ArticlePDF AvailableLiterature Review

BMP type II receptor as a therapeutic target in pulmonary arterial hypertension

Authors:

Abstract and Figures

Pulmonary arterial hypertension (PAH) is a chronic disease characterized by a progressive elevation in mean pulmonary arterial pressure. This occurs due to abnormal remodeling of small peripheral lung vasculature resulting in progressive occlusion of the artery lumen that eventually causes right heart failure and death. The most common cause of PAH is inactivating mutations in the gene encoding a bone morphogenetic protein type II receptor (BMPRII). Current therapeutic options for PAH are limited and focused mainly on reversal of pulmonary vasoconstriction and proliferation of vascular cells. Although these treatments can relieve disease symptoms, PAH remains a progressive lethal disease. Emerging data suggest that restoration of BMPRII signaling in PAH is a promising alternative that could prevent and reverse pulmonary vascular remodeling. Here we will focus on recent advances in rescuing BMPRII expression, function or signaling to prevent and reverse pulmonary vascular remodeling in PAH and its feasibility for clinical translation. Furthermore, we summarize the role of described miRNAs that directly target the BMPR2 gene in blood vessels. We discuss the therapeutic potential and the limitations of promising new approaches to restore BMPRII signaling in PAH patients. Different mutations in BMPR2 and environmental/genetic factors make PAH a heterogeneous disease and it is thus likely that the best approach will be patient-tailored therapies.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Cell. Mol. Life Sci. (2017) 74:2979–2995
DOI 10.1007/s00018-017-2510-4
REVIEW
BMP type II receptor asatherapeutic target inpulmonary
arterial hypertension
MarOrriols1· MariaCatalinaGomez‑Puerto1· PetertenDijke1
Received: 22 December 2016 / Revised: 9 March 2017 / Accepted: 17 March 2017 / Published online: 26 April 2017
© The Author(s) 2017. This article is an open access publication
Keywords Endothelial cell· Vascular smooth muscle
cell· Signal transduction· Inflammation· Vascular
remodeling and autophagy
Abbreviations
ACE Angiotensin-converting enzyme
ACVRII Activin type II receptor type
ACVRL1 Gene encoding activin receptor-like kinase
ALK Activin receptor-like kinase
BMP Bone morphogenetic protein
BMPRI Bone morphogenetic protein type I receptor
BMPRII Bone morphogenetic protein type II receptor
BMPR2 Gene encoding bone morphogenetic protein
receptor II
Cav1 Caveolin-1
cGMP Cyclic guanosine monophosphate
DN Dominant negative
EC Endothelial cell
EndMT Endothelial-to-mesenchymal transition
FKBP12 FK-binding protein-12
GDF Growth and differentiation factor
GDF-2 Gene encoding BMP9
HMGA1 High-mobility group protein
iTOP Induced transduction by osmocytosis and
propanebetaine
MCT Monocrotaline
mPAP Mean pulmonary arterial pressure
miRNA MicroRNA
NMD Non-sense mediated decay
PAEC Pulmonary arterial endothelial cells
PAH Pulmonary arterial hypertension
PASMC Pulmonary arterial smooth muscle cells
PGI2 Prostacyclin
PKG Protein kinase G
PTC Premature termination codon
Abstract Pulmonary arterial hypertension (PAH) is a
chronic disease characterized by a progressive elevation
in mean pulmonary arterial pressure. This occurs due to
abnormal remodeling of small peripheral lung vasculature
resulting in progressive occlusion of the artery lumen that
eventually causes right heart failure and death. The most
common cause of PAH is inactivating mutations in the
gene encoding a bone morphogenetic protein type II recep-
tor (BMPRII). Current therapeutic options for PAH are lim-
ited and focused mainly on reversal of pulmonary vasocon-
striction and proliferation of vascular cells. Although these
treatments can relieve disease symptoms, PAH remains a
progressive lethal disease. Emerging data suggest that res-
toration of BMPRII signaling in PAH is a promising alter-
native that could prevent and reverse pulmonary vascular
remodeling. Here we will focus on recent advances in res-
cuing BMPRII expression, function or signaling to prevent
and reverse pulmonary vascular remodeling in PAH and its
feasibility for clinical translation. Furthermore, we summa-
rize the role of described miRNAs that directly target the
BMPR2 gene in blood vessels. We discuss the therapeutic
potential and the limitations of promising new approaches
to restore BMPRII signaling in PAH patients. Different
mutations in BMPR2 and environmental/genetic factors
make PAH a heterogeneous disease and it is thus likely that
the best approach will be patient-tailored therapies.
Cellular and Molecular LifeSciences
Mar Orriols and Maria Catalina Gomez-Puerto the authors have
contributed equally to this work.
* Peter ten Dijke
p.ten_dijke@lumc.nl
1 Department ofMolecular Cell Biology andCancer Genomics
Center Netherlands, Leiden University Medical Center,
Leiden, TheNetherlands
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2980 M.Orriols et al.
1 3
RV Right ventricle
SMC Smooth muscle cell
TGF-β Transforming growth factor-beta
TNFα Tumor necrosis factor-alfa
Introduction
Pulmonary arterial hypertension (PAH) is a chronic dis-
ease characterized by a progressive elevation in mean pul-
monary arterial pressure (mPAP >25 mmHg) leading to
right heart failure and death [1]. PAH is characterized by
abnormal remodeling of small peripheral lung vascula-
ture resulting in progressive occlusion of the artery lumen.
In addition, at late stages, so-called plexiform lesions are
found, which are complex vascular formations originating
from abnormal endothelial cell (EC) proliferation and vas-
cular smooth muscle cell (SMC) hypertrophy [2]. The basic
pathogenic mechanisms underlying this disease include
vasoconstriction, intimal proliferation, and medial hyper-
trophy. These processes are accompanied by illicit recruit-
ment of inflammatory cells which release factors enhanc-
ing cell proliferation and elastin fibers degradation [3, 4]
(Fig.1).
More than 70% of patients with familial PAH and 20%
of idiopathic PAH show heterozygous mutations in the
bone morphogenetic protein type II receptor (BMPRII)
[58]. BMPRII is a transmembrane serine/threonine kinase
receptor of the bone morphogenetic protein (BMP) pathway
which is essential for embryogenesis, development, and
adult tissue homeostasis. Upon BMP-induced heteromeric
complex formation of BMPRII with BMP type I receptor
(BMPRI), BMPRII activates BMPRI by phosphorylation.
Thereafter, the activated BMPRI propagates the signal into
the cell through phosphorylation of the SMAD1/5/8 tran-
scription factors.
In PAH, over 300 mutations have been found in the
BMPR2 gene. These mutations target sequences that
encode the ligand binding and kinase domain and the
long cytoplasmic tail; the mutations compromise BMPRII
function [9]. Although the BMPRII pathway is essential
for vascular homeostasis and there is a strong correlation
between BMPR2 mutations and PAH, the incomplete pen-
etrance of BMPRII mutations (20–30%) suggests that other
genetic and environmental factors contribute to the disease.
Among them, BMPR2 alternative splicing plays a role in
PAH penetrance. One BMPR2 splice variant lacks exon 12,
which is the largest exon of the gene and encodes the cyto-
plasmic tail. It has been shown that carriers of this variant
are more prone to develop PAH through a dominant-neg-
ative effect (DN) effect on wild-type BMPRII [10]. Fur-
thermore, there are mutations in other genes in the BMP
pathway, which further strengthens the notion of a causal
role for this pathway in PAH [11]. Moreover, the co-exist-
ence of modifier genes, infections, toxic exposure, inflam-
mation, or alterations in estrogen metabolism has been
described [1114] and some of them were found to down-
regulate BMPRII expression. For example, pro-inflam-
matory cytokines such as tumor necrosis factor α (TNFα)
and Interleukin 6 induce the expression of miRNAs that
inhibit BMPRII expression [15]. Furthermore, BMPRII is
essential for maintaining the barrier function of the pulmo-
nary artery endothelial cell lining and BMPRII deficiency
increases endothelial inflammatory responses thereby con-
tributing to adverse vascular remodeling [1618].
Current therapeutic options for PAH are limited and
focused mainly on reversal of pulmonary vasoconstric-
tion and proliferation of vascular cells through targeting of
Fig. 1 Physiopathological mechanisms of pulmonary arterial hyper-
tension development. Presence of genetic risk factors such as BMPR2
mutations together with exposure to deleterious environmental or
biological stimuli in the lung promotes PAH. PAH development is
characterized by a disturbance on the signaling pathways that con-
trol pulmonary vascular homeostasis. It results in pulmonary vascu-
lar thickening and occlusion compromising lung and heart function.
EndMT endothelial-to-mesenchymal transition
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2981BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
prostacyclin (PGI2), endothelin, or nitric oxide pathways
[19]. Although these treatments can relieve disease symp-
toms and slow down its progression, PAH remains a pro-
gressive lethal disease. Abundant research over the past
decade has improved our understanding of the molecular
mechanisms underlying PAH progression revealing novel
potential therapeutic interventions [2022]. Among them
there are several anti-proliferative strategies including cell
cycle inhibitors (e.g., mTOR inhibitor rapamycin) and
anti-apoptotic drugs (e.g., surviving inhibitors) [23]. Fur-
thermore, based on the fact that Rho and ROCK mediate
smooth muscle cell proliferation in a serotonin-BMPR-
dependent pathway, Rho-kinase inhibitors have been also
considered [23, 24]. Although several drugs with possible
benefit in PAH have been identified, only very few have
been approved for use in the clinic due to toxicity or lack of
clinical efficacy. This review will focus on recent advances
on the rescue of BMPRII expression, function, or signaling
to prevent and reverse pulmonary vascular remodeling in
PAH. We will discuss data on the invitro efficacy of the
different approaches together with the physiological out-
comes in pre-clinical models and their feasibility for clini-
cal translation.
BMP signaling invascular biology andPAH
BMPs belong to the multifunctional transforming growth
factor-β (TGF-β) family of secreted dimeric cytokines.
The effects of BMPs are highly dependent on cellular
context [25]. In general, BMPs control cellular prolifera-
tion, differentiation, and apoptosis, and play an important
role in embryonic development and maintaining tissue
homeostasis [26]. Therefore, perturbation of BMP signal-
ing may lead to skeletal diseases, vascular diseases, and
cancer [27]. BMPs can be subdivided into four subgroups
based on their sequence similarity and cell surface recep-
tor affinities: BMP2/4, BMP5/6/7/8, BMP9/10, and growth
and differentiation factor (GDF)-5/6/7 [28, 29]. BMPs sig-
nal via hetero-tetrameric combinations of type I receptors
(activin receptor-like kinase (ALK)1, ALK2, ALK3, or
ALK6) and BMP type II receptors (BMPRII) and activin
type II receptor (ACVRII)A or ACVRIIB complexes [30,
31]. Both, type I and type II receptors have a similar struc-
ture encompassing a short extracellular domain, a single
transmembrane domain and an intracellular domain with
intrinsic serine-threonine kinase activity. In the vascular
endothelium, BMP signaling is mainly activated by BMP2,
4, 6, 9, and 10 [32]. BMP2 and BMP4 bind preferentially to
the BMPRII in complex with ALK3 or ALK6. BMP6 binds
to the ACVRIIA-ALK2 complex, while BMP9 and BMP10
bind to BMPRII or ACVRII in combination with ALK1 or
ALK2. Whereas BMPRII is a specific receptor for BMPs,
ACVRIIA and ACVRIIB can also interact functionally
with other ligands, such as activins, myostatin, and nodal.
Interestingly, ALK2 and 6 are widely expressed in various
cell types, while ALK1 has a more selective expression
pattern and is mainly restricted to ECs. After BMP binding
and receptor complex formation, the type II receptor kinase
phosphorylates the type I receptor on serine and threonine
residues in the glycine-serine rich (GS)-domain causing its
activation and subsequent phosphorylation of the recep-
tor-associated R-SMAD1, 5, and 8 effector proteins. The
R-SMADs that are activated by TGF-β type I and activin
type I receptor (i.e., ALK5 and ALK4, respectively) are
SMAD2 and SMAD3 and these are distinct from BMP
R-SMADs. Activated R-SMAD 1, 5, or 8 forms a hetero-
oligomeric complex with common mediator co-SMAD4.
This complex translocates to the nucleus and regulates the
expression of target genes by binding to specific enhanc-
ers/promoters upstream of these target genes [30, 33, 34]
(Fig. 2). Besides canonical BMP receptor/SMAD signal-
ing, activated BMP receptors can initiate non-SMAD sign-
aling pathways such as ERK, JNK, p38 MAP kinases, and
the phosphatidyl inositol 3 kinase (PI3K)/AKT pathways
[3537]. These non-SMAD pathways are also important for
diversifying and modulating the canonical SMAD signal-
ing pathways that are activated by the BMP receptors [38,
39]. In addition, BMP activity is also regulated by several
extracellular modulators, including BMP binding proteins
NOGGIN, CHORDIN, and FIBULINs. Co-receptors such
as ENDOGLIN, BETAGLYCAN, and DRAGON fam-
ily members may also modulate the interactions between
BMPs and BMP receptors [40, 41]. Moreover, intracellu-
lar kinases/phosphatases and other binding proteins have
been identified as regulators of the trafficking, subcellular
localization, stability, and function of BMP receptors and
SMADs [26].
Genetic depletion of different components of the BMP
signaling cascade leads to embryonic death due to cardio-
vascular malformations and abnormal vascular remodeling.
BMP signaling plays an important role in vasculogenesis
(de novo formation of blood vessels from undifferentiated
mesodermal cells) and angiogenesis (formation of new
blood vessels from the existing vasculature). In this light,
it is not surprising to discover that, besides PAH, dysfunc-
tion of BMP signaling has been found to be associated with
other vascular diseases including hereditary hemorrhagic
telangiectasia, cerebral cavernous malformation, athero-
sclerosis, and vascular calcification among others [42].
Furthermore, BMPRII downregulation has been found to
be involved in pancreatic and lung fibrosis [43, 44].
Blood vessels are composed of three layers: the tunica
adventitia consisting of fibroblasts and associated collagen
fibers; the tunica media composed of SMCs; and the tunica
intima consisting of ECs coating the interior surface [45,
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2982 M.Orriols et al.
1 3
46]. ECs, SMCs, and fibroblasts have been found to play
a role in the pathogenesis of PAH. Abnormal EC prolif-
eration resulting in the formation of plexiform lesions has
been frequently described in many PAH cases [47]. In addi-
tion, pulmonary arterial SMCs show an increased prolifera-
tion and decreased apoptosis leading to vessel wall thicken-
ing and vascular remodeling (Fig.1). The close interaction
between ECs and SMCs has been found to be involved in
vessel formation and maintenance. For instance, endothe-
lial derived factors, like endothelin and angiotensin II,
affect SMCs which increases vascular tone. Similarly, nitric
oxide and PGI2 secreted by ECs modulate the vasodilator
response of SMCs [48]. In particular, PGI2 has been found
to be reduced in PAH patients [49].
BMP signaling is known to control cell migration, pro-
liferation, and apoptosis in ECs and SMCs [45]. BMP9
and BMP10 are present in the circulation and play an
important role in the vasculature. Their associated recep-
tors BMPRII and ALK1 and co-receptor ENDOGLIN are
predominantly expressed on ECs [50, 51] and together
can modulate the ability of ECs to migrate and prolifer-
ate [27]. Furthermore, BMPRII is also expressed in vas-
cular SMCs where it has been shown to be necessary
for the control of proliferation and differentiation [52].
Besides mutations in the BMPR2 gene, mutations in
the genes of other BMP signaling components (such as
GDF-2, ACVRL1, ENDOGLIN, and SMAD8) have also
been linked to PAH development [11, 5359]. This asso-
ciation reinforces the importance of BMP signaling in
the control of vascular homeostasis, and suggests that
there is a causal link between perturbation of canonical
BMP/SMAD signaling and PAH. In support of this view,
recent, new DNA sequencing techniques helped to iden-
tify new gene mutations associated to PAH [Caveolin-1
(CAV1), KCNK3, and EIF2AK4] [60, 61] (Fig.2).
The abnormal vascular remodeling that characterizes
PAH involves an accumulation of α-smooth muscle,
actin-expressing mesenchymal-like cells indicating that
the endothelial-to-mesenchymal transition (EndMT)
may be involved in the pathogenesis of the disease [62].
In addition, BMPRII reduction in pulmonary artery
endothelial cells (PAECs) has been found to promote the
trans-differentiation of epithelial cells into motile mesen-
chymal cells via the transcription factors high-mobility
group protein (HMGA)1 and its target SLUG [63].
Fig. 2 BMP signaling in
endothelial cells. BMP9 and
BMP10 present in the circu-
lation initiate signaling by
binding and bringing together
BMPRII and ALK1. BMPRII
phosphorylates ALK1 which
then propagate the signal
through phosphorylation of
SMAD1/5/8. Subsequently,
SMAD4 forms a complex with
SMAD1/5/8, which translo-
cates to the nucleus regulating
the expression of target genes
such as ID1 and ID3. Known
gene mutations associated with
PAH are highlighted in red.
It includes mutations in BMP
signaling components (GDF2,
BMPR2, ALK1, SMAD8, and
ENDOGLIN) as well as recently
discovered non-directly related
BMP genes (CAV1, KCNK3,
and EIF2AK4). CAV caveolin,
EFI2AK4 eukaryotic translation
initiation factor 2α kinase 4,
ENG ENDOGLIN, ID inhibi-
tor of DNA binding, KCNK3
potassium channel subfamily K
member 3
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2983BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
Animal models ofPAH
PAH has a complex etiology and pathobiology with many
factors contributing to its development [64]. A variety of
pre-clinical rodent models have been used to study the
underlying pathophysiological mechanisms and to test
novel therapeutic strategies for PAH. A proper model
should be reproducible, inexpensive, and faithfully repro-
duce (in a defined period) the basic features of PAH such
as complex destructive neointimal lesions and right ventri-
cle (RV) dysfunction and failure. To date, there is no model
that recapitulates all of the pathophysiological mechanisms
and the clinical course of human PAH. For instance, in
the chronic hypoxic exposure or monocrotaline (MCT)-
induced rat models, pulmonary hypertension rarely devel-
ops with the same severity observed in humans perhaps due
to the absence of obstructive intimal lesions in the periph-
eral pulmonary arteries [65, 66]. Furthermore, the chronic
hypoxia model does not lead to RV failure, while MCT
injection causes myocarditis affecting both ventricles and
causing liver and kidney damage [67]. These limitations
may explain why it is difficult to translate the reversal of
PAH in animal models by several experimental compounds
into therapies for PAH patients.
In recent years, second-generation animal models have
been established based on the combination of multiple
triggers: MCT plus pneumonectomy, MCT plus chronic
hypoxia, and SU5416 plus chronic hypoxia. To circumvent
the problem of the embryonic lethality of BMPR2 knock-
out mice, switchable rodent models have been developed,
by means of BMPR2 conditional knock-out, whereby the
mutation can be activated after birth [6870]. Moreo-
ver, genetic rodent models have been developed includ-
ing overexpression of interleukin-6. These new models
closely mimic the features and the severity of human PAH
although not completely [71]).
Restoring BMPRII signaling asatherapeutic
strategy
While hereditary PAH have been linked to heterozygous
mutations in the BMPR2 gene, non-genetic forms of PAH
show a reduction in BMPRII levels and activity [9]. Con-
sistent with this, heterozygous BMPR2 deletion in PAECs
and pulmonary artery smooth muscle cells (PASMCs)
mimics the PAH phenotype [69, 72]. Furthermore, mice
expressing a dominant-negative BMPRII (lacking an intra-
cellular domain) in vascular SMC, develop vascular lesions
in the lungs [68, 72].
There is strong evidence suggesting that BMPRII signal-
ing has a protective role in the vascular wall by promoting
the survival of PAECs, inhibiting PASMCs proliferation
and triggering anti-inflammatory responses [17, 73, 74].
Based on this, modulation of BMPRII signaling is consid-
ered a promising therapeutic approach for PAH. Impor-
tantly, the rescue of BMPRII expression may not exclu-
sively benefit PAH patients but also patients suffering from
pancreatic and lung fibrosis where BMPRII deficiency has
been implicated [43, 44]. BMPRII restoration can be tar-
geted at different levels: genetic-based therapies, transcrip-
tional and translational regulation, protein activity, and
processing as well as SMAD downstream signaling modu-
lation [27, 75, 76] (Fig.3).
Genetic‑based therapies
Exogenous BMPR2 gene delivery
One strategy to treat PAH patients is to rescue BMPRII
expression through gene therapy targeting ECs. In pre-
clinical models, this was explored by Reynolds etal., who
administrated a vector inducing BMPRII expression via
tail-vein injection. The BMPR2 encoding virus targets the
pulmonary endothelium by binding to the highly expressed
pulmonary endothelial angiotensin-converting enzyme
(ACE) using a bi-specific conjugate antibody. This BMPR2
adenoviral vector restored BMPRII protein levels in human
microvascular PAECs and attenuated the PAH phenotype
in a chronic hypoxia model and MCT-treated rats [77,
78]. Furthermore, BMPRII overexpression in lung tissue
was shown to reverse the imbalance between BMPRII and
TGFβ signaling thus restoring normal levels of pSMAD
1/5/8 and the activation of PI3K and p38 MAP kinase [79].
In contrast, BMPRII administration via an aerosol route
targeting PASMCs did not improve the PAH phenotype
when tested in the MCT model [80]. The later result high-
lights the importance of BMPRII signaling in ECs but not
SMCs. However, further investigations will be required to
elucidate precisely how spatio-temporal control BMPRII
overexpression might provide therapeutic benefit in the
context of BMPR2 mutations. It should be noted that ade-
noviral vectors are only capable of transient gene expres-
sion since the delivered gene is not integrated into the host
chromosome. Stable integration can be achieved and lenti-
viral vectors are potentially an attractive vehicle to deliver
longer term transgene expression since they integrate into
the genome and they can infect non-proliferating cells,
when compared to retroviral vectors. An important poten-
tial limitation of this approach is that integrating vectors
may generate gene mutations upon insertion and newer
advances regarding self-inactivating vectors are needed
[81, 82]. Adeno-associated virus and helper dependent ade-
noviral vectors (the latest generation of recombinant adeno-
viral vectors) are a promising alternative since they deliver
longer durations of transgene expression when compared
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2984 M.Orriols et al.
1 3
to the first-generation vectors. Moreover, they show nei-
ther long-term adverse effects in liver nor an immunologi-
cal response [83, 84]. Taken together, exogenous BMPR2
delivery is a possible therapy for PAH, but further improve-
ments in vector technology are required to translate this
approach to the clinic for the treatment of pulmonary vas-
cular disease.
Transcriptional regulation
miRNA targeting BMPRII
In recent years, there has been an increasing interest in the
role of epigenetics in the development of PAH [85, 86].
Epigenetics refers to heritable changes in gene expres-
sion that do not involve alterations in the DNA sequence.
miRNAs are small non-coding RNAs that negatively, post-
transcriptionally regulate the expression of target genes by
interfering with both the stability of the target transcript as
well as its translation. miRNAs have emerged as essential
players in the development (and diseases) of the cardiovas-
cular system. They also play an important role in vascular
remodeling [87, 88]. miRNAs are expressed in the vascu-
lature and are essential for the regulation of vessel func-
tion. Many miRNAs control proliferation, differentiation,
and apoptosis of ECs and SMCs by targeting components
of the TGF-β/BMP signaling pathways. Several miRNAs,
such as miR-145, miR-21 and the miR17/92 cluster, have
been associated with the disrupted BMPRII pathway in
PAH and can explain the incomplete penetrance of BMPR2
mutations [8991]). Figure4 and Table1 provides an over-
view of currently described miRNAs that target BMPR2
Fig. 3 Rescuing the BMPRII
signaling pathway in pulmonary
arterial hypertension. Modula-
tion of BMPRII signaling is
considered a promising thera-
peutic approach for PAH. This
could be achieved by different
methods aiming to increase the
amounts of BMPRII present
in the cell or to trigger BMP
signaling. These approaches
include exogenous BMPRII
delivery, inhibition of miRNAs
negatively regulating BMPRII
stability and translation, inhibi-
tion of lysosomal degradation,
and delivery of exogenous BMP
ligands or BMP coactivators
among others
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2985BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
expression in vascular cells. In addition, Table 1 shows a
list of other miRNAs that are predicted to target BMPR2 in
silico.
Currently, there are different technologies to inhibit
aberrantly overexpressed miRNAs including the use of
antisense oligonucleotides, masking, sponges, erasers,
or decoys [92, 93]. In addition, administration of miRNA
mimics can enhance the expression of downregulated miR-
NAs [94]. These strategies are still under development
and more research is needed to establish how modulation
of miRNA function could offer therapeutic benefits in a
clinical setting while avoiding off-target effects, especially
in the liver, where systemically administrated miRNAs or
modulating compounds preferentially accumulate [95].
The delivery routes mostly used to target lung dis-
ease are local intranasal and intra-tracheal administration.
These naked miRNAs are directly delivered into the lung
with minimal systemic side effects [96]. Nevertheless, this
method remains ineffective and challenging due to the
complexity of the lung [97]. Recent advances in delivery
strategies, such as the use of liposomes, nanoparticles, or
virus, combined with improvements in chemically modi-
fying miRNAs, represent promising strategies to improve
lung miRNA delivery [98]. More than 20 miRNAs are cur-
rently in clinical trials, several in Phase III stage, highlight-
ing the potential of miRNA therapeutics to restore BMPRII
in PAH [99]. To date, the potential for miRNAs as a thera-
peutic tool is relatively limited. Further studies focusing on
the specificity, safety, efficiency, and stable systemic deliv-
ery of miRNAs into target cells or tissues will improve the
process of translating these findings to the clinic.
Translational regulation
Read-through premature STOP codons
Most BMPR2 mutations (~70%) are non-sense muta-
tions (frame-shift deletions and insertions) generated
by the insertion of a premature termination codon (PTC)
resulting in truncated reading frames which produce non-
functional proteins [7]. To prevent formation of truncated
proteins, mutated transcripts are directly degraded through
non-sense mediated decay (NMD) resulting in insufficient
amounts of the functional protein, which is produced only
by the wild-type allele (haplo-insufficiency) [100, 101].
NMD usually does not completely reduce the levels of
mutated transcripts and as a result truncated proteins per-
sist, and may exert a DN effect [102].
An approach aimed to correct these types of mutations
consists on the induction of PTC read-through. Read-
through of truncated mutations by aminoglycoside anti-
biotics, such as Gentamicin, has been extensively studied
and has reached the clinical trial stage for genetic disorders
such as cystic fibrosis [103] and Duchenne muscular dys-
trophy [104110]. Aminoglycoside antibiotics bind to the
decoding site of ribosomal RNA and eliminates the PTC
Fig. 4 miRNAs targeting
BMPRII in the vascular wall.
The illustration shows hypoxia
and BMPRII mutations as regu-
lators of miRNAs expression in
endothelial or smooth muscle
cells. These miRNAs negatively
regulate BMPRII expression
resulting in increased cell
proliferation and impaired
apoptosis. Green arrows
indicate activation, red arrows
represent inhibition, and black
arrows correspond to unknown
regulation. EC endothelial cells,
IL interleukin, miR micro RNA,
mut mutant, SMC smooth mus-
cle cell, STAT signal transducer,
and activator of transcription
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2986 M.Orriols et al.
1 3
by incorporating an amino acid to generate full-length pro-
teins [111]. Importantly, this read-through function of ami-
noglycosides does not affect normal translation because
of the presence of upstream and downstream regulatory
sequences around a normal termination codon that ensure
optimal efficiency of termination [112]. Gentamicin treat-
ment has been tested in lymphocytes derived from two
PAH patients with PTC mutations [113, 114]. The results
demonstrated increased amounts of full-length BMPRII
protein, a reduction in the mutated BMPRII product and
enhanced BMPRII downstream signaling. Although amino-
glycosides are commonly used in the clinic to treat infec-
tions and are safe when administered directly to the lung by
inhalation, several side effects of long-term treatments and/
or high concentrations of the drug have been shown.
Recently, a high-throughput screening for compounds
that suppress non-sense mutations identified a new small
molecule named Ataluren (PTC124) which mediates
read-through of premature stop codons without acute
side effects [115]. Aldred et al. have demonstrated that
after Ataluren treatment, BMPRII protein levels were
normalized and BMP-dependent phosphorylation of the
downstream target R-SMADs was increased in PAECs
and PASMCs from PAH patients. In addition, the hyper-
proliferative phenotype of these cells was reversed even
in the presence of significant non-sense mediated mRNA
decay. Although, further studies, including animal mod-
els, are required to explore the relevance of Ataluren
in vivo in a PAH context, the compound has emerged
as a promising therapeutic strategy for a subset of PAH
patients.
Table 1 miRNA targeting BMPRII expression
MicroRNA Cell type Function Model Expression References
miR-17/92 EC Interleukin-6 modulates the expres-
sion of the BMPRII through a novel
STAT3–microRNA Cluster 17/92
pathway
PAEC Brock etal. [13]
SMC Inhibition of miR-17 enhances
BMPRII expression and improves
heart and lung function in experi-
mental PH
PASMC Hypoxia-induced PH mice
MCT-induced PH rats
? Pullamsetti etal. [164]
miR-20A SMC Treatment with antagomiR-20a
restores functional levels of BMPRII
in pulmonary arteries and prevents
the development of vascular remod-
eling
PASMC Hypoxia-induced PH mice ? Brock etal. [165]
miR-21 EC Hypoxia and BMPRII signaling
independently upregulate miR-21. In
a reciprocal feedback loop, miR-21
downregulates BMP receptor type II
expression
PAEC Several rodent models of PH
miR-21-null mice
# Parikh etal. [166]
SMC BMPRII was downregulated in
PASMCs overexpressing miR-21
PASMC Hypoxia-induced PH mice # Yang etal. [167]
miR-125 EC Inhibition of miR-125a resulted in
upregulated BMPRII expression
accompanied by increased prolifera-
tion of EC
PAEC Hypoxia-induced PH mice
Plasma PAH patients
# Huber etal. [168]
miR-143/145 SMC miR-145 expression is increased in
primary PASMCs cultured from
patients with BMPRII mutations and
in the lungs of BMPRII-deficient
mice
PASMC Hypoxia-induced PH mice
BMPRII R899X knock-In Mice
miR-145 knock-Out Mice Lugn tis-
sue PAH patients
# Caruso etal. 2012 [169]
miR-302 SMC Inhibition of miR-302 by BMP4
increases BMPRII expression
and facilitates the BMP signaling
pathway
PASMC ? Kang etal. 2012 [170]
miR-181c cardiac Increased miR-181c expression
in human cardiac samples from
individuals with ventricular septal
defects (VSD) was correlated with
downregulated BMPRII levels
Human VSD cardiac samples # Li etal. [171]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2987BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
Protein processing regulation
Rescue ofBMPRII trafficking
30% of BMPR2 mutations are missense mutations leading
to single amino acid substitutions in a conserved domain
affecting the overall function of the protein [7]. Mutations
resulting in the substitution of cysteine residues in the
ligand binding and kinase domains disrupt protein folding
and trafficking of BMPRII to the cell surface leading to
retention of the mutant receptor in the endoplasmic reticu-
lum (ER) [113, 116]. A potentially promising therapeutic
strategy to increase the expression of BMPRII at the plasma
membrane is to enhance the activity of chaperones which
facilitate protein folding and trafficking. This can be done
by means of chemical chaperones such as sodium phenyl-
butyrate (4-PBA), probenecid, and tauroursodeoxycholic
acid (TUDCA). These have been shown to improve protein
trafficking via several distinct mechanisms [117126]. Dif-
ferent groups have demonstrated that treatment with chemi-
cal chaperones can partially restore cell surface expression
of BMPRII in ECs. As a result, BMP-induced SMAD 1/5/8
phosphorylation and the expression of the target gene ID1
is restored [127129]. These agents are showing promise
in clinical trials for other diseases caused by misfolded
proteins, such as cystic fibrosis. Since the currently used
chemical chaperones are federal drug administration (FDA)
approved drugs, there is an immediate translational poten-
tial to treat PAH patients [118, 130133]. However, fur-
ther invivo studies are required to test the viability of this
approach.
Even though chemical chaperones have the potential to
rescue the BMPRII mutants which are retained in the ER, it
remains to be investigated whether the amount of BMPRII
reaching the plasma membrane is enough to induce a clini-
cally relevant effect. Also, BMPRII with a protein-folding
defect expressed at the cell surface may have a dominant-
negative activity and adverse effects on BMP signaling
[127]. Moreover, patients harboring missense mutations
that affect the activity of the receptor (kinase domain) may
not benefit from this therapeutic strategy. Further research,
taking this mutation variability into account, is required to
determine which patients might benefit from this approach.
Inhibition oflysosomal degradation
The deciphering of mechanisms which regulate cell sur-
face expression levels of BMPRII are of potential clini-
cal importance, particularly those mechanisms that pre-
vent its rapid turnover and thereby restore downstream
BMPRII signaling and function. In this context, several
studies focused on the potential of targeting the degra-
dation of BMPRII by preventing lysosomal degradation
[134]. Durrington et al. have demonstrated that after
Kaposi sarcoma-associated herpesvirus infection,
BMPRII is ubiquitinated by K5 (membrane-associated
RING E3 viral ubiquitin protein ligase) leading to lyso-
somal degradation in primary cultured pulmonary vascu-
lar cells [134]. In addition, cells treated with the lysoso-
mal inhibitor concanamycin A exhibit increase levels of
BMPRII. Furthermore, through siRNA screening of the
NEDD4-like family E3 ubiquitin protein ligases, it was
found that knockdown of ITCH expression resulted in
increased BMPRII protein levels [134]. Whether ITCH
ubiquitinates BMPRII, leading to lysosomal degrada-
tion, has yet to be investigated. Satow etal. have dem-
onstrated that BMPRII is degraded via the proteosomal
pathway in HEK 293T cells, when it is associated with
Dullard phosphatase [135]. This might suggest that more
than one mechanism accounts for BMPRII proteasome-
mediated degradation. It is noteworthy that Satow etal.
used a BMPRII overexpression system, whereas Dur-
rington et al. studied the degradation of endogenous
BMPRII [134]. Different membrane trafficking pathways
such as endocytosis, phagocytosis, micropinocytosis, and
autophagy, use lysosomes for the digestion of diverse
macromolecules [136]. Caveolae-mediated endocytosis
affects multiple cellular signaling pathways by the redis-
tribution of transmembrane receptors and receptor-ligand
complexes [137139]. BMPRII localization has been
found to be regulated by CAV1 in vascular SMC [137].
Recently, ELAFIN (endogenous serine protease inhibitor)
treatment has been shown to prevent and reverse PAH in
the SU-hypoxia rat model. This occurs via elastase inhi-
bition and by promoting the interaction of BMPRII with
CAV1. Interestingly, when ELAFIN was combined with
BMP9, there was enhanced co-localization of CAV1 and
BMPRII on PAEC surfaces, which led at an increase in
BMP9-dependent SMAD1/5 phosphorylation and induc-
tion of ID1 [137]. Furthermore, transgenic mice overex-
pressing human ELAVIN in the cardiovascular system
(by placing ELAVIN expression under the control of the
pre-proendothelin-1 promoter), exhibited reduced SMC
proliferation and medial/intimal thickening after carotid
artery wire injury [140] and were protected from hypoxic
pulmonary hypertension [141]. In agreement with this,
peptidyl trifluoromethylketone serine elastase inhibitors
such as M249314 or ZD0892, have been used to prevent
and reverse PAH in the MCT rat model [142]. However,
the clinical use of these compounds was not pursued due
to hepatotoxicity. ELAFIN has been shown to inhibit
myocardial ischaemia-reperfusion injury induced during
coronary artery bypass graft surgery [143]. Even though
ELAFIN infusion was safe and resulted in >50% inhi-
bition of elastase activity in the first 24 h, myocardial
injury was not reduced after 48h. Based on the biology
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2988 M.Orriols et al.
1 3
of ischemia–reperfusion injury and PAH, we believe it
is worth testing whether ELAFIN together with BMP9
could reverses PAH in patients.
Interestingly, autophagy has also been found to be
involved in PAH. Autophagy (literally, “self-eating” in
Greek) is a highly regulated catabolic process that involves
sequestration and lysosomal degradation of cytosolic
components such as dysfunctional organelles, misfolded
proteins, lipid droplets, and invading pathogens [144].
Autophagy can be considered to be a general housekeep-
ing mechanism maintaining the integrity of intracellular
organelles and proteins. It is also triggered during devel-
opment, differentiation, infection, and stress conditions.
Thus, autophagy can be activated in the presence of dam-
aged organelles, protein aggregates, intracellular patho-
gens, hypoxia, amino acid starvation, reactive oxygen spe-
cies, and DNA damage [145]. Long etal. have shown that
in rats suffering from PAH induced by MCT treatment,
there is increased autophagy together with a decrease of
BMPRII protein expression [146]. Moreover, inhibition of
autophagic degradation by the lysosomal inhibitors chloro-
quine and hydroxychloroquine [147] prevents the develop-
ment of PAH as well as its progression. The authors dem-
onstrated that chloroquine and ATG5 (an autophagy protein
involved in the elongation and closure of the autophago-
somal membrane) knockdown inhibited proliferation and
increased apoptosis of PASMCs and these effects corre-
lated with increased levels of BMPRII via lysosomal inhi-
bition. Although autophagy seems to be involved in the
degradation of BMPRII, the exact mechanism by which
this takes place has yet to be elucidated. Chloroquine and
hydroxychloroquine have been widely utilized in malaria
prophylaxis [148]. They have also been used to treat rheu-
matoid arthritis and lupus erythematosus (as anti-inflam-
matory agents) [148]. Since inflammation is thought to be
a crucial second hit in PAH [149], these drugs might be
effective at inhibiting PAH progression by impairing the
degradation of BMPRII as well as inhibiting the inflam-
matory response. However, it is important to keep in mind
that since lysosomal degradation is a ubiquitous cellular
mechanism for regulating protein processing, this approach
can lead to widespread and non-specific off-target effects
independent of BMPRII signaling. Therefore, an improved
understanding of the molecular mechanisms underlying
BMPRII turnover is required for the development of more
directed interventions.
BMPRII signaling regulation
Delivery ofexogenous BMP ligand
As mentioned previously, BMP signaling in the vascular
endothelium is mainly activated by BMP2, 4, 6, 9, and 10
[32]. In particular, BMP9 and BMP10 appear to play an
important role in the vasculature due to their presence in
the circulation and based on the fact that they are known
to signal through receptors expressed on the endothelium,
such as ALK1 and BMPRII or ACVRIIB. Therefore, the
stimulation of BMP signaling with exogenous recombi-
nant ligand is an interesting approach for PAH treatment
[11, 150]. Long et al. have shown that BMP9 prevents
apoptosis and enhances the integrity of ECs in PAECs and
blood outgrowth ECs from PAH patients. Furthermore,
therapeutic BMP9 delivery prevents and reverses PAH in
several mouse models [70]. BMP10 is the least studied
BMP ligand; however, it may present a better treatment
than BMP9 since it binds to ALK1 and BMPRII with
higher affinity and because of its lack of osteogenic activ-
ity in vitro [151]. Further studies have to be performed
to evaluate the delivery strategies, efficiency, and poten-
tial side effects of BMP9 and BMP10 invivo. Finally, the
development of a small peptide mimetics of BMP9 or
BMP10, with an increased affinity for the receptor, is a the-
oretical alternative for efficiently activating BMP signaling
and thereby reversing PAH [150].
Enhance downstream SMAD signaling
An additional approach to reverse the effect of mutant
BMPRII is use small molecules to enhance signaling of
the wild-type functional proteins. Sildenafil is a phospho-
diesterase type-5 (PDE5) inhibitor currently used in the
clinic for PAH treatment [152154]. Its mode of action is to
block the degradation of cyclic guanosine monophosphate
(cGMP) resulting in corrective vasodilatory and anti-pro-
liferative effects in the arterial wall [155]. Furthermore, it
has been described that protein kinase G (PKG) activated
by cGMP, is a modulator of BMP signaling [156] and that
PASMCs expressing a BMPRII mutant, showed an increase
in BMP signaling after Sildenadil treatment via a cGMP/
PKG-dependent mechanism. In addition, in vivo stud-
ies confirmed that Sildenafil treatment enhanced BMP
signaling and partially reversed PAH development in the
MCT rat model [157, 158]. Although Sildenafil therapy
during 12 weeks improves multiple clinical symptoms
in PAH patients, it appears to have no effect on reducing
either mortality or serious adverse events [159]. Further-
more, the long-term efficiency and safety of Sildenafil
therapy in PAH requires further studies based on large and
well-designed clinical trials [159].
Another promising strategy is to identify compounds in
drug libraries that activate BMP/SMAD signaling. FK506
(Tacrolimus) was identified as the best BMP coactiva-
tor among 3756 FDA-approved drugs and bioactive com-
pounds (using a high-throughput BMP/SMAD-driven tran-
scriptional reporter assay) [160]. FK506 promotes BMP
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2989BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
signaling and endothelial-specific gene regulation of genes
such as APELIN. This occurs even in the absence of exog-
enous ligand via a dual mechanism of action: acting as an
inhibitor of phosphatase CALCINEURIN and binding FK-
binding protein-12 (FKBP12), a repressor of BMP signal-
ing. FK506 promotes the release of FKBP12 from the type
I receptor which leads to activation of SMAD1/5 down-
stream of BMP as well as MAPK signaling and ID1 gene
regulation [161]. Furthermore, FK506 treatment increases
ALK1 and ENDOGLIN expression in ECs [162]. Recently,
a randomized, double-blind, placebo-controlled phase IIa
trial was performed to investigate the efficacy of FK506
treatment in three patients with end-stage PAH. The results
suggest a potential clinical benefit of low-dose FK506 (the
evidence being that patients demonstrated cardiac function
stabilization and required less intensive hospital care for
RV failure despite the severity of the illness). It was also
found that changes in serologic biomarkers indicated that
BMPRII had been successfully targeted [163]. However,
these results are based on a limited group of patients and
the efficacy of this therapy must be validated in appropri-
ate, well-designed clinical trials. FK506 (also known as
Tacrolimus) is an immunosuppressive drug with a known
pharmacokinetic and toxicity profile. It is widely used in
solid organ transplantations to lower the risk of organ rejec-
tion [164]. High doses of FK506 caused systemic hyper-
tension and transplant vasculopathy in animal models
[165]. Also, organ transplant patients treated with FK506,
have an increased risk of renal injury, which might occur
due to the inhibition of calcineurin expression in the kid-
ney [166168]. In contrast, low doses of FK506 did not
induce systemic hypertension in animal models, even after
3 weeks of treatment. FK506 has shown significant clinical
benefits, nonetheless long-term use of this agent for treat-
ing PAH still needs to be rigorously monitored for toxicity
effects.
Conclusions andperspectives
Exogenous BMPRII delivery to ECs has been shown to
be an effective means to restore BMPRII expression and
function [77, 78]. An interesting approach, which is yield-
ing promising results in mice, is to deliver BMPRII spe-
cifically to ECs using BMPRII adenoviral vectors carry-
ing a bi-specific conjugate antibody that targets the virus
to ACE, a membrane-bound protease highly expressed on
pulmonary endothelial cells [77, 78]. One of the draw-
backs of this strategy is the use of two components namely,
adenovirus and antibody. Additional restrictions related
to the use of viral transduction such as safety, specificity,
and delivery of sufficient protein to revert the phenotype
must also be taken into consideration. The utilization of
CRISPR/Cas9 may overcome some of these limitations, for
instance by minimizing the risk that the foreign gene will
be integrated in the wrong place in the genome. Further-
more, it will place the gene under the control of its natu-
ral promoter. However, the delivery of CRISPR/Cas9 into
the patient is still challenging and the Cas9 enzyme could
cleave at unwanted locations. Similarly, the use of miRNAs
targeting BMPRII has to be evaluated for off-target effects
and an effective delivery system has to be found in order
to consider this approach as a promising treatment. A solu-
tion for both plasmid DNA and miRNA delivery might be
the use of liposomes [169] or iTOP (induced transduction
by osmocytosis and propanebetaine), which is an active
uptake mechanism in which NaCl-mediated hyperosmo-
larity together with propanebetaine triggers the uptake of
macromolecules [170]. Another therapeutic strategy is the
use of FDA-approved drugs that have been found to be ben-
eficial in PAH mice models or similar diseases. Ataluren,
for example, allows the cellular machinery to read-through
premature stop codons [115]. Although most of the BMPR2
mutations (~70%) are non-sense mutations, not all patients
will benefit from this approach. Nevertheless, further
invivo studies are worth pursuing in the context of PAH.
Likewise, clinical trials using chloroquine have to be per-
formed to test its effectiveness in PAH patients. The use of
chloroquine has to be carefully evaluated because blocking
lysosomal degradation might trigger non-specific off-target
effects when used as a long-term treatment. An alterna-
tive drug showing significant clinical benefits for PAH is
FK506/Tacrolimus. However, it still needs to be monitored
for side effects since it is an immunosuppressive drug (cur-
rently utilized after allogeneic organ transplant). Moreover,
the effectiveness of FK506 at low doses has to be rigor-
ously tested.
It is important to highlight that although several drugs
showed beneficial outcomes in animal models, most of the
drugs have failed in the clinic. In light of this, we should
focus on a more personalized approach which takes into
account the co-existence of modifier genes, infections,
toxic exposure, inflammation, or alterations in estrogen
metabolism. Combining treatments which target not only
BMPRII signaling but also inflammation and hypoxia
should improve outcomes. Lastly, the use of human exvivo
models such as lung or vessel on a chip [171] could be ben-
eficial for drug discovery and efficacy testing in the context
of PAH. We anticipate that such models may improve the
relevance of pre-clinical results by using patient derived
cells, especially since animal models of PAH are frequently
difficult to translate into clinical practice.
Taken together, previously discussed data suggest that
modulation of BMPRII signaling in PAH is a promising
alternative that could prevent and reverse pulmonary vascu-
lar remodeling. However, different therapeutic approaches
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2990 M.Orriols et al.
1 3
aimed at to increasing the levels of BMPRII signaling are
needed, and these approaches will depend on the particular
genetic background of each patient. In addition, for more
efficient treatments, targeting other genetic and environ-
mental factors that contribute to the disease must be taken
into consideration. In this regard, modulators of the inflam-
matory response and estrogen metabolism could be used to
help restore BMPRII signaling.
Acknowledgements We acknowledge the support from the Neth-
erlands CardioVascular Research Initiative: the Dutch Heart Foun-
dation, Dutch Federation of University Medical Centers, the Neth-
erlands Organisation for Health Research and Development, and the
Royal Netherlands Academy of Sciences.
Open Access This article is distributed under the terms of the
Creative Commons Attribution 4.0 International License (http://
creativecommons.org/licenses/by/4.0/), which permits unrestricted
use, distribution, and reproduction in any medium, provided you give
appropriate credit to the original author(s) and the source, provide a
link to the Creative Commons license, and indicate if changes were
made.
References
1. Rosenkranz S (2015) Pulmonary hypertension 2015: current
definitions, terminology, and novel treatment options. Clin Res
Cardiol 104(3):197–207. doi:10.1007/s00392-014-0765-4
2. Guignabert C, Dorfmuller P (2013) Pathology and pathobiol-
ogy of pulmonary hypertension. Semin Respir Crit Care Med
34(5):551–559. doi:10.1055/s-0033-1356496
3. Farber HW, Loscalzo J (2004) Pulmonary arterial hypertension.
N Engl J Med 351(16):1655–1665. doi:10.1056/NEJMra035488
4. Thompson K, Rabinovitch M (1996) Exogenous leukocyte and
endogenous elastases can mediate mitogenic activity in pul-
monary artery smooth muscle cells by release of extracellular-
matrix bound basic fibroblast growth factor. J Cell Physiol
166(3):495–505. doi:10.1002/(sici)1097-4652(199603)
5. Consortium IP, Lane KB, Machado RD, Pauciulo MW, Thom-
son JR, Phillips JA, Loyd JE, Nichols WC, Trembath RC (2000)
Heterozygous germline mutations in BMPR2, encoding a TGF-
beta receptor, cause familial primary pulmonary hypertension.
Nat Genet 26(1):81–84. doi:10.1038/79226
6. Deng Z, Morse JH, Slager SL, Cuervo N, Moore KJ, Venetos
G, Kalachikov S, Cayanis E, Fischer SG, Barst RJ, Hodge SE,
Knowles JA (2000) Familial primary pulmonary hypertension
(gene PPH1) is caused by mutations in the bone morphoge-
netic protein receptor-II gene. Am J Hum Genet 67(3):737–744.
doi:10.1086/303059
7. Machado RD, Aldred MA, James V, Harrison RE, Patel B,
Schwalbe EC, Gruenig E, Janssen B, Koehler R, Seeger W,
Eickelberg O, Olschewski H, Elliott CG, Glissmeyer E, Car-
lquist J, Kim M, Torbicki A, Fijalkowska A, Szewczyk G,
Parma J, Abramowicz MJ, Galie N, Morisaki H, Kyotani S,
Nakanishi N, Morisaki T, Humbert M, Simonneau G, Sitbon O,
Soubrier F, Coulet F, Morrell NW, Trembath RC (2006) Muta-
tions of the TGF-beta type II receptor BMPR2 in pulmonary
arterial hypertension. Hum Mutat 27(2):121–132. doi:10.1002/
humu.20285
8. Thomson JR, Machado RD, Pauciulo MW, Morgan NV, Hum-
bert M, Elliott GC, Ward K, Yacoub M, Mikhail G, Rogers P,
Newman J, Wheeler L, Higenbottam T, Gibbs JS, Egan J, Cro-
zier A, Peacock A, Allcock R, Corris P, Loyd JE, Trembath RC,
Nichols WC (2000) Sporadic primary pulmonary hypertension
is associated with germline mutations of the gene encoding
BMPR-II, a receptor member of the TGF-beta family. J Med
Genet 37(10):741–745
9. Machado RD, Southgate L, Eichstaedt CA, Aldred MA, Aus-
tin ED, Best DH, Chung WK, Benjamin N, Elliott CG, Eyries
M, Fischer C, Gräf S, Hinderhofer K, Humbert M, Keiles SB,
Loyd JE, Morrell NW, Newman JH, Soubrier F, Trembath RC,
Viales RR, Grünig E (2015) Pulmonary arterial hypertension:
a current perspective on established and emerging molecular
genetic defects. Hum Mutat 36(12):1113–1127. doi:10.1002/
humu.22904
10. Cogan J, Austin E, Hedges L, Womack B, West J, Loyd
J, Hamid R (2012) Role of BMPR2 alternative splic-
ing in heritable pulmonary arterial hypertension pen-
etrance. Circulation 126(15):1907–1916. doi:10.1161/
CIRCULATIONAHA.112.106245
11. Guignabert C, Bailly S, Humbert M (2017) Restoring BMPRII
functions in pulmonary arterial hypertension: opportuni-
ties, challenges and limitations. Expert Opin Ther Targets
21(2):181–190. doi:10.1080/14728222.2017.1275567
12. Song Y, Coleman L, Shi J, Beppu H, Sato K, Walsh K, Los-
calzo J, Zhang Y-Y (2008) Inflammation, endothelial injury, and
persistent pulmonary hypertension in heterozygous BMPR2-
mutant mice. Am J Physiol Heart Circ Physiol 295(2):H677–
H690. doi:10.1152/ajpheart.91519.2007
13. Austin ED, Cogan JD, West JD, Hedges LK, Hamid R,
Dawson EP, Wheeler LA, Parl FF, Loyd JE, Phillips JA
(2009) Alterations in oestrogen metabolism: implica-
tions for higher penetrance of familial pulmonary arterial
hypertension in females. Eur Respir J 34(5):1093–1099.
doi:10.1183/09031936.00010409
14. Mair KM, Yang XD, Long L, White K, Wallace E, Ewart M-A,
Docherty CK, Morrell NW, MacLean MR (2015) Sex affects
bone morphogenetic protein type II receptor signaling in pul-
monary artery smooth muscle cells. Am J Respir Crit Care Med
191(6):693–703. doi:10.1164/rccm.201410-1802OC
15. Brock M, Trenkmann M, Gay RE, Michel BA, Gay S,
Fischler M, Ulrich S, Speich R, Huber LC (2009) Interleu-
kin-6 modulates the expression of the bone morphogenic pro-
tein receptor type II through a novel STAT3-microRNA clus-
ter 17/92 pathway. Circ Res 104(10):1184–1191. doi:10.1161/
circresaha.109.197491
16. Kim CW, Song H, Kumar S, Nam D, Kwon HS, Chang KH,
Son DJ, Kang DW, Brodie SA, Weiss D, Vega JD, Alberts-Grill
N, Griendling K, Taylor WR, Jo H (2013) Anti-inflammatory
and antiatherogenic role of BMP receptor II in endothelial cells.
Arterioscler Thromb Vasc Biol 33(6):1350–1359. doi:10.1161/
ATVBAHA.112.300287
17. Soon E, Crosby A, Southwood M, Yang P, Tajsic T, Toshner M,
Appleby S, Shanahan CM, Bloch KD, Pepke-Zaba J, Upton P,
Morrell NW (2015) Bone morphogenetic protein receptor type
II deficiency and increased inflammatory cytokine production.
A gateway to pulmonary arterial hypertension. Am J Respir Crit
Care Med 192(7):859–872. doi:10.1164/rccm.201408-1509OC
18. Burton VJ, Ciuclan LI, Holmes AM, Rodman DM, Walker C,
Budd DC (2011) Bone morphogenetic protein receptor II regu-
lates pulmonary artery endothelial cell barrier function. Blood
117(1):333–341. doi:10.1182/blood-2010-05-285973
19. Frumkin LR (2012) The pharmacological treatment of pul-
monary arterial hypertension. Pharmacol Rev 64(3):583–620.
doi:10.1124/pr.111.005587
20. Perrin S, Chaumais M-C, O’Connell C, Amar D, Savale L,
Jaïs X, Montani D, Humbert M, Simonneau G, Sitbon O
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2991BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
(2015) New pharmacotherapy options for pulmonary arterial
hypertension. Expert Opin Pharmacother 16(14):2113–2131.
doi:10.1517/14656566.2015.1074177
21. Morrell NW, Archer SL, Defelice A, Evans S, Fiszman M,
Martin T, Saulnier M, Rabinovitch M, Schermuly R, Stew-
art D, Truebel H, Walker G, Stenmark KR (2013) Antici-
pated classes of new medications and molecular targets for
pulmonary arterial hypertension. Pulm Circ 3(1):226–244.
doi:10.4103/2045-8932.109940
22. Stenmark KR, Rabinovitch M (2010) Emerging therapies for
the treatment of pulmonary hypertension. Pediatr Crit Care
Med 11(2):S85–S90. doi:10.1097/PCC.0b013e3181c76db3
23. Balliga RS, MacAllister RJ, Hobbs AJ (2011)
New perspectives for the treatment of pulmo-
nary hypertension. Br J Pharmacol 163(1):125–140.
doi:10.1111/j.1476-5381.2010.01164.x
24. Liu Y, Ren W, Warburton R, Toksoz D, Fanburg BL (2009)
Serotonin induces Rho/ROCK-dependent activation of Smads
1/5/8 in pulmonary artery smooth muscle cells. FASEB J
23(7):2299–2306. doi:10.1096/.08-127910
25. Akhurst RJ, Padgett RW (2015) Matters of context guide
future research in TGFβ superfamily signaling. Sci Signal
8(399):re10. doi:10.1126/scisignal.aad0416
26. Sánchez-Duffhues G, Hiepen C, Knaus P, Ten Dijke P (2015)
Bone morphogenetic protein signaling in bone homeostasis.
Bone 80:43–59. doi:10.1016/j.bone.2015.05.025
27. Morrell NW, Bloch DB,Ten Dijke P, Goumans M-JTH, Hata
A, Smith J, Yu PB, Bloch KD (2016) Targeting BMP signal-
ling in cardiovascular disease and anaemia. Nat Rev Cardiol
13(2):106–120. doi:10.1038/nrcardio.2015.156
28. Kawabata M, Imamura T, Miyazono K (1998) Signal trans-
duction by bone morphogenetic proteins. Cytokine Growth
Factor Rev 9(1):49–61
29. Miyazono K, Kamiya Y, Morikawa M (2010) Bone morpho-
genetic protein receptors and signal transduction. J Biochem
147(1):35–51. doi:10.1093/jb/mvp148
30. Heldin CH, Miyazono K, Ten Dijke P (1997) TGF-beta sig-
nalling from cell membrane to nucleus through SMAD pro-
teins. Nature 390(6659):465–471. doi:10.1038/37284
31. Derynck R, Zhang YE (2003) Smad-dependent and Smad-
independent pathways in TGF-beta family signalling. Nature
425(6958):577–584. doi:10.1038/nature02006
32. David L, Feige J-J, Bailly S (2009) Emerging role of bone
morphogenetic proteins in angiogenesis. Cytokine Growth
Factor Rev 20(3):203–212. doi:10.1016/j.cytogfr.2009.05.001
33. Feng X-H, Derynck R (2005) Specificity and versatility in
tgf-beta signaling through Smads. Annu Rev Cell Dev Biol
21:659–693. doi:10.1146/annurev.cellbio.21.022404.142018
34. Shi Y, Massagué J (2003) Mechanisms of TGF-beta signaling
from cell membrane to the nucleus. Cell 113(6):685–700
35. Zhang YE (2009) Non-Smad pathways in TGF-beta signal-
ing. Cell Res 19(1):128–139. doi:10.1038/cr.2008.328
36. Moustakas A, Heldin C-H (2005) Non-Smad TGF-beta sig-
nals. J Cell Sci 118(Pt 16):3573–3584. doi:10.1242/jcs.02554
37. Mu Y, Gudey SK, Landström M (2012) Non-Smad signal-
ing pathways. Cell Tissue Res 347(1):11–20. doi:10.1007/
s00441-011-1201-y
38. Massagué J, Wotton D (2000) Transcriptional control by the
TGF-beta/Smad signaling system. EMBO J 19(8):1745–1754.
doi:10.1093/emboj/19.8.1745
39. Mulder KM (2000) Role of ras and mapks in TGFbeta signal-
ing. Cytokine Growth Factor Rev 11(1–2):23–35
40. Canalis E, Economides AN, Gazzerro E (2003) Bone mor-
phogenetic proteins, their antagonists, and the skeleton.
Endocr Rev 24(2):218–235. doi:10.1210/er.2002-0023
41. Sánchez-Duffhues G, Hiepen C, Knaus P, Ten Dijke P
(2016) Emerging regulators of BMP bioavailability. Bone.
doi:10.1016/j.bone.2016.01.022
42. Cai J, Pardali E, Sánchez-Duffhues G, Ten Dijke P (2012) BMP
signaling in vascular diseases. FEBS Lett 586(14):1993–2002.
doi:10.1016/j.febslet.2012.04.030
43. Gao X, Cao Y, Staloch DA, Gonzales MA, Aronson JF, Chao
C, Hellmich MR, Ko TC (2014) Bone morphogenetic protein
signaling protects against cerulein-induced pancreatic fibrosis.
PLoS ONE 9(2):e89114. doi:10.1371/journal.pone.0089114
44. Chen NY, S DC, Luo F, Weng T, Le TT, A MH, Philip K,
Molina JG, Garcia-Morales LJ, Cao Y, Ko TC, Amione-Guerra
J, Al-Jabbari O, Bunge RR, Youker K, Bruckner BA, Hamid
R, Davies J, Sinha N, Karmouty-Quintana H (2016) Mac-
rophage bone morphogenic protein receptor 2 depletion in idi-
opathic pulmonary fibrosis and Group III pulmonary hyperten-
sion. Am J Physiol Lung Cell Mol Physiol 311(2):L238–L254.
doi:10.1152/ajplung.00142.2016
45. De Vinuesa AG, Abdelilah-Seyfried S, Knaus P, Zwijsen A,
Bailly S (2016) BMP signaling in vascular biology and dys-
function. Cytokine Growth Factor Rev 27:65–79. doi:10.1016/j.
cytogfr.2015.12.005
46. Stenmark KR, Davie N, Frid M, Gerasimovskaya E, Das M
(2006) Role of the adventitia in pulmonary vascular remod-
eling. Physiology (Bethesda) 21:134–145. doi:10.1152/
physiol.00053.2005
47. Humbert M, Morrell NW, Archer SL, Stenmark KR, MacLean
MR, Lang IM, Christman BW, Weir EK, Eickelberg O, Voel-
kel NF, Rabinovitch M (2004) Cellular and molecular pathobi-
ology of pulmonary arterial hypertension. J Am Coll Cardiol
43(12):13S-24 S. doi:10.1016/j.jacc.2004.02.029
48. Lilly B (2014) We have contact: endothelial cell-smooth mus-
cle cell interactions. Physiology (Bethesda) 29(4):234–241.
doi:10.1152/physiol.00047.2013
49. Christman BW, McPherson CD, Newman JH, King GA, Ber-
nard GR, Groves BM, Loyd JE (1992) An imbalance between
the excretion of thromboxane and prostacyclin metabo-
lites in pulmonary hypertension. N Engl J Med 327:70–75.
doi:10.1056/NEJM199207093270202
50. David L, Mallet C, Mazerbourg S, Feige J-J, Bailly S (2007)
Identification of BMP9 and BMP10 as functional activa-
tors of the orphan activin receptor-like kinase 1 (ALK1) in
endothelial cells. Blood 109(5):1953–1961. doi:10.1182/
blood-2006-07-034124
51. Scharpfenecker M, van Dinther M, Liu Z, van Bezooijen RL,
Zhao Q, Pukac L, Löwik CWGM, Ten Dijke P (2007) BMP-9
signals via ALK1 and inhibits bFGF-induced endothelial cell
proliferation and VEGF-stimulated angiogenesis. J Cell Sci
120(Pt 6):964–972. doi:10.1242/jcs.002949
52. Upton PD, Long L, Trembath RC, Morrell NW (2008) Func-
tional characterization of bone morphogenetic protein binding
sites and Smad1/5 activation in human vascular cells. Mol Phar-
macol 73(2):539–552. doi:10.1124/mol.107.041673
53. Shintani M, Yagi H, Nakayama T, Saji T, Matsuoka R (2009) A
new nonsense mutation of SMAD8 associated with pulmonary
arterial hypertension. J Med Genet 46:331–337. doi:10.1136/
jmg.2008.062703
54. Nasim MT, Ogo T, Ahmed M, Randall R, Chowdhury HM,
Snape KM, Bradshaw TY, Southgate L, Lee GJ, Jackson I, Lord
GM, Gibbs JS, Wilkins MR, Ohta-Ogo K, Nakamura K, Girerd
B, Coulet F, Soubrier F, Humbert M, Morrell NW, Trembath
RC, Machado RD (2011) Molecular genetic characterization of
SMAD signaling molecules in pulmonary arterial hypertension.
Hum Mutat 32(12):1385–1389. doi:10.1002/humu.21605
55. Wang G, Fan R, Ji R, Zou W, Penny DJ, Varghese NP, Fan Y
(2016) Novel homozygous BMP9 nonsense mutation causes
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2992 M.Orriols et al.
1 3
pulmonary arterial hypertension: a case report. BMC Pulm
Med 16:17. doi:10.1186/s12890-016-0183-7
56. Shintani M, Yagi H, Nakayama T, Saji T, Matsuoka R (2009)
A new nonsense mutation of SMAD8 associated with pul-
monary arterial hypertension. J Med Genet 46(5):331–337.
doi:10.1136/jmg.2008.062703
57. Harrison RE, Berger R, Haworth SG, Tulloh R, Mache CJ,
Morrell NW, Aldred MA, Trembath RC (2005) Transform-
ing growth factor-β receptor mutations and pulmonary arte-
rial hypertension in childhood. Circulation 111(4):435–441.
doi:10.1161/01.CIR.0000153798.78540.87
58. Girerd B, Montani D, Coulet F, Sztrymf B, Yaici A, Jais X,
Tregouet D, Reis A, Drouin-Garraud V, Fraisse A, Sitbon
O, O’Callaghan DS, Simonneau G, Soubrier F, Humbert M
(2010) Clinical outcomes of pulmonary arterial hyperten-
sion in patients carrying an ACVRL1 (ALK1) mutation.
Am J Respir Crit Care Med 181(8):851–861. doi:10.1164/
rccm.200908-1284OC
59. Pousada G, Baloira A, Fontan D, Nunez M, Valverde D
(2016) Mutational and clinical analysis of the ENG gene in
patients with pulmonary arterial hypertension. BMC Genet
17(1):72. doi:10.1186/s12863-016-0384-3
60. Austin ED, Ma L, LeDuc C, Berman Rosenzweig E, Borczuk
A, Phillips JA, Palomero T, Sumazin P, Kim HR, Talati MH,
West J, Loyd JE, Chung WK (2012) Whole exome sequencing
to identify a novel gene (caveolin-1) associated with human
pulmonary arterial hypertension. Circ Cardiovasc Genet
5(3):336–343. doi:10.1161/circgenetics.111.961888
61. Ma L, Roman-Campos D, Austin ED, Eyries M, Sampson
KS, Soubrier F, Germain M, Trégouët D-A, Borczuk A,
Rosenzweig EB, Girerd B, Montani D, Humbert M, Loyd JE,
Kass RS, Chung WK (2013) A novel channelopathy in pul-
monary arterial hypertension. N Engl J Med 369(4):351–361.
doi:10.1056/NEJMoa1211097
62. Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A,
Pechoux C, Bogaard HJ, Dorfmuller P, Remy S, Lecerf F,
Plante S, Chat S, Fadel E, Houssaini A, Anegon I, Adnot S,
Simonneau G, Humbert M, Cohen-Kaminsky S, Perros F
(2015) Endothelial-to-mesenchymal transition in pulmonary
hypertension. Circulation 131(11):1006–1018. doi:10.1161/
CIRCULATIONAHA.114.008750
63. Hopper RK, Moonen JR, Diebold I, Cao A, Rhodes CJ, Tojais
NF, Hennigs JK, Gu M, Wang L, Rabinovitch M (2016) In
pulmonary arterial hypertension, reduced BMPR2 promotes
endothelial-to-mesenchymal transition via HMGA1 and its
target slug. Circulation 133(18):1783–1794. doi:10.1161/
CIRCULATIONAHA.115.020617
64. Maron BA, Loscalzo J (2013) Pulmonary hypertension:
pathophysiology and signaling pathways. Handb Exp Phar-
macol 218:31–58. doi:10.1007/978-3-642-38664-0_2
65. Voelkel NF, Tuder RM (2000) Hypoxia-induced pulmonary
vascular remodeling: a model for what human disease? J Clin
Invest 106(6):733–738. doi:10.1172/JCI11144
66. Campian ME, Hardziyenka M, Michel MC, Tan HL (2006)
How valid are animal models to evaluate treatments for pul-
monary hypertension? Naunyn Schmiedebergs Arch Pharma-
col 373(6):391–400. doi:10.1007/s00210-006-0087-9
67. Guihaire J, Bogaard HJ, Flecher E, Noly PE, Mercier O,
Haddad F, Fadel E (2013) Experimental models of right heart
failure: a window for translational research in pulmonary
hypertension. Semin Respir Crit Care Med 34(5):689–699.
doi:10.1055/s-0033-1355444
68. West J, Fagan K, Steudel W, Fouty B, Lane K, Har-
ral J, Hoedt-Miller M, Tada Y, Ozimek J, Tuder R, Rod-
man DM (2004) Pulmonary hypertension in transgenic
mice expressing a dominant-negative BMPRII gene in
smooth muscle. Circ Res 94(8):1109–1114. doi:10.1161/01.
res.0000126047.82846.20
69. Hong K-H, Lee YJ, Lee E, Park SO, Han C, Beppu H, Li E,
Raizada MK, Bloch KD, Oh SP (2008) Genetic ablation of
the BMPR2 gene in pulmonary endothelium is sufficient to
predispose to pulmonary arterial hypertension. Circulation
118(7):722–730. doi:10.1161/circulationaha.107.736801
70. Long L, Ormiston ML, Yang X, Southwood M, Graf S,
Machado RD, Mueller M, Kinzel B, Yung LM, Wilkinson JM,
Moore SD, Drake KM, Aldred MA, Yu PB, Upton PD, Mor-
rell NW (2015) Selective enhancement of endothelial BMPR-II
with BMP9 reverses pulmonary arterial hypertension. Nat Med
21(7):777–785. doi:10.1038/nm.3877
71. Maarman G, Lecour S, Butrous G, Thienemann F, Sliwa K
(2013) A comprehensive review: the evolution of animal mod-
els in pulmonary hypertension research; are we there yet?. Pulm
Circ 3(4):739–756. doi:10.1086/674770
72. West J, Harral J, Lane K, Deng Y, Ickes B, Crona D, Albu S,
Stewart D, Fagan K (2008) Mice expressing BMPR2R899X
transgene in smooth muscle develop pulmonary vascular
lesions. Am J Physiol Lung Cell Mol Physiol 295(5):L744–
L755. doi:10.1152/ajplung.90255.2008
73. de Jesus Perez VA, Alastalo T-P, Wu JC, Axelrod JD, Cooke
JP, Amieva M, Rabinovitch M (2009) Bone morphogenetic pro-
tein 2 induces pulmonary angiogenesis via Wnt-beta-catenin
and Wnt-RhoA-Rac1 pathways. J Cell Biol 184(1):83–99.
doi:10.1083/jcb.200806049
74. Hansmann G, de Jesus Perez VA, Alastalo T-P, Alvira CM,
Guignabert C, Bekker JM, Schellong S, Urashima T, Wang
L, Morrell NW, Rabinovitch M (2008) An antiprolifera-
tive BMP-2/PPARgamma/apoE axis in human and murine
SMCs and its role in pulmonary hypertension. J Clin Invest
118(5):1846–1857. doi:10.1172/jci32503
75. Sung YK, Yuan K, de Jesus Perez VA (2016) Novel approaches
to pulmonary arterial hypertension drug discovery. Expert Opin
Drug Discov 11(4):407–414. doi:10.1517/17460441.2016.1153
625
76. Madonna R, Cocco N (2016) Novel strategies in the treat-
ment of pulmonary arterial hypertension. Curr Drug Targets
17(7):817–823
77. Reynolds AM, Xia W, Holmes MD, Hodge SJ, Danilov S,
Curiel DT, Morrell NW, Reynolds PN (2007) Bone morpho-
genetic protein type 2 receptor gene therapy attenuates hypoxic
pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol
292(5):L1182–L1192. doi:10.1152/ajplung.00020.2006
78. Reynolds AM, Holmes MD, Danilov SM, Reynolds
PN (2012) Targeted gene delivery of BMPR2 attenu-
ates pulmonary hypertension. Eur Respir J 39(2):329–343.
doi:10.1183/09031936.00187310
79. Harper RL, Reynolds AM, Bonder CS, Reynolds PN (2016)
BMPR2 gene therapy for PAH acts via Smad and non-Smad
signalling. Respirology 21(4):727–733. doi:10.1111/resp.12729
80. McMurtry MS, Moudgil R, Hashimoto K, Bonnet S, Michelakis
ED, Archer SL (2007) Overexpression of human bone morpho-
genetic protein receptor 2 does not ameliorate monocrotaline
pulmonary arterial hypertension. Am J Physiol Lung Cell Mol
Physiol 292(4):L872–L878. doi:10.1152/ajplung.00309.2006
81. Doi K, Takeuchi Y (2015) Gene therapy using retrovirus vec-
tors: vector development and biosafety at clinical trials. Uirusu
65(1):27–36. doi:10.2222/jsv.65.27
82. Reynolds PN (2011) Viruses in pharmaceutical research: pul-
monary vascular disease. Mol Pharm 8(1):56–64. doi:10.1021/
mp1003477
83. Brunetti-Pierri N, Ng T, Iannitti D, Cioffi W, Stapleton G,
Law M, Breinholt J, Palmer D, Grove N, Rice K, Bauer C,
Finegold M, Beaudet A, Mullins C, Ng P (2013) Transgene
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2993BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
expression up to 7 years in nonhuman primates follow-
ing hepatic transduction with helper-dependent adenovi-
ral vectors. Hum Gene Ther 24(8):761–765. doi:10.1089/
hum.2013.071
84. Gubrij IB, Martin SR, Pangle AK, Kurten R, Johnson LG
(2014) Attenuation of monocrotaline-induced pulmonary
hypertension by luminal adeno-associated virus serotype
9 gene transfer of prostacyclin synthase. Hum Gene Ther
25(6):498–505. doi:10.1089/hum.2013.187
85. Kim J-D, Lee A, Choi J, Park Y, Kang H, Chang W, Lee
M-S, Kim J (2015) Epigenetic modulation as a therapeutic
approach for pulmonary arterial hypertension. Exp Mol Med
47:e175. doi:10.1038/emm.2015.45
86. Xu X-F, Cheng F, Du L-Z (2011) Epigenetic regulation of
pulmonary arterial hypertension. Hypertens Res 34(9):981–
986. doi:10.1038/hr.2011.79
87. Gurha P (2016) MicroRNAs in cardiovascular disease. Curr
Opin Cardiol. doi:10.1097/hco.0000000000000280
88. Welten SMJ, Goossens EAC, Quax PHA, Nossent AY (2016)
The multifactorial nature of microRNAs in vascular remodel-
ling. Cardiovasc Res. doi:10.1093/cvr/cvw039
89. Kurakula K, Goumans M-J, Ten Dijke P (2015) Regulatory
RNAs controlling vascular (dys)function by affecting TGF-β
family signalling. EXCLI J 14:832–850. doi:10.17179/
excli2015-423
90. Courboulin A, Ranchoux B, Cohen-Kaminsky S, Perros F,
Bonnet S (2016) MicroRNA networks in pulmonary arte-
rial hypertension: share mechanisms with cancer? Curr Opin
Oncol 28(1):72–82. doi:10.1097/cco.0000000000000253
91. Gupta S, Li L (2015) Modulation of miRNAs in pul-
monary hypertension. Int J Hypertens 2015:169069.
doi:10.1155/2015/169069
92. Weiler J, Hunziker J, Hall J (2006) Anti-miRNA oligonu-
cleotides (AMOs): ammunition to target miRNAs implicated
in human disease?. Gene Ther 13(6):496–502. doi:10.1038/
sj.gt.3302654
93. Gubrij IB, Pangle AK, Pang L, Johnson LG (2016) Reversal
of microrna dysregulation in an animal model of pulmonary
hypertension. PLoS ONE 11(1):e0147827. doi:10.1371/jour-
nal.pone.0147827
94. Courboulin A, Paulin R, Giguère NJ, Saksouk N, Perreault
T, Meloche J, Paquet ER, Biardel S, Provencher S, Côté J,
Simard MJ, Bonnet S (2011) Role for miR-204 in human pul-
monary arterial hypertension. J Exp Med 208(3):535–548.
doi:10.1084/jem.20101812
95. Bienertova-Vasku J, Novak J, Vasku A (2015) MicroRNAs in
pulmonary arterial hypertension: pathogenesis, diagnosis and
treatment. J Am Soc Hypertens 9(3):221–234. doi:10.1016/j.
jash.2014.12.011
96. Fujita Y, Takeshita F, Kuwano K, Ochiya T (2013) RNAi
therapeutic platforms for lung diseases. Pharmaceuticals
6(2):223–250. doi:10.3390/ph6020223
97. Comer BS, Ba M, Singer CA, Gerthoffer WT (2015) Epige-
netic targets for novel therapies of lung diseases. Pharmacol
Ther 147:91–110. doi:10.1016/j.pharmthera.2014.11.006
98. Zhang Y, Wang Z, Gemeinhart RA (2013) Progress in
microRNA delivery. J Control Release 172(3):962–974.
doi:10.1016/j.jconrel.2013.09.015
99. Bobbin ML, Rossi JJ (2016) RNA interference (RNAi)-
based therapeutics: delivering on the promise? Annu
Rev Pharmacol Toxicol 56:103–122. doi:10.1146/
annurev-pharmtox-010715-103633
100. González CI, Bhattacharya A, Wang W, Peltz SW (2001)
Nonsense-mediated mRNA decay in Saccharomyces cerevi-
siae. Gene 274(1–2):15–25
101. Maquat LE (2004) Nonsense-mediated mRNA decay: splic-
ing, translation and mRNP dynamics. Nat Rev Mol Cell Biol
5(2):89–99. doi:10.1038/nrm1310
102. Noensie EN, Dietz HC (2001) A strategy for disease gene iden-
tification through nonsense-mediated mRNA decay inhibition.
Nat Biotechnol 19(5):434–439. doi:10.1038/88099
103. Howard M, Frizzell RA, Bedwell DM (1996) Aminoglycoside
antibiotics restore CFTR function by overcoming premature
stop mutations. Nat Med 2(4):467–469
104. Barton-Davis ER, Cordier L, Shoturma DI, Leland SE,
Sweeney HL (1999) Aminoglycoside antibiotics restore dys-
trophin function to skeletal muscles of mdx mice. J Clin Invest
104(4):375–381. doi:10.1172/jci7866
105. Diop D, Chauvin C, Jean-Jean O (2007) Aminoglycosides and
other factors promoting stop codon readthrough in human cells.
C R Biol 330(1):71–79. doi:10.1016/j.crvi.2006.09.001
106. Kuzmiak HA, Maquat LE (2006) Applying nonsense-medi-
ated mRNA decay research to the clinic: progress and chal-
lenges. Trends Mol Med 12(7):306–316. doi:10.1016/j.
molmed.2006.05.005
107. Hermann T (2007) Aminoglycoside antibiotics: old drugs and
new therapeutic approaches. Cell Mol Life Sci 64(14):1841–
1852. doi:10.1007/s00018-007-7034-x
108. Zingman LV, Park S, Olson TM, Alekseev AE, Terzic A (2007)
Aminoglycoside-induced translational read-through in dis-
ease: overcoming nonsense mutations by pharmacogenetic
therapy. Clin Pharmacol Ther 81(1):99–103. doi:10.1038/
sj.clpt.6100012
109. Kellermayer R (2006) Translational readthrough induction of
pathogenic nonsense mutations. Eur J Med Genet 49(6):445–
450. doi:10.1016/j.ejmg.2006.04.003
110. Kulyté A, Dryselius R, Karlsson J, Good L (2005) Gene selec-
tive suppression of nonsense termination using antisense
agents. Biochim Biophys Acta 1730(3):165–172. doi:10.1016/j.
bbaexp.2005.07.004
111. Linde L, Kerem B (2008) Introducing sense into nonsense
in treatments of human genetic diseases. Trends Genet
24(11):552–563. doi:10.1016/j.tig.2008.08.010
112. Namy O, Hatin I, Rousset JP (2001) Impact of the six nucleo-
tides downstream of the stop codon on translation termination.
EMBO Rep 2(9):787–793. doi:10.1093/embo-reports/kve176
113. Nasim MT, Ghouri A, Patel B, James V, Rudarakanchana N,
Morrell NW, Trembath RC (2008) Stoichiometric imbalance
in the receptor complex contributes to dysfunctional BMPR-II
mediated signalling in pulmonary arterial hypertension. Hum
Mol Genet 17(11):1683–1694. doi:10.1093/hmg/ddn059
114. Hamid R, Hedges LK, Austin E, Phillips JA, Loyd JE,
Cogan JD (2010) Transcripts from a novel BMPR2 ter-
mination mutation escape nonsense mediated decay by
downstream translation re-initiation: implications for treat-
ing pulmonary hypertension. Clin Genet 77(3):280–286.
doi:10.1111/j.1399-0004.2009.01311.x
115. Ryan NJ (2014) Ataluren: first global approval. Drugs
74(14):1709–1714. doi:10.1007/s40265-014-0287-4
116. Rudarakanchana N, Flanagan JA, Chen H, Upton PD, Machado
R, Patel D, Trembath RC, Morrell NW (2002) Functional analy-
sis of bone morphogenetic protein type II receptor mutations
underlying primary pulmonary hypertension. Hum Mol Genet
11(13):1517–1525
117. Rubenstein RC, Egan ME, Zeitlin PL (1997) Invitro pharma-
cologic restoration of CFTR-mediated chloride transport with
sodium 4-phenylbutyrate in cystic fibrosis epithelial cells con-
taining delta F508-CFTR. J Clin Invest 100(10):2457–2465.
doi:10.1172/jci119788
118. Rubenstein RC, Zeitlin PL (1998) A pilot clinical trial of oral
sodium 4-phenylbutyrate (Buphenyl) in deltaF508-homozygous
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2994 M.Orriols et al.
1 3
cystic fibrosis patients: partial restoration of nasal epithelial
CFTR function. Am J Respir Crit Care Med 157(2):484–490.
doi:10.1164/ajrccm.157.2.9706088
119. Rubenstein RC, Zeitlin PL (2000) Sodium 4-phenylbu-
tyrate downregulates Hsc70: implications for intracellular
trafficking of DeltaF508-CFTR. Am J Physiol Cell Physiol
278(2):C259–C267
120. Rubenstein RC, Lyons BM (2001) Sodium 4-phenylbutyrate
downregulates HSC70 expression by facilitating mRNA degra-
dation. Am J Physiol Lung Cell Mol Physiol 281(1):L43–L51
121. Zhang X-M, Wang X-T, Yue H, Leung SW, Thibodeau PH,
Thomas PJ, Guggino SE (2003) Organic solutes rescue the
functional defect in delta F508 cystic fibrosis transmembrane
conductance regulator. J Biol Chem 278(51):51232–51242.
doi:10.1074/jbc.M309076200
122. Papp E, Csermely P (2006) Chemical chaperones: mechanisms
of action and potential use. Handb Exp Pharmacol 172:405–416
123. Ma L, Liu Y, El-Achkar TM, Wu X-R (2012) Molecular and
cellular effects of Tamm-Horsfall protein mutations and their
rescue by chemical chaperones. J Biol Chem 287(2):1290–
1305. doi:10.1074/jbc.M111.283036
124. Hua Y, Kandadi MR, Zhu M, Ren J, Sreejayan N (2010) Tauro-
ursodeoxycholic acid attenuates lipid accumulation in endoplas-
mic reticulum-stressed macrophages. J Cardiovasc Pharmacol
55(1):49–55. doi:10.1097/FJC.0b013e3181c37d86
125. da-Silva WS, Ribich S, Arrojo e Drigo R, Castillo M, Patti
M-E, Bianco AC (2011) The chemical chaperones tauroursode-
oxycholic and 4-phenylbutyric acid accelerate thyroid hormone
activation and energy expenditure. FEBS Lett 585(3):539–544.
doi:10.1016/j.febslet.2010.12.044
126. Cao SS, Zimmermann EM, Chuang B-M, Song B, Nwokoye
A, Wilkinson JE, Eaton KA, Kaufman RJ (2013) The unfolded
protein response and chemical chaperones reduce protein mis-
folding and colitis in mice. Gastroenterology 144(5):989–1000.
e1006. doi:10.1053/j.gastro.2013.01.023
127. Sobolewski A, Rudarakanchana N, Upton PD, Yang J, Crilley
TK, Trembath RC, Morrell NW (2008) Failure of bone morpho-
genetic protein receptor trafficking in pulmonary arterial hyper-
tension: potential for rescue. Hum Mol Genet 17(20):3180–
3190. doi:10.1093/hmg/ddn214
128. Yang Y-M, Lane KB, Sehgal PB (2013) Subcellular mecha-
nisms in pulmonary arterial hypertension: combinatorial
modalities that inhibit anterograde trafficking and cause bone
morphogenetic protein receptor type 2 mislocalization. Pulm
Circ 3(3):533–550. doi:10.1086/674336
129. Frump AL, Lowery JW, Hamid R, Austin ED, de Caestecker
M (2013) Abnormal trafficking of endogenously expressed
BMPR2 mutant allelic products in patients with heritable
pulmonary arterial hypertension. PLoS ONE 8(11):e80319.
doi:10.1371/journal.pone.0080319
130. Zeitlin PL, Diener-West M, Rubenstein RC, Boyle MP, Lee
CKK, Brass-Ernst L (2002) Evidence of CFTR function in
cystic fibrosis after systemic administration of 4-phenylbu-
tyrate. Mol Ther 6(1):119–126. doi:10.1006/mthe.2002.0639
131. Obici L, Cortese A, Lozza A, Lucchetti J, Gobbi M, Palladini
G, Perlini S, Saraiva MJ, Merlini G (2012) Doxycycline plus
tauroursodeoxycholic acid for transthyretin amyloidosis: a
phase II study. Amyloid 19(Suppl 1):34–36. doi:10.3109/1350
6129.2012.678508
132. Berry GT, Steiner RD (2001) Long-term management
of patients with urea cycle disorders. J Pediatr 138(1
Suppl):S56–60; discussion S60–51
133. Iannitti T, Palmieri B (2011) Clinical and experimental appli-
cations of sodium phenylbutyrate. Drugs R D 11(3):227–249.
doi:10.2165/11591280-000000000-00000
134. Durrington HJ, Upton PD, Hoer S, Boname J, Dunmore BJ,
Yang J, Crilley TK, Butler LM, Blackbourn DJ, Nash GB,
Lehner PJ, Morrell NW (2010) Identification of a lysosomal
pathway regulating degradation of the bone morphogenetic
protein receptor type II. J Biol Chem 285(48):37641–37649.
doi:10.1074/jbc.M110.132415
135. Satow R, Kurisaki A, Chan T-c, Hamazaki TS, Asashima M
(2006) Dullard promotes degradation and dephosphorylation of
BMP receptors and is required for neural induction. Dev Cell
11(6):763–774. doi:10.1016/j.devcel.2006.10.001
136. Luzio JP, Pryor PR, Bright NA (2007) Lysosomes: fusion and
function. Nat Rev Mol Cell Biol 8(8):622–632. doi:10.1038/
nrm2217
137. Wertz JW, Bauer PM (2008) Caveolin-1 regulates BMPRII
localization and signaling in vascular smooth muscle cells. Bio-
chem Biophys Res Commun 375(4):557–561. doi:10.1016/j.
bbrc.2008.08.066
138. Hartung A, Bitton-Worms K, Rechtman MM, Wenzel V,
Boergermann JH, Hassel S, Henis YI, Knaus P (2006) Different
routes of bone morphogenic protein (BMP) receptor endocyto-
sis influence BMP signaling. Mol Cell Biol 26(20):7791–7805.
doi:10.1128/mcb.00022-06
139. Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana JL (2003)
Distinct endocytic pathways regulate TGF-beta receptor signal-
ling and turnover. Nat Cell Biol 5(5):410–421. doi:10.1038/
ncb975
140. Zaidi SH, You XM, Ciura S, O’Blenes S, Husain M, Rabino-
vitch M (2000) Suppressed smooth muscle proliferation and
inflammatory cell invasion after arterial injury in elafin-overex-
pressing mice. J Clin Invest 105(12):1687–1695. doi:10.1172/
jci9147
141. Zaidi SHE, You X-M, Ciura S, Husain M, Rabinovitch M
(2002) Overexpression of the serine elastase inhibitor elafin
protects transgenic mice from hypoxic pulmonary hypertension.
Circulation 105(4):516–521
142. Cowan KN, Heilbut A, Humpl T, Lam C, Ito S, Rabinovitch
M (2000) Complete reversal of fatal pulmonary hypertension
in rats by a serine elastase inhibitor. Nat Med 6(6):698–702.
doi:10.1038/76282
143. Alam SR, Lewis SC, Zamvar V, Pessotto R, Dweck MR, Kris-
han A, Goodman K, Oatey K, Harkess R, Milne L, Thomas
S, Mills NM, Moore C, Semple S, Wiedow O, Stirrat C, Mir-
sadraee S, Newby DE, Henriksen PA (2015) Perioperative
elafin for ischaemia-reperfusion injury during coronary artery
bypass graft surgery: a randomised-controlled trial. Heart
101(20):1639–1645. doi:10.1136/heartjnl-2015-307745
144. Hubbard VM, Valdor R, Macian F, Cuervo AM (2012) Selec-
tive autophagy in the maintenance of cellular homeostasis in
aging organisms. Biogerontology 13(1):21–35. doi:10.1007/
s10522-011-9331-x
145. Kroemer G, Marino G, Levine B (2010) Autophagy and
the integrated stress response. Mol Cell 40(2):280–293.
doi:10.1016/j.molcel.2010.09.023
146. Long L, Yang X, Southwood M, Lu J, Marciniak SJ, Dun-
more BJ, Morrell NW (2013) Chloroquine prevents progres-
sion of experimental pulmonary hypertension via inhibition of
autophagy and lysosomal bone morphogenetic protein type II
receptor degradation. Circ Res 112(8):1159–1170. doi:10.1161/
CIRCRESAHA.111.300483
147. Daniel JK, Fabio CA, Hagai A etal (2012) Guidelines for the
use and interpretation of assays for monitoring autophagy.
Autophagy 8:445–544. doi:10.1080/15548627.2015.1100356
148. Ben-Zvi I, Kivity S, Langevitz P, Shoenfeld Y (2012) Hydroxy-
chloroquine: from malaria to autoimmunity. Clin Rev Allergy
Immunol 42(2):145–153. doi:10.1007/s12016-010-8243-x
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2995BMP type II receptor asatherapeutic target inpulmonary arterial hypertension
1 3
149. Rabinovitch M, Guignabert C, Humbert M, Nicolls MR (2014)
Inflammation and immunity in the pathogenesis of pulmonary
arterial hypertension. Circ Res 115(1):165–175. doi:10.1161/
CIRCRESAHA.113.301141
150. Ormiston ML, Upton PD, Li W, Morrell NW (2015) The prom-
ise of recombinant BMP ligands and other approaches targeting
BMPR-II in the treatment of pulmonary arterial hypertension.
Glob Cardiol Sci Pract 2015(4):47. doi:10.5339/gcsp.2015.47
151. Cheng H, Jiang W, Phillips FM, Haydon RC, Peng Y, Zhou L,
Luu HH, An N, Breyer B, Vanichakarn P, Szatkowski JP, Park
JY, He TC (2003) Osteogenic activity of the fourteen types of
human bone morphogenetic proteins (BMPs). J Bone Joint Surg
Am 85:1544–1552
152. Croom KF, Curran MP (2008) Sildenafil: a review of its use in
pulmonary arterial hypertension. Drugs 68(3):383–397
153. Ito T, Ozawa K, Shimada K (2007) Current drug targets and
future therapy of pulmonary arterial hypertension. Curr Med
Chem 14(6):719–733
154. Zhao L, Sebkhi A, Ali O, Wojciak-Stothard B, Mamanova L,
Yang Q, Wharton J, Wilkins MR (2009) Simvastatin and silde-
nafil combine to attenuate pulmonary hypertension. Eur Respir
J 34(4):948–957. doi:10.1183/09031936.00143508
155. Barnett CF, Machado RF (2006) Sildenafil in the treat-
ment of pulmonary hypertension. Vasc Health Risk Manag
2(4):411–422
156. Schwappacher R, Weiske J, Heining E, Ezerski V, Marom B,
Henis YI, Huber O, Knaus P (2009) Novel crosstalk to BMP
signalling: cGMP-dependent kinase I modulates BMP receptor
and Smad activity. EMBO J 28(11):1537–1550. doi:10.1038/
emboj.2009.103
157. Long L, Crosby A, Yang X, Southwood M, Upton PD, Kim
D-K, Morrell NW (2009) Altered bone morphogenetic pro-
tein and transforming growth factor-beta signaling in rat
models of pulmonary hypertension: potential for activin
receptor-like kinase-5 inhibition in prevention and progres-
sion of disease. Circulation 119(4):566–576. doi:10.1161/
circulationaha.108.821504
158. Yang J, Li X, Al-Lamki RS, Wu C, Weiss A, Berk J, Scher-
muly RT, Morrell NW (2013) Sildenafil potentiates bone mor-
phogenetic protein signaling in pulmonary arterial smooth
muscle cells and in experimental pulmonary hypertension.
Arterioscler Thromb Vasc Biol 33(1):34–42. doi:10.1161/
atvbaha.112.300121
159. Wang R-C, Jiang F-M, Zheng Q-l, Li C-T, Peng X-Y, He C-Y,
Luo J, Liang Z-A (2014) Efficacy and safety of sildenafil treat-
ment in pulmonary arterial hypertension: a systematic review.
Respir Med 108(3):531–537. doi:10.1016/j.rmed.2014.01.003
160. Spiekerkoetter E, Tian X, Cai J, Hopper RK, Sudheendra D, Li
CG, El-Bizri N, Sawada H, Haghighat R, Chan R, Haghighat
L, de Jesus Perez V, Wang L, Reddy S, Zhao M, Bernstein
D, Solow-Cordero DE, Beachy PA, Wandless TJ, Ten Dijke
P, Rabinovitch M (2013) FK506 activates BMPR2, rescues
endothelial dysfunction, and reverses pulmonary hypertension.
J Clin Invest 123(8):3600–3613. doi:10.1172/jci65592
161. Wu X, Wang L, Han Y, Regan N, Li P-K, Villalona MA, Hu
X, Briesewitz R, Pei D (2011) Creating diverse target-binding
surfaces on FKBP12: synthesis and evaluation of a rapamycin
analogue library. ACS Comb Sci 13(5):486–495. doi:10.1021/
co200057n
162. Albiñana V, Sanz-Rodríguez F, Recio-Poveda L, Bernabéu C,
Botella LM (2011) Immunosuppressor FK506 increases endog-
lin and activin receptor-like kinase 1 expression and modulates
transforming growth factor-β1 signaling in endothelial cells.
Mol Pharmacol 79(5):833–843. doi:10.1124/mol.110.067447
163. Spiekerkoetter E, Sung YK, Sudheendra D, Bill M, Aldred MA,
van de Veerdonk MC, Vonk Noordegraaf A, Long-Boyle J,
Dash R, Yang PC, Lawrie A, Swift AJ, Rabinovitch M, Zama-
nian RT (2015) Low-dose FK506 (Tacrolimus) in end-stage
pulmonary arterial hypertension. Am J Respir Crit Care Med
192(2):254–257. doi:10.1164/rccm.201411-2061LE
164. Plosker GL, Foster RH (2000) Tacrolimus: a further update of
its pharmacology and therapeutic use in the management of
organ transplantation. Drugs 59(2):323–389
165. Takeda Y, Miyamori I, Furukawa K, Inaba S, Mabuchi H
(1999) Mechanisms of FK 506-induced hypertension in the rat.
Hypertension 33(1):130–136
166. Drake KM, Dunmore BJ, McNelly LN, Morrell NW, Aldred
MA (2013) Correction of nonsense BMPR2 and SMAD9
mutations by ataluren in pulmonary arterial hypertension.
Am J Respir Cell Mol Biol 49(3):403–409. doi:10.1165/
rcmb.2013-0100OC
167. Bloom RD, Reese PP (2007) Chronic kidney disease after
nonrenal solid-organ transplantation. J Am Soc Nephrol
18(12):3031–3041. doi:10.1681/asn.2007040394
168. Randhawa PS, Starzl TE, Demetris AJ (1997) Tacroli-
mus (FK506)-associated renal pathology. Adv Anat Pathol
4(4):256–276
169. Endo-Takahashi Y, Negishi Y, Nakamura A, Ukai S, Ooaku K,
Oda Y, Sugimoto K, Moriyasu F, Takagi N, Suzuki R, Maruy-
ama K, Aramaki Y (2014) Systemic delivery of miR-126 by
miRNA-loaded bubble liposomes for the treatment of hindlimb
ischemia. Sci Rep 4:3883. doi:10.1038/srep03883
170. D’Astolfo DS, Pagliero RJ, Pras A, Karthaus WR, Clevers H,
Prasad V, Lebbink RJ, Rehmann H, Geijsen N (2015) Efficient
intracellular delivery of native proteins. Cell 161(3):674–690.
doi:10.1016/j.cell.2015.03.028
171. Esch EW, Bahinski A, Huh D (2015) Organs-on-chips at the
frontiers of drug discovery. Nat Rev Drug Discov 14(4):248–
260. doi:10.1038/nrd4539
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Bone morphogenetic protein receptor type 2 (BMPR2) is a gene involved in cell development and division [202,203]. The loss of BMPR2 activity has been linked to increased toxicity in PAH [204,205], and aberrant hypermethylation of the BMPR2 promoter region has been reported in familial PAH patients [206], suggesting that DNA methylation may suppress BMPR2 gene expression. ...
Article
Full-text available
Vascular smooth muscle cells (VSMCs) are crucial components of the arterial wall, controlling blood flow and pressure by contracting and relaxing the artery walls. VSMCs can switch from a contractile to a synthetic state, leading to increased proliferation and migratory potential. Epigenetic pathways, including DNA methylation, play a crucial role in regulating VSMC differentiation and phenotypic flexibility. DNA methylation involves attaching a methyl group to the 5’ carbon of a cytosine base, which regulates gene expression by interacting with transcription factors. Understanding the key factors influencing VSMC plasticity may help to identify new target molecules for the development of innovative drugs to treat various vascular diseases. This review focuses on DNA methylation pathways in VSMCs, summarizing mechanisms involved in controlling vascular remodeling, which can significantly enhance our understanding of related mechanisms and provide promising therapeutic approaches for complex and multifactorial diseases.
... Next, bone defect and ovariectomy-induced postmenopausal osteoporosis are attractive models that could be used to test the in vivo efficacy of the indolyl-benzimidazoles [55,75,76]. Moreover, inducible models of PAH or chronic kidney disease are also appealing avenues for pre-clinical investigation of the in vivo efficacy of these small molecules [2,77,78]. With the advantages associated with small molecules, the SY-LB compounds could potentially be cost-effective replacements for rBMP-based therapies targeting a number of organ systems. ...
Article
Background: Increasing or restoring Bone Morphogenetic Protein- (BMP-) signaling through administration of recombinant BMPs (rBMPs) has demonstrated therapeutic efficacy for treating bone fractures or to enhance repair following spinal surgeries. However, direct use of rBMPs has come up against significant obstacles like high cost and incidence of adverse effects. Recently, we reported our findings on the novel indolyl-benzimidazoles, SY-LB-35 and SY-LB-57, that fully activated BMP receptor signaling demonstrating activity profiles that mirrored rBMPs. Here, we explored the potential of these compounds to substitute for rBMPs in processes like wound healing and osteogenesis. Methods: Cell-based assays including cell viability, short- and long-term phosphorylation, protein expression, wound healing and bone differentiation assays were carried out in the pluripotent myoblast C2C12 cell line with select assays performed in multiple cell lines. Several assays included conditions in the presence of a selective inhibitor of type I BMP receptor, Activin-like kinase 2 (ALK2), or inhibitors of BMP-stimulated downstream signaling. All assays were repeated at least 3 times with replicates per condition where indicated. Statistical tests were carried out using Student's two-tailed, t-test. Results: Sustained activation of non-canonical BMP signaling pathways was observed after 24-hour exposure to SY-LB-35 and SY-LB-57. Moreover, this treatment increased the expression of targets of BMP-mediated transcription such as the Id1 transcription factor. SY-LB-35 and SY-LB-57 promoted substantial increases in cell viability in three distinct cell types and increased the rate of wound closure in scrape-wounded C2C12 cell cultures. Cell viability and wound closure induced by SY-LB compounds required ALK2-, PI3K- and p38-dependent pathways. In contrast, responses to SY-LB compounds were not affected by ERK inhibition. Expression of bone differentiation markers beginning at 4 hours and evidence of calcium deposition detected after 21 days in C2C12 cell cultures exposed to SY-LB-35 and SY-LB-57 demonstrated the osteogenic potential of these compounds. Conclusions: The functional similarities between these novel compounds and rBMPs indicates that SY-LB-35 or SY-LB-57, acting as potent activators of BMP receptor signaling and inducers of osteogenic processes, could potentially replace rBMPs for treating BMP-related pathologies such as bone fracture repair or other wound healing processes.
... 8 Upon ligand binding, a heterodimer complex, formed by a combination of two type I receptors and two type II receptors, propagates the signal through a canonical pathway by phosphorylation of the Smad1/5/8 transcription factors or a noncanonical pathway by phosphorylation of p38 MAPK. 9 To date, about 500 distinct bmpr2 variants have been identified and most of them are pathogenic. 10 In addition, mutations in other genes including the activin receptor-like kinase 1 (alk1), endoglin (eng), 11 small mothers against decapentaplegic homolog 1 (smad1), smad4 12 and smad9 13 have been found being associated to PAH, highlighting the crucial role of the BMP pathway in the pathogenesis of PAH. 14 Among the multifactorial processes associated with PAH, endothelial dysfunction and defective angiogenesis play a key role in initiating structural changes in the pulmonary vasculature. ...
Article
Full-text available
Mutations in the bone morphogenetic protein receptor type 2 ( bmpr2 ) gene and signaling pathway impairment are observed in heritable and idiopathic pulmonary arterial hypertension (PAH). In PAH, endothelial dysfunction is currently handled by drugs targeting the endothelin‐1 (ET‐1), nitric oxide (NO), and prostacyclin (PGI 2 ) pathways. The role of angiogenesis in the disease process and the effect of PAH therapies on dysregulated angiogenesis remain inconclusive. We aim to investigate in vitro whether (i) bmpr2 silencing can impair angiogenic capacity of human lung microvascular endothelial cells (HLMVECs) and (ii) PAH therapies can restore them. The effects of macitentan (ET‐1), tadalafil (NO), and selexipag (PGI 2 ), on BMPRII pathway activation, endothelial barrier function, and angiogenesis were investigated in bmpr2 ‐silenced HLMVECs. Stable bmpr2 silencing resulted in impaired migration and tube formation in vitro capacity. Inhibition of ET‐1 pathway was able to partially restore tube formation in bmpr2 ‐silenced HLMVECs, whereas none of the therapies was able to restore endothelial barrier function, no deleterious effects were observed. Our findings highlight the potential role of BMPRII signaling pathway in driving pulmonary endothelial cell angiogenesis. In addition, PAH drugs display limited effects on endothelial function when BMPRII is impaired, suggesting that innovative therapeutic strategies targeting BMPRII signaling are needed to better rescue endothelial dysfunction in PAH.
... The BMP10 mutations were recently identified in young patients with severe PAH 27,28 , and mutations in GDF2, a gene encoding BMP9, resulting in BMP9 loss of function, result in reduced circulating levels of both, BMP9 and BMP10 29 . Mutational or epigenetic down-regulation at the receptor (predominantly BMPR2) level is strongly associated with familial and idiopathic PAH 30 . Importantly, BMPR2 down-regulation contributes to hyper-proliferative, apoptosis resistant phenotype in PAH pulmonary vascular cells [31][32][33][34] , and BMP9 is deficient in portopulmonary hypertension and is a biomarker for transplant-free survival 35 . ...
Article
Full-text available
Pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by a progressive increase in pulmonary vascular resistance leading to right ventricular failure and often death. Here we report that deficiency of transcription factor GATA6 is a shared pathological feature of PA endothelial (PAEC) and smooth muscle cells (PASMC) in human PAH and experimental PH, which is responsible for maintenance of hyper-proliferative cellular phenotypes, pulmonary vascular remodeling and pulmonary hypertension. We further show that GATA6 acts as a transcription factor and direct positive regulator of anti-oxidant enzymes, and its deficiency in PAH/PH pulmonary vascular cells induces oxidative stress and mitochondrial dysfunction. We demonstrate that GATA6 is regulated by the BMP10/BMP receptors axis and its loss in PAECs and PASMC in PAH supports BMPR deficiency. In addition, we have established that GATA6-deficient PAEC, acting in a paracrine manner, increase proliferation and induce other pathological changes in PASMC, supporting the importance of GATA6 in pulmonary vascular cell communication. Treatment with dimethyl fumarate resolved oxidative stress and BMPR deficiency, reversed hemodynamic changes caused by endothelial Gata6 loss in mice, and inhibited proliferation and induced apoptosis in human PAH PASMC, strongly suggesting that targeting GATA6 deficiency may provide a therapeutic advance for patients with PAH.
... BMPs are part of the transforming growth factor (TGF)-β family; unopposed TGF-β signals happen when we have a loss of function of the BMPR2 genes, which is essential in regulating vascular cell apoptosis and maintaining normal function of the endothelium. When BMPR2 is suppressed, there is unopposed signaling of TGF-β, resulting in proliferation of the vascular cells and remodeling [29,30]. Sotatercept is a novel drug that works on the BMPR2 gene and the TGF-β pathway. ...
Article
Pulmonary arterial hypertension (PAH) is a progressive disease that despite advances in therapy is associated with a 7-year survival of approximately 50%. Several risk factors are associated with developing PAH, include methamphetamine use, scleroderma, human immunodeficiency virus, portal hypertension, and genetic predisposition. PAH can also be idiopathic. There are traditional pathways underlying the pathophysiology of PAH involving nitric oxide, prostacyclin, thromboxane A2, and endothelin-1, resulting in impaired vasodilation, enhanced vasoconstriction and proliferation in the pulmonary vasculature. Established PAH medications targets these pathways; however, this paper aims to discuss novel drugs for treating PAH by targeting new and alternative pathways.
Article
Background: The purpose of this study was to evaluate the effect of 18β-glycyrrhetinic acid (18β-GA) on isolated rat pulmonary artery vascular rings as well as the mechanism that lies behind the diastolic effects of this compound. Methods: To examine the effects of various doses of 18β-GA on resting normal vascular rings, we used isolated rat pulmonary artery vascular rings. The isolated rat pulmonary artery vascular ring was precontracted with phenylephrine (PE) and potassium chloride solution (KCl), which allowed for the observation of the diastolic effects of 18β-GA, as well as the effects of various concentrations of 18β-GA on blocking the four potassium channels of glibenclamide, barium chloride (BaCl2), tetraethylamine (TEA), or 4-aminopyridine; the impact of various 18β-GA concentrations on the pulmonary artery vasodilation effect pre-use of the nitric oxide synthase inhibitors l-nitroarginine methyl ester and indomethacin. Results: It was shown that 18β-GA has concentration-dependent diastolic effects on isolated rat pulmonary vascular rings that have been precontracted with PE and KCl but it has no effect on resting isolated thoracic aortic vascular rings. Conclusion: The two Kir and Kv channels may be connected to the endothelium-dependent vasodilation mechanism of 18β-GA.
Article
Full-text available
Hypertension is a major harbinger of cardiovascular morbidity and mortality. It predisposes to higher rates of myocardial infarction, chronic kidney failure, stroke, and heart failure than most other risk factors. By 2025, the prevalence of hypertension is projected to reach 1.5 billion people. The pathophysiology of this disease is multifaceted, as it involves nitric oxide and endothelin dysregulation, reactive oxygen species, vascular smooth muscle proliferation, and vessel wall calcification, among others. With the advent of new biomolecular techniques, various studies have elucidated a gaping hole in the etiology and mechanisms of hypertension. Indeed, epigenetics, DNA methylation, histone modification, and microRNA-mediated translational silencing appear to play crucial roles in altering the molecular phenotype into a hypertensive profile. Here, we critically review the experimentally determined associations between microRNA (miRNA) molecules and hypertension pharmacotherapy. Particular attention is given to the epigenetic mechanisms underlying the physiological responses to antihypertensive drugs like candesartan, and other relevant drugs like clopidogrel, aspirin, and statins among others. Furthermore, how miRNA affects the pharmaco-epigenetics of hypertension is especially highlighted.
Article
Full-text available
Cardiovascular disease is the leading cause of death worldwide. Its prevalence is rising due to ageing populations and the increasing incidence of diseases such as chronic kidney disease, obesity and diabetes which are associated with elevated cardiovascular risk. Despite currently available treatments, there remains a huge burden of cardiovascular disease-associated morbidity for patients and healthcare systems, and newer treatments are needed. The apelin system, comprising the apelin receptor and its two endogenous ligands apelin and elabela, is a broad regulator of physiology that opposes the actions of the renin-angiotensin and vasopressin systems. Activation of the apelin receptor promotes endothelium-dependent vasodilatation and inotropy, lowers blood pressure and promotes angiogenesis. The apelin system appears to protect against arrhythmias, inhibits thrombosis and has broad anti-inflammatory and anti-fibrotic actions. It also promotes aqueous diuresis through direct and indirect (central) effects in the kidney. Thus, the apelin system offers therapeutic promise for a range of cardiovascular, kidney and metabolic diseases. This review will discuss current cardiovascular disease targets of the apelin system and future clinical utility of apelin receptor agonism.
Article
Full-text available
No studies have explored a functional role for bone morphogenetic protein (BMP)-9, a transforming growth factor-β superfamily ligand, in cardiac remodeling after myocardial infarction (MI). Using BMP-9 null mice, we observed that loss of BMP-9 decreases survival and increases cardiac rupture after MI. We further observed that loss of BMP-9 not only increases collagen abundance, but also promotes matrix metalloproteinase-9 activity and collagen degradation after MI. These findings identify BMP-9 as a necessary component of cardiac remodeling after MI and a potentially important target of therapy to improve outcomes after MI.
Article
Pulmonary arterial hypertension (PAH) is a devastating rare disease, which despite currently available treatments, still represents a high unmet medical need. Specific E3 ubiquitin protein ligase 1 (SMURF1) is a HECT E3 ligase that ubiquitinates key signaling molecules from the TGFβ/BMP pathways, which are of great relevance in the pathophysiology of PAH. Herein, the design and synthesis of novel potent small-molecule SMURF1 ligase inhibitors are described. Lead molecule 38 has demonstrated good oral pharmacokinetics in rats and significant efficacy in a rodent model of pulmonary hypertension.
Article
Full-text available
Background: Pulmonary arterial hypertension (PAH) is a rare vascular disorder characterized by a capillary wedge pressure ≤ 15 mmHg and a mean pulmonary arterial pressure ≥ 25 mmHg at rest. PAH can be idiopathic, heritable or associated with other conditions. The aim of this study was to analyze the Endoglin (ENG) gene and assess the influence of the c.572G > A (p.G191D) mutation in patients with idiopathic or associated PAH. The correlation between the pathogenic mutations and clinical and functional parameters was further analyzed. Results: Sixteen different changes in the ENG gene were found in 44 out of 57 patients. After in silico analysis, we classified eight mutations as pathogenic in 16 of patients. The c.572G>A (p.G191D) variation was observed in ten patients, and the analysis for the splicing process using hybrid minigenes, with pSPL3 vector to assess splicing alterations, do not generate a new transcript. Age at diagnosis (p = 0.049) and the 6-min walking test (p = 0.041) exhibited statistically significant differences between carriers and non-carriers of pathogenic mutations. Patients with pathogenic mutations exhibited disease symptoms 8 years before non-carriers. Five patients with pathogenic mutations were carriers of another mutation in the BMPR2 or ACVRL1 genes. Conclusions: We present a series of PAH patients with mutations in the ENG gene, some of them not previously described, exhibiting clinical and hemodynamic alterations suggesting that the presence of these mutations may be associated with the severity of the disease. Moreover, genetic analysis in patients with PAH may be of clinical relevance and indicates the complexity of the genetic background.
Article
Introduction: Pulmonary arterial hypertension (PAH) is a cardiopulmonary disorder in which mechanical obstruction of the pulmonary vascular bed is largely responsible for the rise in pulmonary arterial pressures. The discovery of heterozygous BMPR2 germline mutations as critical predisposing factors together with a remarkable progress in our understanding of the pathogenic mechanisms have helped identify the significant and complex roles of the BMPRII axis in PAH. However, their precise contributions to the condition are still incompletely understood. Areas covered: This review aims to assemble and discuss the cellular actions of BMPs together with the possible clinical applications and prospects for their use in the near future. Expert opinion: Although major advances have been made, several questions remain unanswered regarding development of efficacious therapies targeting the BMPRII axis in PAH. Specifically, the reasons why BMPRII signaling is reduced in PAH and how the alterations influence or even drive the pathogenesis need to be understood. Because the BMPRII axis is ubiquitously expressed and exhibits a wide variety of functions in organ regeneration and homeostasis, a better understanding of the overall risk-benefit ratio of these strategies with long-term follow-up is needed. This knowledge will lay the foundation for discovery of innovative therapeutics for PAH.
Article
Idiopathic Pulmonary Fibrosis (IPF) is a lethal lung disease of unknown etiology. The development of pulmonary hypertension (PH) is considered the single most significant predictor of mortality in patients with chronic lung diseases. The processes that govern the progression and development of fibroproliferative and vascular lesions in IPF are not fully understood. Using human lung explant samples from patients with IPF with or without a diagnosis of PH as well as normal control tissue, we report reduced BMPR2 expression in patients with IPF or IPF+PH. These changes were consistent with dampened P-SMAD 1/5/8 and elevated P-SMAD 2/3 demonstrating reduced BMPR2 signaling and elevated TGF-β activity in IPF. In the bleomycin (BLM) model of lung fibrosis and PH, we also report decreased BMPR2 expression compared to control animals that correlated with vascular remodeling and PH. We show that genetic abrogation or pharmacological inhibition of interleukin-6 leads to diminished markers of fibrosis and PH consistent with elevated levels of BMPR2 and reduced levels of a collection of microRNAs (miRs) that are able to degrade BMPR2. We also demonstrate that isolated bone-marrow derived macrophages from BLM-exposed mice show reduced BMPR2 levels upon exposure with IL6 or the IL6+IL6R complex that are consistent with IHC showing reduced BMPR2 in CD206 expressing macrophages from lung sections from IPF and IPF+PH patients. In conclusion, our data suggest that depletion of BMPR2 mediated by a collection of miRs induced by IL-6 and subsequent STAT3 phosphorylation as a novel mechanism participating to fibroproliferative and vascular injuries in IPF.
Article
Background: -We previously reported high-throughput RNA sequencing analyses that identified heightened expression of the chromatin architectural factor High Mobility Group AT-hook 1 (HMGA1) in pulmonary arterial (PA) endothelial cells (ECs) from idiopathic PA hypertension (IPAH) patients compared to controls. Since HMGA1 promotes epithelial to mesenchymal transition in cancer, we hypothesized that increased HMGA1 could induce transition of PAECs to a smooth muscle (SM)-like mesenchymal phenotype (EndMT), explaining both dysregulation of PAEC function and possible cellular contribution to the occlusive remodeling that characterizes advanced IPAH. Methods and results: -We documented increased HMGA1 in PAECs cultured from IPAH vs. donor control lungs. Confocal microscopy of lung explants localized the increase in HMGA1 consistently to PA endothelium, and identified many cells double-positive for HMGA1 and smooth muscle 22 alpha (SM22α) in occlusive and plexogenic lesions. Since decreased expression and function of bone morphogenetic protein receptor (BMPR)2 is observed in PAH, we reduced BMPR2 by siRNA in control PAECs and documented an increase in HMGA1 protein. Consistent with transition of PAECs by HMGA1, we detected reduced PECAM-1 (CD31) and increased EndMT markers, αSMA, SM22α, calponin, phospho-vimentin and Slug. The transition was associated with spindle SM-like morphology, and the increase in αSMA was largely reversed by joint knockdown of BMPR2 and HMGA1 or Slug. Pulmonary ECs from mice with EC-specific loss of BMPR2 showed similar gene and protein changes. Conclusions: -Increased HMGA1 in PAECs resulting from dysfunctional BMPR2 signaling can transition endothelium to SM-like cells associated with PAH.
Article
The most common mutation of the cystic fibrosis transmembrane conductance regulator (CFTR), Delta F508, is a trafficking mutant that has prolonged associations with molecular chaperones and is rapidly degraded, at least in part by the ubiquitin-proteasome system. Sodium 4-phenylbutyrate (4PBA) improves Delta F508-CFTR trafficking and function in vitro in cystic fibrosis epithelial cells and in vivo. To further understand the mechanism of action of 4PBA, we tested the hypothesis that 4PBA modulates the targeting of Delta F508-CFTR for ubiquitination and degradation by reducing the expression of Hsc70 in cystic fibrosis epithelial cells. IB3-1 cells (genotype Delta F508/W1282X) that were treated with 0.05-5 mM 4PBA for 2 days in culture demonstrated a dose-dependent reduction in Hsc70 protein immunoreactivity and mRNA levels. Immunoprecipitation with Hsc70-specific antiserum demonstrated that Hsc70 and CFTR associated under control conditions and that treatment with 4PBA reduced these complexes. Levels of immunoreactive Hsp40, Hdj2, Hsp70, Hsp90, and calnexin were unaffected by 4PBA treatment. These data suggest that 4PBA may improve Delta F508-CFTR trafficking hy allowing a greater proportion of mutant CFTR to escape association with Hsc70.
Article
Retrovirus vectors (gammaretroviral and lentiviral vectors) have been considered as promising tools to transfer therapeutic genes into patient cells because they can permanently integrate into host cellular genome. To treat monogenic, inherited diseases, retroviral vectors have been used to add correct genes into patient cells. Conventional gammaretroviral vectors achieved successful results in clinical trials: treated patients had therapeutic gene expression in target cells and had improved symptoms of diseases. However, serious side-effects of leukemia occurred, caused by retroviral insertional mutagenesis (IM). These incidences stressed the importance of monitoring vector integration sites in patient cells as well as of re-consideration on safer vectors. More recently lentiviral vectors which can deliver genes into non-dividing cells started to be used in clinical trials including neurological disorders, showing their efficacy. Vector integration site analysis revealed that lentiviruses integrate less likely to near promoter regions of oncogenes than gammaretroviruses and no adverse events have been reported in lentiviral vector-mediated gene therapy clinical trials. Therefore lentiviral vectors have promises to be applied to a wide range of common diseases in near future. For example, T cells from cancer patients were transduced to express chimeric T cell receptors recognizing their tumour cells enhancing patients' anti-cancer immunity.
Article
Vascular remodelling is a multifactorial process which involves both adaptive and maladaptive changes of the vessel wall through amongst others, cell proliferation and migration, but also apoptosis and necrosis of the various cell types in the vessel wall. Vascular remodelling can be beneficial e.g. during neovascularization after ischemia, as well as pathological e.g. during atherosclerosis and aneurysm formation. In recent years, it has become clear that microRNAs are able to target many genes that are involved in vascular remodelling processes and either can promote or inhibit structural changes of the vessel wall. Since many different processes of vascular remodelling are regulated by similar mechanisms and factors, both positive and negative vascular remodelling can be affected by the same microRNAs.A large number of microRNAs has been linked to various aspects of vascular remodelling and indeed, several of these microRNAs regulate multiple vascular remodelling processes, including both the adaptive processes angiogenesis and arteriogenesis as well as maladaptive processes of atherosclerosis, restenosis and aneurysm formation. Here, we discuss the multifactorial role of microRNAs and microRNA clusters that were reported to play a role in multiple forms of vascular remodelling and are clearly linked to cardiovascular disease. The microRNAs reviewed are miR-126, miR-155 and the microRNA gene clusters 17-92, 23/24/27, 143/145 and 14q32. Understanding the contribution of these microRNAs to the entire spectrum of vascular remodelling processes is important, especially as these microRNAs may have great potential as therapeutic targets for treatment of various cardiovascular diseases.
Article
Introduction: Pulmonary arterial hypertension (PAH) is a rare disorder associated with abnormally elevated pulmonary pressures that, if untreated, leads to right heart failure and premature death. The goal of drug development for PAH is to develop effective therapies that halt, or ideally, reverse the obliterative vasculopathy that results in vessel loss and obstruction of blood flow to the lungs. Areas Covered: This review summarizes the current approach to candidate discovery in PAH and discusses the currently available drug discovery methods that should be implemented to prioritize targets and obtain a comprehensive pharmacological profile of promising compounds with well-defined mechanisms. Expert opinion: To improve the successful identification of leading drug candidates, it is necessary that traditional pre-clinical studies are combined with drug screening strategies that maximize the characterization of biological activity and identify relevant off-target effects that could hinder the clinical efficacy of the compound when tested in human subjects. A successful drug discovery strategy in PAH will require collaboration of clinician scientists with medicinal chemists and pharmacologists who can identify compounds with an adequate safety profile and biological activity against relevant disease mechanisms.
Article
Purpose of review: Noncoding RNAs regulate many aspects of cardiovascular biology and are potential therapy targets. In this review, we summarize and highlight current discoveries in the field of microRNAs, a class of noncoding RNAs. Recent findings: miRNAs regulate posttranscriptional gene expression and have been shown to control cardiac development, hypertrophy, fibrosis, and regeneration. Of note are the miRNAs that regulate cardiac contractility (for example, miR-25 and miR-22), cardiac regeneration (like miR-302-367 and miR99/100 families), and fibrosis (as miR-125b). Consistently with these roles of miRNAs, pharmacological intervention using anti-miRNA oligonucleotides (antagomirs or LNA-anti-miRs) has been shown to improve cardiac contractility and mitigate fibrosis, alleviating cardiac dysfunction in the setting of heart failure. Summary: miRNAs are crucial regulators of cardiac phenotype and have enthused both basic scientists and clinicians alike. With advancement of technology and better understanding of mechanisms governing miRNA deregulation, we are at the crossroads for deciphering miRNA function and modulating it for therapeutics.