grade prostate
cancer, Ye et al. investigated the influence of BMP9 on
the biology of prostate cancer cells [76]. They showed
that the overexpression of BMP9 reduced cell growth,
matrix adhesion, invasion, and migration of prostate can-
cer PC-3 cells. They also showed that BMP9 induced
apoptosis in those particular cells through the overex-
pression of the prostate apoptosis response-4. The effect
of BMP9 on PC-3 cells relied on Smad-dependent signal
transduction. Thus, knockdown of BMPR-IB or BMPR-
II could promote PC-3 growth in vitro, but it had no sig-
nificant effect on their invasion potential. This suggests
that BMP9 may function as a tumor suppressor and
apoptosis regulator in prostate cancer through diverse
mechanisms [76].
Regarding angiogenesis, BMP9 has been shown to be
linked to vascular maturation [22]. However, contradic-
tory studies about the role of TGF-
and other family
members with reported vascular functions, such as BMP9,
in physiological and pathological angiogenesis make the
need for more comprehensive studies.
7. BMP9 AND ANGIOGENESIS
The effect of BMP9 on angiogenesis remains controver-
sial as both pro [79] and anti-angiogenic [22] effects
have been demonstrated. This discrepancy can be ex-
plained by different dosages, but also microenvironment
and the subtle interplay between vascular sprouting and
maturation [84]. The interest of studying BMP9 in an-
giogenesis resides in two principal pathologic angio-
genesis conditions: cancer angiogenesis and vascular
diseases per se (Hereditary Hemorrhagic Telangiectasia
(HHT), pre-eclampsia, pulmonary arterial hypertension
(PAH)).
BMP9 has been reported to bind to ALK1 in endothe-
lial cells [24]. It was also shown using rat aortic cross
sections that circulating BMP9 induces a constitutive
Smad1/5/8 phosphorylation in endothelial cells [21].
However, the roles of BMP9-ALK1 signaling in the
regulation of endothelial cells have not yet been fully
elucidated. Suzuki et al. examined the effects of BMP9
on the proliferation of endothelial cells. Vascular tube
formation from ex vivo mouse embryonic allantois ex-
plants and proliferation of in vitro cultured mouse stem
cell-derived endothelial cells (MESECs) was both pro-
moted by BMP9 [79]. This effect was shown to be re-
lated to ALK1 signaling , supported by the mimicked pro-
angiogenic effect of the expression of constitutively ac-
tive ALK1 in MESECs. Furthermore, in vivo angiogene-
sis was promoted by BMP9 in a Matrigel plug assay and
in a mouse xenograft model after subcutaneous injection
of BxPC3 human pancreatic adenocarcinoma cells in-
fected with lentiviruses enco ding BMP9. Taken together,
these results showed that BMP9 enhanced the prolifera-
tion of normal endoth elial cells and tumor-associated en-
dothelial cells [79].
Through genetic and pharmacological means, Cunha
demonstrated that ALK1 represents a new therapeutic
target for tumor angiogenesis [78]. Using a soluble fu-
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 37
sion protein of ALK1 with an immunoglobulin (ALK1-
Fc), they have been able to inhibit the BMP9 -ALK1 sig-
naling. Systemic treatment with the ALK1-Fc fusion pro-
tein RAP-041 (12 mg/kg), which acts as a competitive
inhibitor of BMP9, delayed pancreatic tumor growth and
progression, as diminution of ALK 1 ge ne d o s age also di d.
This delayed growth was thought to be related to the
inhibition of angiogenesis. Furthermore, the use of RAP-
041 significantly impaired angiogenic response toward
vascular endothelial growth factor A (VEGFA). In that
study, they found that the observed effect was related to
an unexpected synergy between TGF-β and BMP9. In
fact, the combined action of the two factors augmented
the response to angiogenic stimuli o f the endo thelial cells
[78]. This finding reinforces the idea that BMP9 can
have a dual effect on angiogenesis and that its pro-an-
giogenic effect may be dependent on TGF-
synergy.
The same year, another team showed a diminution of
vessel formation in a chick chorioallantoic assay and a
reduced tumor burden in both melanoma explants and
mammary adenocarcinomas with ALK1-Fc treatment
[85].
Later, Cunha et al. presented another paper on the
critical role for ALK1, reviewing its effect on the endo-
thelial phenotype in vitro and in vivo [25]. These findings
have led to the development of new ALK1 pathway in-
hibitors that target different malignant diseases. In the
same vein, another group used soluble endoglin ECD as
an inhibitor in the regulation of angiogenesis and high-
lighted the efficacy of this fusion protein as a potential
anti-angiogenic drug [75]. They showed that this inhibi-
tor reduced the VEGF-induced angiogenesis and the tu-
mor burden in a mouse colon cancer model.
Angiogenesis requires a finely tuned balance between
numerous signals and although BMP9 has been shown to
have pro-angiogenic potential (above), other studies have
shown the opposite. Indeed, a team has been working a
lot on the relationship between endothelial cells and
ALK1 [21,24,86]. Using NIH3T3 fibroblasts, found that
BMP9 signal transduction involved ALK1 and both
BMPRII and ActRIIA. Moreover, by overexpressing en-
doglin in NIH3T3 fibroblasts, they promoted the BMP9
stimulation of ALK1. BMP9 could inhibit endothelial
cell migration and growth, and activate the expression of
genes encoding proteins related to vascular maturation
such as Id1, Id2, Smad6, Smad7, endoglin and BMPRII
[24].
A year after, they reviewed the presence of BMP9 in
human serum to verify whether in fact it could be im-
plied in vascular quiescence. First, they found that hu-
man serum alone could indu ce Smad1/5 phosphorylatio n
in NIH3T3 transfected by ALK1 expression plasmid.
Second, they identified the active factor by using anti-
bodies agains t all BMP members and found that only the
anti-BMP9 inhibited this Smad1/5 phosphorylation [22].
These results were refined by data from Herrera and
Inman (2009) where they identified circulating BMP4
and 9 as active BMPs in human sera with a luciferase
reporter fused with the promoter BMP response element
Id1, transfected in a C2C12 line [87]. Present at a con-
centration between 2 and 12 ng/mL in human plasma,
BMP9 is circulating at a biologically active concentra-
tion (EC50 = 50 pg/mL). BMP9 was then tested in vivo in
angiogenic assays. Both in vivo mouse cellulose sponge
angiogenesis and chicken chorioallantoic membrane as-
says showed that BMP9 could negatively affect angio-
genesis may play a role in vascular quiescence [22]. In-
terestingly, the role of BMPs in angiogenesis opened the
door to a better understanding of genetic vascular dis-
eases caused by mutations in genes encoding for recap-
tors of this pathway (endoglin, ALK1 and BMPRII). This
leads to an important review in which the authors pro-
posed a model to distinguish BMP2, BMP7 and GDF5
from BMP9 in their functional implication in vessel for-
mation and maturation [84].
Hereditary Hemorrhagic Telangiectasia (HHT), also
known as Rendu-Osler-Weber, is a disease that causes
frequent nose bleeds, mucocutaneous and visceral te-
langiectasia [88]. This pathology follows an autosomal-
dominant inheritance pattern which has been shown to be
linked in 80% to a mutation in ALK1 or the co-receptor
endoglin [89]. Endoglin is a tran smembrane glycoprotein
on human vascular endothelial cells and is a co-receptor
implicated in this dominant vascular dysplasia as well as
in preeclampsia [90]. The alteration of ALK1 or endoglin
affects the maturation phase of angiogenesis, leading to
the absence of capillaries connecting the arterial and ve-
nous networks [89]. As BMP9 acts as the specific ligand
of the receptor ALK1 and to endoglin as its co-receptor,
a defect in either members of the receptor complex
means a disrupted BMP9 signaling pathway. Heterozy-
gous mice for endoglin or ALK1 supports the involve-
ment of endothelial hyperproliferation in the pathogene-
sis of the disease while the maturation is disrupted [91].
More recently, Choi et al. demonstrated that ALK1 defi-
cient endothelial cells have high migratory and invasion
properties both in vitro and in vivo. This new data con-
firms the importance of BMP9-ALK1 for angiogenesis
and into the differentiation and maturation of neocapil-
laries [92]. This BMP9-ALK1 signaling in angiogenesis
was shown to be transmitted downstream through Tmem
100, a gene encoding a previously uncharacterized intra-
cellular transmembrane protein [93].
On the other hand, pulmonary arterial hypertension
(PAH) is characterized by dysregulation in pulmonary
artery endothelial cell proliferation, apoptosis and per-
meability. The most common cause of inheritable PAH is
a mutation in the BMPR-II, leading to a loss of its func-
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
38
tion [94]. In the same way as the defective signaling of
ALK1-endoglin in HHT, a deficient BMPRII signaling
leads to an absent function of BMP9 and to an anarchic
development of blood vessels [94].
Recently, it has been shown that chloroquine, an anti-
malarial drug, is able to indirectly increase cell surface
expression of BMPRII in pulmonary artery endothelial
cells [95]. This increase seemed to be independent from
transcription since chloroquine enhanced BMPRII ex-
pression even after siRNA knockdown of the BMPRII
transcript. Using BMPRII deficient cells from PAH sick
patients, BMP9 signaling was also shown to be decreas-
ed and treatment with chloroquine significantly increased
expression of BMP9-BMPRII signaling gene targets.
These findings prov ide ne w insigh ts for the restor atio n of
cell surface receptors in treating hereditary vascular dis-
eases, specifically BMPR-II with chloroquine for treat-
ment of PAH [95].
The effect of BMP9 on angiogenesis remains contro-
versial as both pro [79] and anti-angiogenic [22] effects
have been demonstrated. This discrepancy can be ex-
plained by different dosages, but also microenvironment
and the subtle interplay between vascular sprouting and
maturation [84]. The interest of studying BMP9 in an-
giogenesis resides in two principal pathologic angioge-
nesis conditions: cancer angiogenesis and vascular dis-
eases per se (Hereditary Hemorrhagic Telangiectasia
(HHT), pre-eclampsia, pulmonary arterial hypertension
(PAH)).
BMP9 has been reported to bind to ALK1 in endothe-
lial cells [24]. It was also shown using rat aortic cross
sections that circulating BMP9 induces a constitutive
Smad1/5/8 phosphorylation in endothelial cells [21].
However, the roles of BMP9-ALK1 signaling in the
regulation of endothelial cells have not yet been fully
elucidated. Suzuki et al. examined the effects of BMP9
on the proliferation of endothelial cells. Vascular tube
formation from ex vivo mouse embryonic allantois ex-
plants and proliferation of in vitro cultured mouse stem
cell-derived endothelial cells (MESECs) was both pro-
moted by BMP9 [79]. This effect was shown to be re-
lated to ALK1 signaling , supported by the mimicked pro-
angiogenic effect of the expression of constitutively ac-
tive ALK1 in MESECs. Furthermore, in vivo angiogene-
sis was promoted by BMP9 in a Matrigel plug assay and
in a mouse xenograft model after subcutaneous injection
of BxPC3 human pancreatic adenocarcinoma cells in-
fected with lentiviruses enco ding BMP9. Taken together,
these results showed that BMP9 enhanced the prolifera-
tion of normal endoth elial cells and tumor-associated en-
dothelial cells [79].
Through genetic and pharmacological means, Cunha
demonstrated that ALK1 represents a new therapeutic
target for tumor angiogenesis [78]. Using a soluble fu-
sion protein of ALK1 with an immunoglobulin (ALK1-
Fc), they have been able to inhibit the BMP9 -ALK1 sig-
naling. Systemic treatment with the ALK1-Fc fusion pro-
tein RAP-041 (12 mg/kg), which acts as a competitive
inhibitor of BMP9, delayed pancreatic tumor growth and
progression, as diminution of ALK 1 ge ne d o s age also di d.
This delayed growth was thought to be related to the in-
hibition of angiogenesis. Furthermore, the use of RAP-
041 significantly impaired angiogenic response toward
vascular endothelial growth factor A (VEGFA). In that
study, they found that the observed effect was related to
an unexpected synergy between TGF-β and BMP9. In
fact, the combined action of the two factors augmented
the response to angiogenic stimuli o f the endo thelial cells
[78]. This finding reinforces the idea that BMP9 can
have a dual effect on angiogenesis and that its pro-an-
giogenic effect may be dependent on TGF-b synergy.
The same year, another team showed a diminution of
vessel formation in a chick chorioallantoic assay and a
reduced tumor burden in both melanoma explants and
mammary adenocarcinomas with ALK1-Fc treatment
[85].
Later, Cunha et al. presented another paper on the
critical role for ALK1, reviewing its effect on the endo-
thelial phenotype in vitro and in vivo [25]. These findings
have led to the development of new ALK1 pathway in-
hibitors that target different malignant diseases. In the
same vein, another group used soluble endoglin ECD as
an inhibitor in the regulation of angiogenesis and high-
lighted the efficacy of this fusion protein as a potential
anti-angiogenic drug [75]. They showed that this inhibi-
tor reduced the VEGF-induced angiogenesis and the tu-
mor burden in a mouse colon cancer model.
Angiogenesis requires a finely tuned balance between
numerous signals and although BMP9 has been shown to
have pro-angiogenic potential (above), other studies have
shown the opposite. Indeed, a team has been working a
lot on the relationship between endothelial cells and
ALK1 [21,24,86]. Using NIH3T3 fibroblasts, found that
BMP9 signal transduction involved ALK1 and both
BMPRII and ActRIIA. Moreover, by overexpressing en-
doglin in NIH3T3 fibroblasts, they promoted the BMP9
stimulation of ALK1. BMP9 could inhibit endothelial
cell migration and growth, and activate the expression of
genes encoding proteins related to vascular maturation
such as Id1, Id2, Smad6, Smad7, endoglin and BMPRII
[24].
A year after, they reviewed the presence of BMP9 in
human serum to verify whether in fact it could be im-
plied in vascular quiescence. First, they found that hu-
man serum alone could indu ce Smad1/5 phosphorylatio n
in NIH3T3 transfected by ALK1 expression plasmid. Se-
cond, they identified th e active factor by using an tibod ies
against all BMP members and found that only the anti-
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 39
BMP9 inhibited this Smad1/5 phosphorylation [22].
These results were refined by data from Herrera and
Inman (2009) where they identified circulating BMP4
and 9 as active BMPs in human sera with a luciferase re-
porter fused with the promoter BMP response element
Id1, transfected in a C2C12 line [87]. Present at a con-
centration between 2 and 12 ng/mL in human plasma,
BMP9 is circulating at a biologically active concentra-
tion (EC50 = 50 pg/mL). BMP9 was then tested in vivo
in angiogenic assays. Both in vivo mouse cellulose
sponge angiogenesis and chicken chorioallantoic mem-
brane assays showed that BMP9 could negatively affect
angiogenesis may play a role in vascular quiescence [22].
Interestingly, the role of BMPs in angiogenesis opened
the door to a better understanding of genetic vascular
diseases caused by mutations in genes encoding for re-
ceptors of this pathway (endoglin, ALK1 and BMPRII).
This lead to an important review in which the authors
proposed a model to distinguish BMP2, BMP7 and
GDF5 from BMP9 in their functional implication in ves-
sel formation and maturation [84].
Hereditary Hemorrhagic Telangiectasia (HHT), also
known as Rendu-Osler-Weber, is a disease that causes
frequent nose bleeds, mucocutaneous and visceral te-
langiectasia [88]. This pathology follows an autosomal-
dominant inheritance pattern which has been shown to be
linked in 80% to a mutation in ALK1 or the co-receptor
endoglin [89]. Endoglin is a tran smembrane glycoprotein
on human vascular endothelial cells and is a co-receptor
implicated in this dominant vascular dysplasia as well as
in preeclampsia [90]. The alteration of ALK1 or endoglin
affects the maturation phase of angiogenesis, leading to
the absence of capillaries connecting the arterial and ve-
nous networks [89]. As BMP9 acts as the specific ligand
of the receptor ALK1 and to endoglin as its co-receptor,
a defect in either members of the receptor complex
means a disrupted BMP9 signaling pathway. Heterozy-
gous mice for endoglin or ALK1 supports the involve-
ment of endothelial hyperproliferation in the pathogene-
sis of the disease while the maturation is disrupted [91].
More recently, Choi et al. demonstrated that ALK1 defi-
cient endothelial cells have high migratory and invasion
properties both in vitro and in vivo. This new data con-
firms the importance of BMP9-ALK1 for angiogenesis
and into the differentiation and maturation of neocapil-
laries [92]. This BMP9-ALK1 signaling in angiogenesis
was shown to be transmitted downstream through Tmem
100, a gene encoding a previously uncharacterized intra-
cellular transmembrane protein [93].
On the other hand, pulmonary arterial hypertension
(PAH) is characterized by dysregulation in pulmonary
artery endothelial cell proliferation, apoptosis and per-
meability. The most common cause of inheritable PAH is
a mutation in the BMPR-II, leading to a loss of its func-
tion [94]. In the same way as the defective signaling of
ALK1-endoglin in HHT, a deficient BMPRII signaling
leads to an absent function of BMP9 and to an anarchic
development of blood vessels [94].
Recently, it has been shown that chloroquine, an anti-
malarial drug, is able to indirectly increase cell surface
expression of BMPRII in pulmonary artery endothelial
cells [95]. This increase seemed to be independent from
transcription since chloroquine enhanced BMPRII ex-
pression even after siRNA knockdown of the BMPRII
transcript. Using BMPRII deficient cells from PAH sick
patients, BMP9 signaling was also shown to be decreas-
ed and treatment with chloroquine significantly increased
expression of BMP9-BMPRII signaling gene targets.
These findings prov ide ne w insigh ts for the restor atio n of
cell surface receptors in treating hereditary vascular dis-
eases, specifically BMPR-II with chloroquine for treat-
ment of PAH [95].
Indeed, there still remain s contro v ersy on pro and an ti-
angiogenic effects of BMP9. Recent literature highlights
the importance of understanding its role in vessel matu-
ration as opposed to formation and also the role of the
microenvironment and synergy with TGF-b. On the other
hand, it is clear that the signaling pathway is through re-
ceptor and co-receptor ALK1-endoglin, and that this can
be inhibited by the use of ALK1 or endoglin fusion pro-
teins. Certainly, it will be interesting to test whether
some drugs derived from inhibitors like ALK1-Fc could
be used experimentally to treat cancer and on the other
hand develop ALK1 pathway enhancers that could help
to treat hereditary vascular diseases.
8. BMP9 AND NEUROGENESIS
BMPs are critical in neurogenesis, forebrain formation
and more globally in the dorsov entral axis determination
[96,97]. BMPs promote cell survival and differentiation
of neurons in both peripheral and central nervous sys-
tems (CNS) [98] Lopez-Coviella et al. have been work-
ing specifically on the cholinergic neurons of the fore-
brain [99]. Basal forebrain cholinergic neurons (BFCN)
participate in processes of learning, memory, attention
and their degeneration is involved in several human dis-
orders such as Alzheimer’s disease [100]. They have
shown that 14 days old mice embryos express BMP9
mRNA principally in the forebrain and spine [99]. This
developmental stage is also the one where the cholinergic
neurons are most sensitive to BMP9. Thus, cultured pri-
mary cells derived from the septal area increase their
acetylcholine (Ach) expression by 20 fold when exposed
to BMP9 during that period. Compared to 5 other BMPs
(BMP2-4-6-7-12), BMP9 is the most potent in inducing
Ach expression by septal cells. Moreover, when injected
into mouse embryo cerebral ventricles, BMP9 is able to
increase cerebral Ach level [99]. The genes responsive to
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
40
this BMP9 induction of basal forebrain neurons include
cell-cycle related proteins, transcription factors, signal
transduction molecules, ex tracellular matrix proteins, ad-
hesion molecules, enzymes, transporters, and chaperon-
ins. The set of genes induced with BMP9 treatment sug-
gests that BMP9 creates a trophic environment for basal
forebrain neurons and contributes to the maturation of
this part of the brain in vivo [ 101].
During the development of cholinergic neurons of the
basal forebrain, BMP9 activates the canonical BMP sig-
naling pathway, through phosphorylation of Smad1 and
Smad5 which associate to co-Smad4 and translocate into
the nucleus [98]. Finally, this team demonstrated that
BMP9 administration can prevent lesion-evoked impair-
ment of the cholinergic hippocampal neurons in adult
mice by inducing a favorable environment for these cells
[102].
Interestingly, Bissonnette et al. have been able to gen-
erate BFCN from human embryonic stem cells (hESCs)
using relevant ligands of the developing forebrain [103].
hESCs respond to BMP9 signaling after sonic hedge-
hog/fibroblast growth factor 8 pretreatment and differen-
tiate the progenitors toward a BFCN phenotype. These
derived neurons express only markers characteristic of
BFCN and have their functional characteristics (action
potentials, functional cholinergic synapses). The ability
to derive BFCN from hESCs is a significant step for the
development of therapeutic interventions in diseases af-
fecting the forebrain (e.g. Alzheimer’s disease) [103].
BMP9 expression in the CNS seems to be related to
the development and maintenance of the cholinergic phe-
notype. The cited studies give new insights on a clearer
comprehension of the role of BMP9 during early embry-
ology and the later ability of damage repair. The proven
expression of BMP9 in CNS is also interesting for dif-
ferent reasons: the relation of brain/spinal cord injury
and increased ossification potential. As shown in many
papers, CNS injury (brain and spinal cord) is associated
with an increase in ossification potential (bone healing
and HO) [104-109]. However, no specific growth factor
has been identified to date [105]. BMP9 would be an in-
teresting candidate to evaluate.
9. BMP9 AND HEPATOCYTES
Since BMP9 was first described to be expressed in non
parenchymal cells of the liver [20,45], studies have been
conducted to determine the effect of BMP9 on liver cells
and hepatocellular regeneration [110]. Miller et al. have
determined the binding affinity of BMP9 and its number
of receptors on rat hepatic endothelial cells (Kd of ~85
pM, 3600 receptors/cell) and Kupffer cells (Kd of ~25
pM, 1100 receptors/cell), cells identified as main BMP9
producers. They also found that the binding of BMP9 on
these cells was reversible and BMP9/recep tor complexes
can be internalized [20].
However, while Miller et al. did not detect BMP9 in
rat hepatocytes [20], Bidart et al. recently found that
BMP9 expression (mRNA and protein) was mainly lo-
cated in human h epatocytes and biliary hepatic ep ithelial
cells [21]. The effect of BMP9 on hepatocytes was first
evaluated by Song et al. [45]. They found that rhBMP9
bind to human HepG2 liver tumor cells with high affinity.
They also found that HepG2 cells express two BMP9
receptors similar in size to the Type I and Type II recap-
tors reported by others for TGF-
members, but specific
for BMP9 [45]. Increasing doses (0.1 - 10 ng/mL) of
BMP9 stimulated human HepG2 and primary rat heap-
tocytes cell proliferation. In contrast, TGF-
(0.1 - 100
ng/mL) had no effect on HepG2 cell proliferation but
inhibited proliferation in primary hepatocytes [45]. This
study was the first to report the effect of BMP9 on heap-
tocyte proliferation. The direct effect of BMP9 on he-
patic progenitor cells was also studied through infection s
of progenitors with a recombinant adenovirus containing
human BMP9 gene (AdBMP9) by Gao et al. (2011). The
maturation and differentiation of progenitor cells were
examined. AdBMP9 enhanced glycogen storage as well
as albumin production in hepatic progenitor cells, a ma-
ture hepatic cell behavior [111].
BMP9 is expressed in the liver and seems to have an
effect on hepatocytes, but more studies are needed to cla-
rify its differential effect over proliferation [45] and dif-
ferentiation [111] as well as its neoplastic influence on
liver cells [110].
10. BMP9 AND GLUCOSE METABOLISM
Insulin resistance is a systemic multifactorial impair ment
of glucose uptake. Muscle, the most important glucose
consuming organ, need Akt2 to be able to activate insu-
lin-induced glucose uptake and this pathway seems to be
severely impaired in insulin resistance [112,113]. Inter-
estingly, a combination of bioinformatic and high-
throughput functional analyses have shown BMP9 to be
the first hepatic factor to regulate blood glucose concen-
tration [46]. Moreover, this effect was thought to be me-
diated by activation of Akt kinase in differentiated myo-
tubes [46]. Then, it has been demonstrated that recombi-
nant BMP9 (1 and 5 mg/kg) improves glucose homeo-
stasis in vivo in diabetic and non-diabetic rodents [46].
The mechanism relied on the upregulation of Smad5 and
Akt2 in differentiated rats myotubes [114]. On the oppo-
site side, Smad5 was downregulated in myotubes by de-
xamethasone, a well known hyperglycemia inducer and
Smad5 knockdown in rats decreased Akt2 expression and
phosphorylation leading to a decrease in insulin-induced
glucose uptake by myotubes. It was then hypothesized
that Smad5 regulated glucose uptake in skeletal muscle
through Akt2 expression and phosphorylation. These
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 41
findings also revealed Smad5 as a potential target for the
treatment of type 2 diabetes. Hence, BMP9 could be seen
as a potential activator of Smad5 for that purpose [114].
This promising effect of BMP9 could lead to a better
control on hepatic glucose, furthermore on diabetes. In-
deed, BMP9 expression is severely reduced in the liver
of insulin-resistant rats [115]. In this study, the authors
have been reporting several assays showing that BMP9
directly controls glucose homeostasis [115]. Direct in
situ exposition of normal fasting rats-liver to glucose an d
insulin leads to BMP9 expression. Moreover, neutraliza-
tion of BMP9 with an anti-BMP9 antibody induces glu-
cose intolerance and insulin resistance. Analyzed toge-
ther, these results reinforce the eligibility of BMP9 as a
hepatic insulin-sensitizing substance (HISS) in glucose
homeostasis physiology [115].
11. CONCLUSION
BMP9 is a powerful osteogenic growth factor and was
shown to be able to activate bone differentiation in many
animal studies. The osteogenic potential of BMP9 was
first studied through gene therapy, but nowadays it is
used through its recombinant human form, rhBMP9. The
study of BMP9 bone formation has led to a better under-
standing on endochondral bone healing, but also of the
new insights in delivery systems that can enhance its
effect as well as improved comprehension of the trau-
matic HO and its relation to BMP9. As bone formation is
always associated with resorption and remodeling, it has
also been shown that BMP9 can directly affect human
osteoclasts, acting on their function and their survival. In
cancer development, BMP9 has two major effects. First
of all, its anti-proliferative/pro-differentiation potential
seems to be protective for many neoplastic conditions.
On the other hand, its inhibition through ALK1 and en-
doglin fusion proteins seemed to decrease tumor angio-
genesis. Therefore, more studies are warranted in order
to clarify its influence on tumorigenesis. On the angio-
genic side, we have discussed the sometimes confusing
contradictions about the pro and anti-angiogenic effects
of BMP9. We have presented the importance of under-
standing BMP9’s role in blood vessel maturation as op-
posed to formation. The importance of BMP9 in the cen-
tral nervous system resides in its ability to promote cho-
linergic neurons proliferation and survival. Looking for
new innovative treatment paths in Alzheimer’s disease,
BMP9 could be an interesting candidate. The relation
between liver homeostasis and BMP9 is still not clear,
but it is now to our knowledge that it is expressed and
produced in that organ. Some studies have tried to show
its hepatocellular regeneration potential, but still, more
studies are needed. Finally, we have discussed its com-
pelling effect in glucose metabolism, principally pro-
moting sensitivity to insuline in skeletal muscle. Thus,
BMP9 is a maj or memb er of th e TGF-
superfamily that
is implied in many fundamental developmental and pa-
thologic processes. Future research will certainly bring
answers to the many questions left open, and those an-
swers will unquestionably lead to clinical applications.
12. ACKNOWLEDGEMENTS
We are grateful to Dr Juan Sebastian Rendon who did a critical reading
of this review anf helped for corrections.
REFERENCES
[1] Urist, M.R. (1965) Bone: Formation by autoinduction.
Science, 150, 893-899.
http://dx.doi.org/10.1126/science.150.3698.893
[2] NCBI. GDF2 growth differe ntiation 2 [Homo sapiens (hu-
man)], Gene, NCBI.
http://www.ncbi.nlm.nih.gov/gene/2658
[3] Kishigami, S. and Mishina, Y. (2005) BMP signaling and
early embryonic patterning. Cytokine Growth Factor Re-
views, 16, 265-278.
[4] Israel, D.I. , Nove, J., Kerns, K.M., Kaufman, R.J., Rosen,
V., Cox, K.A. and Wozney, J.M. (1996) Heterodimeric
bone morphogenetic proteins show enhanced activity in
vitro and in vivo. Growth Factors, 13, 291-300.
http://dx.doi.org/10.3109/08977199609003229
[5] Aono, A., Hazama, M., Notoya, K., Taketomi, S., Yama-
saki, H., Tsukuda, R., Sasaki, S. and Fujisawa, Y. (1995)
Potent ectopic bone-inducing activity of bone morpho-
genetic protein-4/7 heterodimer. Biochemical and Bio-
physical Research Communications, 210, 670-677.
http://dx.doi.org/10.1006/bbrc.1995.1712
[6] Miyazono, K., Maeda, S. and Imamura, T. (2005) BMP
receptor signaling: Transcriptional targets, regulation of
signals, and signaling cross-talk. Cytokine Growth Factor
Reviews, 16, 251-263.
http://dx.doi.org/10.1016/j.cytogfr.2005.01.009
[7] Mazerbourg, S. and Hsueh, A.J. (2006) Genomic analys-
es facilitate identification of receptors and signalling
pathways for growth differentiation factor 9 and related
orphan bone morphogenetic protein/growth differentia-
tion factor ligands. Human Reproduction Update, 12,
373-383. http://dx.doi.org/10.1093/humupd/dml014
[8] Bessa, P.C., Casal, M. and Reis, R.L. (2008) Bone mor-
phogenetic proteins in tissue engineering: The road from
the laboratory to the clinic, part I (basic concepts). Jour-
nal of Tissue Engineering and Regenerative Medicine, 2,
1-13. http://dx.doi.org/10.1002/term.63
[9] Wotton, K.R., Alcaine Colet, A., Jaeger, J. and Jimenez-
Guri, E. (2013) Evolution and expression of BMP genes
in flies. Development Genes and Evolution, 223, 335-340.
http://dx.doi.org/10.1007/s00427-013-0445-9
[10] Kitisin, K., Saha, T., Blake, T., Golestaneh, N., Deng, M.,
Kim, C., Tang, Y., Shetty, K., Mishra, B. and Mishra, L.
(2007) Tgf-Beta signaling in development. Science STKE,
399, cm1. http://dx.doi.org/10.1126/stke.3992007cm1
[11] Chen, D., Zhao, M. and Mundy, G.R. (2004) Bone mor-
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
42
phogenetic proteins. Growth Factors, 22, 233-241.
http://dx.doi.org/10.1080/08977190412331279890
[12] Bergeron, E., Senta, H., Mailloux, A., Park, H., Lord, E.
and Faucheux, N. (2009) Murine preosteoblast differen-
tiation induced by a peptide derived from bone morpho-
genetic proteins-9. Tissue Eng Part A, 15, 3341-3349.
http://dx.doi.org/10.1089/ten.tea.2009.0189
[13] Kang, Q., Song, W.X., Luo, Q., Tang, N., Luo, J., Luo,
X., et al. (2009) A comprehensive analysis of the dual
roles of BMPs in regulating adipogenic and osteogenic
differentiation of mesenchymal progenitor cells. Stem
Cells and Development, 18, 545-559.
http://dx.doi.org/10.1089/scd.2008.0130
[14] Kang, Q., Sun, M.H., Cheng, H., Peng, Y., Montag, A.G.,
Deyrup, A.T., et al., (2004) Characterization of the dis-
tinct orthotopic bone-forming activity of 14 BMPs using
recombinant adenovirus-mediated gene delivery. Gene
Therapy, 11, 1312-1320.
http://dx.doi.org/10.1038/sj.gt.3302298
[15] Cheng, H., Jiang, W., Phillips, F.M., Haydon, R.C., Peng,
Y., Zhou, L., et al. (2003) Osteogenic activity of the
fourteen types of human bone morphogenetic proteins
(BMPs). The Journal of Bone & Joint Surgery, 85-A,
1544-1552.
[16] Peng, Y., Kang, Q., Cheng, H., Li, X., Sun, M.H., Jiang,
W., et al. (2003) Transcriptional characterization of bone
morphogenetic proteins (BMPs)-mediated osteogenic sig-
naling. Journal of Cellular Biochemistry, 90, 1149-1165.
http://dx.doi.org/10.1002/jcb.10744
[17] Li, J.Z., Hankins, G.R., Kao, C., Li , H., Kamma uff, J. and
Helm, G.A. (2003) Osteogenesis in rats induced by a no-
vel recombinant helper-dependent bone morphogenetic
protein-9 (BMP-9) adenovirus. The Journal of Gene Me-
dicine, 5, 748-756.
http://dx.doi.org/10.1002/jgm.412
[18] Wang, Y., Hong, S., Li, M., Zhang, J., Bi, Y., He, Y., et
al. (2013) Noggin resistance contributes to the potent
osteogenic capability of BMP9 in mesenchymal stem
cells. Journal of Orthopaedic Research.
http://dx.doi.org/10.1002/jor.22427
[19] van der Horst, G., van Bezooijen, R.L., Deckers, M.M.,
Hoogendam, J., Visser, A., Lowik, C.W. and Karperien,
M. (2002) Differentiation of murine preosteobla stic KS483
cells depends on autocrine bone morphogenetic protein
signaling during all phases of osteoblast formation. Bone,
31, 661-669.
http://dx.doi.org/10.1016/S8756-3282(02)00903-1
[20] Miller, A.F., Harvey, S.A., Thies, R.S. and Olson, M.S.
(2000) Bone morphogenetic protein-9. An autocrine/pa-
racrine cytokine in the liver. The Journal of Biological
Chemistry, 275, 17937-17945.
http://dx.doi.org/10.1074/jbc.275.24.17937
[21] Bidart, M., Ricard, N., Levet, S., Sa mson, M., Mallet , C.,
David, L., Subileau, M., Tillet, E., Feige, J.J. and Bailly,
S. (2012) BMP9 is produced by hepatocytes and cir-
culates mainly in an active mature form complexed to its
prodomain. Cellular and Molecular Life Sciences, 69,
313-324. http://dx.doi.org/10.1007/s00018-011-0751-1
[22] David, L., Mallet, C., Keramidas, M., Lamande, N., Gasc,
J.M., Dupuis-Girod, S., Plauchu, H., Feige, J.J. and Bailly,
S. (2008) Bone morphogenetic protein-9 is a circulating
vascular quiescence factor. Circulation Research, 102,
914-922.
http://dx.doi.org/10.1161/CIRCRESAHA.107.165530
[23] Brown, M.A., Zhao, Q., Baker, K.A., Naik, C., Chen, C.,
Pukac, L., et al. (2005) Crystal structure of BMP-9 and
functional interactions with pro-region and receptors. The
Journal of Biological Chemistry, 280, 25111-25118.
http://dx.doi.org/10.1074/jbc.M503328200
[24] David, L., Mallet, C., Mazerbourg, S., Feige, J.J. and
Bailly, S. (2007) Identification of BMP9 and BMP10 as
functional activators of the orphan activin receptor-like
kinase 1 (ALK1) in endothelial cells. Blood, 109, 1953-
1961. http://dx.doi.org/10.1182/blood-2006-07-034124
[25] Cunha, S.I. and Pietras, K. (2011) ALK1 as an emerging
target for antiangiogenic therapy of cancer. Blood, 117,
6999-7006.
http://dx.doi.org/10.1182/blood-2011-01-330142
[26] Attisano, L., Carcamo, J., Ventura, F., Weis, F.M., Mas-
sague, J. and Wrana, J.L. (1993) Identification of human
activin and TGF beta type I receptors that form hetero-
meric kinase complexes with type II receptors. Cell, 75,
671-680.
http://dx.doi.org/10.1016/0092-8674(93)90488-C
[27] Lamplot, J.D., Qin, J., Nan, G. , Wang, J., Liu, X., Yin, L.,
et al. (2013) BMP9 signaling in stem cell differentiation
and osteogenesis. American Journal of Stem Cells, 2, 1-
21.
[28] Bandyopadhyay, A., Yadav, P.S. and Prashar, P. (2013)
BMP signaling in development and diseases: A pharma-
cological perspective. Biochemical Pharmacology, 85,
857-864. http://dx.doi.org/10.1016/j.bcp.2013.01.004
[29] Kim, S.I., Kwak, J.H., Zachariah, M., He, Y., Wang, L.
and Choi, M.E. (2007) TGF-beta-activated kinase 1 and
TAK1-binding protein 1 cooperate to mediate TGF-be-
ta1-induced MKK3-p38 MAPK activation and stimula-
tion of type I collagen. AJP: Renal Physiology, 292, F1471-
F1478.
[30] Luo, J., Tang, M., Huang, J., He, B.C., Gao, J.L., Chen,
L., et al. (2010) TGF{beta}/BMP type I receptors ALK1
and ALK2 are essential for BMP9-induced osteogenic
signaling in mesenchymal stem cells. The Journal of Bi-
ological Chemistry.
[31] Townson, S.A., Martinez-Hackert, E., Greppi, C., Low-
den, P., Sako, D., Liu, J., et al. (2012) Specificity and
structure of a high affinity activin receptor-like kinase 1
(ALK1) signaling complex. The Journal of Biological
Chemistry, 287, 27313-27325.
http://dx.doi.org/10.1074/jbc.M112.377960
[32] Luther, G., Wagner, E.R., Zhu, G., Kang, Q., Luo, Q.,
Lamplot, J., et al. (2011) BMP-9 induced osteogenic dif-
ferentiation of mesenchymal stem cells: Molecular me-
chanism and therapeutic potential. Current Gene Therapy,
11, 229-240.
http://dx.doi.org/10.2174/156652311795684777
[33] Huang, E., Zhu, G., Jiang, W., Yang, K., Gao, Y., Luo,
Q., et al. (2012) Growth hormone synergizes with BMP9
in osteogenic differentiation by activating the JAK/STAT/
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 43
IGF1 pathway in murine multilineage cells. Journal of
Bone and Mineral Research, 27, 1566-1575.
http://dx.doi.org/10.1002/jbmr.1622
[34] Liu, X., Qin, J., Luo, Q., Bi, Y., Zhu, G., Jiang, W., et al.
(2013) Cross-talk between EGF and BMP9 signalling
pathways regulates the osteogenic differentiation of me-
senchymal stem cells. Journal of Cellular and Molecular
Medicine. http://dx.doi.org/10.1111/jcmm.12097
[35] Suttapreyasri, S., Koontongkaew, S., Phongdara, A. and
Leggat, U. (2006) Expression of bone morphogenetic pro-
teins in normal human intramembranous and endochon-
dral bones. International Journal of Oral and Maxillo-
facial Surgery, 35, 444-452.
http://dx.doi.org/10.1016/j.ijom.2006.01.021
[36] Celeste, A.J., Song, J.J., Cox, K.A., Rosen, V. and
Wozney, J.M. (1994) Bone morphogenetic protein-9, a
new member of the TGF beta superfamily. Journal of
Bone and Mineral Research, 9, 136.
[37] Alden, T.D., Beres, E.J., Laurent, J.S., Engh, J.A., Das, S.,
London, S.D., Jane Jr., J.A., Hudson, S.B. and Helm, G.A.
(2000) The use of bone morphogenetic protein gene the-
rapy in craniofacial bone repair. Journal of Craniofacial
Surgery, 11, 24-30.
http://dx.doi.org/10.1097/00001665-200011010-00005
[38] Epstein, N.E. (2011) Pros, cons, and costs of INFUSE in
spinal surgery. Surgical Neurology International, 2, 10.
http://dx.doi.org/10.4103/2152-7806.76147
[39] Helm, G.A., Alden, T.D., Beres, E.J., Hudson, S.B., Das,
S., Engh, J.A., Pittman, D.D., Kerns, K.M. and Kallmes,
D.F. (2000) Use of bone morphogenetic protein-9 gene
therapy to induce spinal arthrodesis in the rodent. Journal
of Neurosurgery, 92, 191-196.
http://dx.doi.org/10.3171/spi.2000.92.2.0191
[40] Dumont, R.J., Dayoub, H., Li, J.Z., Dumont, A.S.,
Kallmes, D.F., Hankins, G.R. and Helm, G.A. (2002) Ex
vivo bone morphogenetic protein-9 gene therapy using
human mesenchymal stem cells induces spinal fusion in
rodents. Neurosurgery, 51, 1239-1244.
[41] Dayoub, H., Dumont, R.J., Li, J.Z., Dumont, A.S.,
Hankins, G.R., Kallmes, D.F. and Helm, G.A. (2003) Hu-
man mesenchymal stem cells transduced with recombi-
nant bone morphogenetic protein-9 adenovirus promote
osteogenesis in rodents. Tissue Engineering, 9, 347-356.
http://dx.doi.org/10.1089/107632703764664819
[42] Abdelaal, M.M., Tholpady, S.S., Kessler, J.D., Morgan,
R.F. and Ogle, R.C. (2004) BMP-9-transduced prefabri-
cated muscular flaps for the treatment of bony defects.
Journal of Craniofacial Surgery, 15, 736-741.
http://dx.doi.org/10.1097/00001665-200409000-00007
[43] Aslan, H., Zilberman, Y., Arbeli, V., Sheyn, D., Matan,
Y., Liebergall, M., Li, J.Z., Helm, G.A., Gazit, D. and
Gazit, Z. (2006) Nucleofection-based ex vivo nonviral
gene delivery to human stem cells as a platform for tissue
regeneration. Tissue Engineering, 12, 877-889.
http://dx.doi.org/10.1089/ten.2006.12.877
[44] Itakura, K., Hirose, T., Crea, R., Riggs, A.D., Heyneker,
H.L., Bolivar, F. and Boyer, H.W. (1977) Expression in
Escherichia coli of a chemically synthesized gene for the
hormone somatostatin. Science, 198, 1056-1063.
http://dx.doi.org/10.1126/science.412251
[45] Song, J.J., Celeste, A.J., Kong, F.M., Jirtle, R.L., Rosen,
V. and Thies, R.S. (1995) Bone morphogenetic protein-9
binds to liver cells and stimulates proliferation. Endocri-
nology, 136, 4293-4297.
http://dx.doi.org/10.1210/en.136.10.4293
[46] Chen, C., Grzegorzewski, K.J., Barash, S., Zhao, Q.,
Schneider, H., Wang, Q., et al. (2003) An integrated
functional genomics screening program reveals a role for
BMP-9 in glucose homeostasis. Nature Biotechnology, 21,
294-301.
[47] Bessa, P.C., Cerqueira, M.T., Rada, T., Gomes, M.E.,
Neves, N.M., Nobre, A., Reis, R.L. and Casal, M. (2009)
Expression, purification and osteogenic bioactivity of re-
combinant human BMP-4, -9, -10, -11 and -14. Protein
Expression and Purification, 63, 89-94.
http://dx.doi.org/10.1016/j.pep.2008.09.014
[48] Bergeron, E., Marquis, M.E., Chretien, I. and Faucheux,
N. (2007) Differentiation of preosteoblasts using a de-
livery system with BMPs and bioactive glass micros-
pheres. Journal of Materials Science: Materials in Me-
dicine, 18, 255-263.
http://dx.doi.org/10.1007/s10856-006-0687-4
[49] Wozney, J.M., Rosen, V., Celeste, A.J., Mitsock, L.M.,
Whitters, M.J., Kriz, R.W., Hewick, R.M. and Wang, E.A.
(1988) Novel regulators of bone formation: Molecular
clones and activities. Science, 242, 1528-1534.
http://dx.doi.org/10.1126/science.3201241
[50] Glaser, D.L., Economides, A.N., Wang, L., Liu, X.,
Kimble, R.D., Fandl, J.P., Wilson, J.M., Stahl, N., Kaplan,
F.S. and Shore, E.M. (2003) In vivo somatic cell gene
transfer of an engineered Noggin mutein prevents BMP4-
induced heterotopic ossification. The Journal of Bone &
Joint Surgery, 85- A, 2332-2342.
[51] Clever, J.L., Sakai, Y., Wang, R.A. and Schneider, D.B.
(2010) Inefficient skeletal muscle repair in inhibitor of
differentiation knockout mice suggests a crucial role for
BMP signaling during adult muscle regeneration. Ameri-
can Journal of Physiology, 298, C1087-C1099.
http://dx.doi.org/10.1152/ajpcell.00388.2009
[52] Ono, Y., Calhabeu, F., Morgan, J.E., Katagiri, T., Amthor,
H. and Zammit, P.S. (2010) BMP signalling permits
population expansion by preventing premature myogenic
differentiation in muscle satellite cells. Cell Death and
Differentiation, 18, 222-234.
http://dx.doi.org/10.1038/cdd.2010.95
[53] Katagiri, T., Yamaguchi, A., Komaki, M., Abe, E., Ta-
kahashi, N., Ikeda, T., Rosen, V., Wozney, J.M., Fujisa-
wa-Sehara, A. and Suda, T. (1994) Bone morphogenetic
protein-2 converts the differentiation pathway of C2C12
myoblasts into the osteoblast lineage. The Journal of Cell
Biology, 127, 1755-1766.
http://dx.doi.org/10.1083/jcb.127.6.1755
[54] Oishi, T., Uezumi, A., Kanaji, A., Yamamoto, N., Yama-
guchi, A., Yamada, H. and Tsuchida, K. (2013) Osteo-
genic differentiation capacity of human skeletal muscle-
derived progenitor cells. PLoS One, 8, e56641.
http://dx.doi.org/10.1371/journal.pone.0056641
[55] Jackson, W.M., Aragon, A.B., Bulken-Hoover, J.D.,
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
44
Nesti, L.J. and Tuan, R.S. (2009) Putative heterotopic os-
sification progenitor cells derived from traumatized mu-
scle. Journal of Orthopaedic Research, 27, 1645-1651.
http://dx.doi.org/10.1002/jor.20924
[56] Leblanc, E., Trensz, F., Haroun, S., Drouin, G., Bergeron,
E., Penton, C.M., Montanaro, F., Roux, S., Faucheux, N.
and Grenier, G. (2011) BMP9-Induced muscle hetero-
topic ossification requires changes to the skeletal muscle
microenvironment. Journal of Bone and Mineral Re-
search, 26, 1166-1177.
http://dx.doi.org/10.1002/jbmr.311
[57] Lauzon, M.A., Bergeron, E., Marcos, B. and Faucheux, N.
(2012) Bone repair: New developments in growth factor
delivery systems and their mathematical modeling. Jour-
nal of Controlled Release, 162, 502 -520.
http://dx.doi.org/10.1016/j.jconrel.2012.07.041
[58] Bergeron, E., Leblanc, E., Drevelle, O., Giguere, R.,
Beauvais, S., Grenier, G. and Faucheux, N. (2012) The
evaluation of ectopic bone formation induced by delivery
systems for bone morphogenetic protein-9 or its derived
peptide. Tissue Engineering Part A, 18, 342-352.
http://dx.doi.org/10.1089/ten.tea.2011.0008
[59] Bessa, P.C., Balmayor, E.R., Azevedo, H.S., Nurnberger,
S., Casal, M., van Griensven, M., Reis, R.L. and Redl, H.
(2010) Silk fibroin microparticles as carriers for delivery
of human recombinant BMPs. Physical characterization
and drug release. Journal of Tissue Engineering and Re-
generative Medicine, 4, 349-355.
[60] Drevelle, O., Daviau, A., Lauzon, M.A. and Faucheux, N.
(2013) Effect of BMP-2 and/or BMP-9 on preosteoblasts
attached to polycaprolactone functionalized by adhesive
peptides derived from bone sialoprotein. Biomaterials, 34,
1051-1062.
http://dx.doi.org/10.1016/j.biomaterials.2012.10.066
[61] Marquis, M.E., Lord, E., Bergeron, E., Bourgoin, L. and
Faucheux, N. (2008) Short-term effects of adhesion pep-
tides on the responses of preosteoblasts to pBMP-9. Bi-
omaterials, 29, 1005-1016.
http://dx.doi.org/10.1016/j.biomaterials.2007.10.047
[62] Miller, M.D. (2008) Review of orthopaedics. Saunders/
Elsevier, Philadelphia.
[63] Goldring, M.B., Tsuchimochi, K. and Ijiri, K. (2005) The
control of chondrogenesis. Journal of Cellular Bioche-
mistry, 97, 33-44.
http://dx.doi.org/10.1002/jcb.20652
[64] Hidaka, C. and Goldring, M.B. (2008) Regulatory me-
chanisms of chondrogenesis and implications for under-
standing articular cartilage homeostasis. Current Rheu-
matology Reviews, 4, 136-147.
http://dx.doi.org/10.2174/157339708785133541
[65] Pizette, S. and Niswander, L. (2000) BMPs are required
at two steps of limb chondrogenesis: Formation of pre-
chondrogenic condensations and their differentiation into
chondrocytes. Developmental Biology, 219, 237-249.
http://dx.doi.org/10.1006/dbio.2000.9610
[66] Yoon, B.S., Ovchinnikov, D.A., Yoshii, I., Mishina, Y.,
Behringer, R.R. and Lyons, K.M. (2005) Bmpr1a and
Bmpr1b have overlapping functions and are essential for
chondrogenesis in vivo. Proceedings of the National Aca-
demy of Sciences of the United States of America, 102,
5062-5067.
[67] Kramer, J., Hegert, C., Guan, K., Wobus, A.M., Muller,
P.K. and Rohwedel, J. (2000) Embryonic stem cell-de-
rived chondrogenic differentiation in vitro: Activation by
BMP-2 and BMP-4. Mechanisms of Development, 92,
193-205.
http://dx.doi.org/10.1016/S0925-4773(99)00339-1
[68] Chubinskaya, S. and Kuettner, K.E. (2003) Regulation of
osteogenic proteins by chondrocytes. The International
Journal of Biochemistry & Cell Biology, 35, 1323-1340.
http://dx.doi.org/10.1016/S1357-2725(03)00035-9
[69] Yeh, L.C., Mallein-Gerin, F. and Lee, J.C. (2002)
Differential effects of osteogenic protein-1 (BMP-7) on
gene expression of BMP and GDF family members du-
ring differentiation of the mouse MC615 chondrocyte
cells. Journal of Cellular Physiology, 191, 298-309.
http://dx.doi.org/10.1002/jcp.10094
[70] Chubinskaya, S., Merrihew, C., Cs-Szabo, G., Mollen-
hauer, J., McCartney, J., Rueger, D.C. and Kuettner, K.E.
(2000) Human articular chondrocytes express osteogenic
protein-1. Journal of Histochemistry & Cytochemistry, 48,
239-250.
[71] Varady, P., Li, J.Z., Cunningham, M., Beres, E.J., Das, S.,
Engh, J., Alden, T.D., Pittman, D.D., Kerns, K.M., Kall-
mes, D.F. and Helm, G.A. (2001) Morphologic analysis
of BMP-9 gene therapy-induced osteogenesis. Human
Gene Therapy, 12, 697-710.
http://dx.doi.org/10.1089/104303401300057423
[72] Majumdar, M.K., Wang, E. and Morris, E.A. (2001)
BMP-2 and BMP-9 promotes chondrogenic differentia-
tion of human multipotential mesenchymal cells and over-
comes the inhibitory effect of IL-1. Journal of Cellular
Physiology, 189, 275-284.
http://dx.doi.org/10.1002/jcp.10025
[73] Blunk, T., Sieminski, A.L., Appel, B., Croft, C., Courter,
D.L., Chieh, J.J., Goepferich, A., Khurana, J.S. and
Gooch, K.J. (2003) Bone morphogenetic protein 9: A po-
tent modulator of cartilage development in vitro. Growth
Factors, 21, 71-77.
http://dx.doi.org/10.1080/0897719031000148822
[74] Fong, D., Bisson, M., Laberge, G., McManus, S., Grenier,
G., Faucheux, N. and Roux, S. (2013) Bone morpho-
genetic protein-9 activates smad and ERK pathways and
supports human osteoclast function and survival in vitro.
Cellular Signalling, 25, 717-728.
http://dx.doi.org/10.1016/j.cellsig.2012.12.003
[75] Castonguay, R., Werner, E.D., Matthews, R.G., Presman,
E., Mulivor, A.W., Solban, N., et al. (2011) Soluble en-
doglin specifically binds bone morphogenetic proteins 9
and 10 via its orphan domain, inhibits blood vessel for-
mation, and suppresses tumor growth. The Journal of Bi-
ological Chemistry, 286, 30034-30046.
http://dx.doi.org/10.1074/jbc.M111.260133
[76] Ye, L., Kynaston, H. and Jiang, W.G. (2008) Bone mor-
phogenetic protein-9 induces apoptosis in prostate cancer
cells, the role of prostate apoptosis response-4. Molecular
Cancer Research, 6, 1594-1606.
http://dx.doi.org/10.1158/1541-7786.MCR-08-0171
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46 45
[77] Li, B., Yang, Y., Jia ng, S., Ni, B., Chen, K. and Jia ng, L.
(2012) Adenovirus-mediated overexpression of BMP-9
inhibits human osteosarcoma cell growth and migration
through downregulation of the PI3K/AKT pathway.
International Journal of Oncology, 41, 1809-1819.
[78] Cunha, S.I., Pardali, E., Thorikay, M., Anderberg, C.,
Hawinkels, L., Goumans, M.J., Seehra, J., Heldin, C.H.,
ten Dijke, P. and Pietras, K. (2010) Genetic and phar-
macological targeting of activin receptor-like kinase 1
impairs tumor growth and angiogenesis. The Journal of
Experimental Medicine, 207, 85-100.
[79] Suzuki, Y., Ohga, N., Morishita, Y., Hida, K., Miyazono,
K. and Watabe, T. (2010) BMP-9 induces proliferation of
multiple types of endothelial cells in vitro and in vivo.
Journal of Cell Science, 123, 1684-1692.
[80] Luo, X., Chen, J., Song, W.X., Tang, N., Luo, J., Deng,
Z.L., et al. (2008) Osteogenic BMPs promote tumor
growth of human osteosarcomas that harbor differen-
tiation defects. Laboratory Investigation, 88, 1264-1277.
http://dx.doi.org/10.1038/labinvest.2008.98
[81] Park, H., Drevelle, O., Da viau, A., Senta, H. , Bergeron, E.
and Faucheux, N. (2012) Preventing MEK1 activation
influences the responses of human osteosarcoma cells to
bone morphogenetic proteins 2 and 9. Anticancer Drugs,
24, 278-290.
http://dx.doi.org/10.1097/CAD.0b013e32835cbde7
[82] Lv, Z., Yang, D., Li, J., Hu, M., Luo, M., Zhan, X., Song,
P., Liu, C., Bai, H., Li, B., Yang, Y., Chen, Y., Shi, Q.
and Weng, Y. (2013) Bone morphogenetic protein 9
overexpression reduces osteosarcoma cell migration and
invasion. Molecules and Cells, 36, 119-126.
http://dx.doi.org/10.1007/s10059-013-0043-8
[83] Spanjol, J., Dj ordje vic , G. , Marki c, D., Klar ic, M. , Fuc kar,
D. and Bobinac, D. (2010) Role of bone morphogenetic
proteins in human prostate cancer pathogenesis and de-
velopment of bone metastases: Immunohistochemical stu-
dy. Collegium Antropologicum, 34, 119-125.
[84] David, L., Feige, J.J. and Bailly, S. (2009) Emerging role
of bone morphogenetic proteins in angiogenesis. Cyto-
kine & Growth Factor Reviews, 20, 203-212.
http://dx.doi.org/10.1016/j.cytogfr.2009.05.001
[85] Mitchell, D., Pobre, E.G., Mulivor, A.W., Grinberg, A.V.,
Castonguay, R., Monnell, T.E., Solban, N., Ucran, J.A.,
Pearsall, R.S., Underwood, K.W., Seehra, J. and Kumar,
R. (2010) ALK1-Fc inhibits multiple mediators of angi-
ogenesis and suppresses tumor growth. Molecular Cancer
Therapy, 9, 379-388.
http://dx.doi.org/10.1158/1535-7163.MCT-09-0650
[86] Ricard, N., Bidart, M., Mallet, C., Lesca, G., Giraud, S.,
Prudent, R., Feige, J.J. and Bailly, S. (2010) Functional
analysis of the BMP9 response of ALK1 mutants from
HHT2 patients: A diagnostic tool for novel ACVRL1 mu-
tations. Blood, 116, 1604-1612.
[87] Herrera, B. and Inman, G.J. (2009) A rapid and sensitive
bioassay for the simultaneous measurement of multiple
bone morphogenetic proteins. Identification and quanti-
fication of BMP4, BMP6 and BMP9 in bovine and hu-
man serum. BMC Cell Biology, 10, 20.
http://dx.doi.org/10.1186/1471-2121-10-20
[88] Haitjema, T., Westermann, C.J., Overtoom, T.T., Timmer,
R., Disch, F., Mauser, H. and Lammers, J.W. (1996) He-
reditary hemorrhagic telangiectasia (Osler-Weber-Rendu
disease): New insights in pathogenesis, complications,
and treatment. Archives of Internal Medicine, 156, 714-
719.
http://dx.doi.org/10.1001/archinte.1996.00440070028004
[89] Mahmoud, M., Upton, P.D. and Arthur, H.M. (2011) An-
giogenesis regulation by TGFbeta signalling: Clues from
an inherited vascular disease. Biochemical Society Tran-
sactions, 39, 1659-1666.
http://dx.doi.org/10.1042/BST20110664
[90] Alt, A., Miguel-Romero, L., Donderis, J., Aristorena, M.,
Blanco, F.J., Round, A., Rubio, V., Bernabeu, C. and Ma-
rina, A. (2012) Structural and functional insights into en-
doglin ligand recognition and binding. PLoS One, 7,
e29948. http://dx.doi.org/10.1371/journal.pone.0029948
[91] Bailly, S., Dupuis-Girod, S. and Plauchu, H. (2010) Ren-
du-osler disease: Clinical and molecular update. Medi-
cine/Science (Paris), 26, 855-860.
http://dx.doi.org/10.1051/medsci/20102610855
[92] Choi, E.J., Kim, Y.H., Choe, S.W., Tak, Y.G., Garrido-
Martin, E.M., Chang, M., Lee, Y.J. and Oh, S.P. (2013)
Enhanced responses to angiogenic cues underlie the pa-
thogenesis of hereditary hemorrhagic telangiectasia 2.
PLoS One, 8, e63138.
http://dx.doi.org/10.1371/journal.pone.0063138
[93] Somekawa, S., Imagawa, K., Hayashi, H., Sakabe, M.,
Ioka, T., Sato, G.E ., Inada , K., Iwamoto, T., Mori , T. , Ue-
mura, S., Nakagawa, O. and Saito, Y. (2012) Tmem100,
an ALK1 receptor signaling-dependent gene essential for
arterial endothelium differentiation and vascular morpho-
genesis. Proceedings of the National Academy of Scien-
ces of the United States of America, 109, 12064-12069.
[94] Chida, A., Shintani, M., Yagi, H., Fujiwara, M., Kojima,
Y., Sato, H., et al. (2013) Outcomes of childhood pul-
monary arterial hypertension in BMPR2 and ALK1 mu-
tation carriers. American Journal of Cardiology, 110,
586-593.
[95] Dunmore, B.J., Drake, K.M., Upton, P. D., Toshner, M.R.,
Aldred, M.A. and Morrell, N.W. (2013) The lysosomal
inhibitor, chloroquine, increases cell surface BMPR-II
levels and restores BMP9 signalling in endothelial cells
harbouring BMPR-II mutations. Human Molecular Ge-
netics, 22, 3667-3679
[96] Bachiller, D., Klingensmith, J., Kemp, C., Belo, J.A.,
Anderson, R.M., May, S.R., McMahon, J.A., McMahon,
A.P., Harland, R.M., Rossant, J. and De Robertis, E.M.
(2000) The organizer factors chordin and noggin are re-
quired for mouse forebrain development. Nature, 403,
658-661.
[97] Weinstein, D.C. and Hemmati-Brivanlou, A. (1999) Neu-
ral induction. Annual Review of Cell and Developmental
Biology, 15, 411-433.
http://dx.doi.org/10.1146/annurev.cellbio.15.1.411
[98] Lopez-Coviella, I., Mellott, T.M., Kovacheva, V.P.,
Berse, B., Slack, B.E., Zemelko, V., Schnitzler, A. and
Blusztajn, J.K. (2006) Developmental pattern of ex-
pression of BMP receptors and smads and activation of
Copyright © 2013 SciRes. ABB
E. Leblanc et al. / Advances in Bioscience and Biotechnology 4 (2013) 31-46
Copyright © 2013 SciRes.
46
ABB
Smad1 and Smad5 by BMP9 in mouse basal forebrain.
Brain Research, 1088, 49-56.
http://dx.doi.org/10.1016/j.brainres.2006.02.073
[99] Lopez-Coviella, I., Berse, B., Krauss, R., Thies, R.S. and
Blusztajn, J.K. (2000) Induction and maintenance of the
neuronal cholinergic phenotype in the central nervous
system by BMP-9. Science, 289, 313-316.
http://dx.doi.org/10.1016/j.brainres.2006.02.073
[100] Wu, C.K., Thal, L., Pizzo, D., Hansen, L., Masliah, E.
and Geula, C. (2005) Apoptotic signals within the basal
forebrain cholinergic neurons in Alzheimer’s disease.
Experimental Neurology, 195, 484-496.
http://dx.doi.org/10.1016/j.expneurol.2005.06.020
[101] Lopez-Coviella, I., Follettie, M.T., Mellott, T.J., Kova-
cheva, V.P., Slack, B.E., Diesl, V., Berse, B., Thies, R.S.
and Blusztajn, J.K. (2005) Bone morphogenetic protein 9
induces the transcriptome of basal forebrain cholinergic
neurons. Proceedings of the National Academy of Scien-
ces of the United States of America, 102, 6984-6989.
[102] Lopez-Coviella, I., Mellott, T.J., Schnitzler, A.C. and
Blusztajn, J.K. (2011) BMP9 protects septal neurons
from axotomy-evoked loss of cholinergic phenotype.
PLoS One, 6, e21166.
http://dx.doi.org/10.1371/journal.pone.0021166
[103] Bissonnette, C.J., Lyass, L., Bhattacharyya, B.J., Bel-
madani, A., Miller, R.J. and Kessler, J.A. (2011) The
controlled generation of functional basal forebrain cho-
linergic neurons from human embryonic stem cells. Stem
Cells, 29, 802-811.
http://dx.doi.org/10.1002/stem.626
[104] Citta-Pietrolungo, T.J., Alexander, M.A. and Steg, N.L.
(1992) Early detection of heterotopic ossification in
young patients with traumatic brain injury. Archives of
Physical Medicine and Rehabilitation, 73, 258-262.
[105] Gautschi, O.P., Cadosch, D., Frey, S.P., Skirving, A.P.,
Filgueira, L. and Zellweger, R. (2009) Serum-mediated
osteogenic effect in traumatic brain-injured patients. ANZ
Journal of Surgery, 79, 449-455.
http://dx.doi.org/10.1111/j.1445-2197.2008.04803.x
[106] Cadosch, D., Gautschi, O.P., Thyer, M., Song, S., Ski-
rving, A.P., Filgueira, L. and Zellweger, R. (2009) Hu-
moral factors enhance fracture-healing and callus forma-
tion in patients with traumatic brain injury. The Journal
of Bone & Joint Surgery, 91, 282-288.
http://dx.doi.org/10.2106/JBJS.G.01613
[107] Cipriano, C.A., Pill, S.G. and Keenan, M.A. (2009) He-
terotopic ossification following traumatic brain injury and
spinal cord injury. Journal of the American Academy of
Orthopaedic Surgeons, 17, 689-697.
[108] Gautschi, O.P., Toffoli, A.M., Joesbury, K.A., Skirving,
A.P., Filgueira, L. and Zellweger, R. (2007) Osteoinduc-
tive effect of cerebrospinal fluid from brain-injured pa-
tients. Journal of Neurotrauma, 24, 154-162.
http://dx.doi.org/10.1089/neu.2006.0166
[109] Toffoli, A.M., Gautschi, O.P., Frey, S.P., Filgueira, L.
and Zellweger, R. (2008) From brain to bone: Evidence
for the release of osteogenic humoral factors after trau-
matic brain injury. Brain Injury, 22, 511-518.
http://dx.doi.org/10.1080/02699050802158235
[110] Sosa, I., Cvijanovic, O., Celic, T., Cuculic, D., Crncevic-
Orlic, Z., Vukelic, L., Zoricic-Cvek, S., Dudaric, L., Bos-
nar, A. and Bobinac, D. (2011) Hepatoregenerative role
of bone morphogenetic protein-9. Medical Science Moni-
tor, 17, 33-35. http://dx.doi.org/10.12659/MSM.882108
[111] Gao, Y.L., Cai, X.F., Liu, J., Shan, X.L., Chen, Q.M.,
Zhou, F. and Tang, N. (2011) Hepatic lineage differen-
tiation of hepatic progenitor cells by bone morphogenetic
protein or leukemia inhibitory factor. Zhonghua Gan
Zang Bing Za Zhi, 19, 692-695.
[112] George, S., Rochford, J.J., Wolfrum, C., Gray, S.L.,
Schinner, S., Wilson, J.C., et al. (2004) A family with se-
vere insulin resistance and diabetes due to a mutation in
AKT2. Science, 304, 1325-1328.
[113] Cho, H., Mu, J., Kim, J.K., Thorvaldsen, J.L., Chu, Q.,
Crenshaw, E.B., 3rd, Kaestner, K.H., Bartolomei, M.S.,
Shulman, G.I. and Birnbaum, M.J. (2001) Insulin resis-
tance and a diabetes mellitus-like syndrome in mice lack-
ing the protein kinase Akt2 (PKB beta). Science, 292,
1728-1731.
[114] Anhe, F.F., Lellis-Santos, C., Leite, A.R., Hirabara, S.M.,
Boschero, A.C., Curi, R., Anhe, G.F. and Bordin, S.
(2010) Smad5 regulates Akt2 expression and insulin-in-
duced glucose uptake in L6 myotubes. Molecular and
Cellular Endocrinology, 319, 30-38.
http://dx.doi.org/10.1126/science.292.5522.1728
[115] Caperuto, L.C., Anhe, G.F., Cambiaghi, T.D., Akamine,
E.H., do Carmo Buonfiglio, D., Cipolla-Neto, J., Curi, R.
and Bordin, S. (2008) Modulation of bone morphogenetic
protein-9 expression and processing by insulin, glucose,
and glucocorticoids: Possible candidate for hepatic insu-
lin-sensitizing substance. Endocrinology, 149, 6326-
6335.