Anda di halaman 1dari 9

Hum Genet (2014) 133:471479

DOI 10.1007/s00439-014-1419-3

Review Paper

The genetic basis ofpulmonary arterial hypertension


LijiangMa WendyK.Chung

Received: 14 October 2013 / Accepted: 7 January 2014 / Published online: 21 January 2014
Springer-Verlag Berlin Heidelberg 2014

Abstract Pulmonary arterial hypertension (PAH) is a rare


disease characterized by distinctive changes in pulmonary
arterioles that lead to progressive elevation of pulmonary
artery pressure, pulmonary vascular resistance, right ventricular failure, and a high mortality rate. The etiology of
PAH is heterogeneous and incompletely understood. Based
on clinical classification, WHO Group 1 PAH includes
sporadic disease (idiopathic PAH), inherited PAH (heritable PAH), and association with certain medical conditions
(associated PAH). Genes play an important role in idiopathic and heritable PAH. Mutations in bone morphogenetic protein receptor 2 (BMPR2), a member of the transforming growth factor (TGF) superfamily of receptors,
have been identified in 70% of cases of familial PAH, as
well as in 1040% of cases of idiopathic PAH. Mutations
in ALK-1, ENG, SMAD4 and SMAD8, other TGF family
members, are additional rare causes of PAH. CAV1 regulates SMAD2/3 phosphorylation, and mutations in CAV1
are a rare cause of PAH. KCNK3 is a member of the twopore domain potassium channels expressed in pulmonary
artery smooth muscle cells, and mutations in KCNK3 are
a rare cause of both familial and IPAH. The genetics of
PAH are complex due to incomplete penetrance and genetic

L.Ma W.K.Chung(*)
Department ofPediatrics, Columbia University, New York, USA
e-mail: wkc15@columbia.edu
L. Ma
e-mail: lm2689@columbia.edu
W.K.Chung
Department ofMedicine, Columbia University, New York, USA
W.K.Chung
1150 St. Nicholas Avenue, Room 620, New York, NY 10032,
USA

heterogeneity. In addition to rare mutations as a monogenic


cause of HPAH, common variants in cerebellin 2 (CBLN2)
increase the risk of PAH by approximately twofold. PAH
in children is much more heterogeneous than in adults and
can be associated with several genetic syndromes, specifically syndromes with congenital heart disease, vascular
disease, and hepatic disease. Clinical genetic testing is
available for PAH and should be considered in families to
allow for more definitive risk stratification and allow for
reproductive planning.

Genetics ofPAH
Pulmonary arterial hypertension (PAH) is a rare disease
characterized by increased pulmonary artery pressure in
the absence of common causes of pulmonary hypertension such as heart, lung, and thrombo-embolic chronic
diseases (Gali etal. 2009). PAH remains a progressive,
fatal disease in many patients despite advances in treatment
(Humbert etal. 2010) with a 1-year incident mortality rate
of 15% (DAlonzo etal. 1991). The pathogenesis of pulmonary arterial hypertension is complex and incompletely
understood, but includes both genetic and environmental
factors that alter vascular structure and function (Humbert
etal. 2010).
Familial cases of PAH have been long recognized (Dresdale etal. 1954) and are usually autosomal dominantly
inherited. The most recent pulmonary hypertension (PH)
classification (4th PH world symposium, Dana Point, 2008)
replaces familial PAH with HPAH at least in part to recognize the fact that 1040% of cases previously thought
to be IPAH harbor identifiable mutations in BMPR2 and,
therefore, pose a hereditary risk to other family members.
Only 6% of PAH patients reported a family history of PAH

13


472

in the prospective National Institutes of Health registry


(Rich etal. 1987). However, a family history of PAH may
go unrecognized in IPAH cases with BMPR2 mutations, as
a consequence of either undiagnosed disease, incomplete
penetrance, or de novo (spontaneous) mutations.
Various strategies that have been used to identify the
PAH associated genes and have evolved as genomic technologies evolved. Initially large families were required
for linkage analysis which mapped PPH1 to chromosome
2q31-q32 (Morse etal. 1997). Positional candidate genes at
2q21-32 were sequenced, and bone morphogenetic protein
receptor type II (BMPR2) was identified as the first major
genetic cause of PAH (International PPH Consortium etal.
2000; Deng etal. 2000a, b). With the advent of whole
exome sequencing, rarer monogenic causes of PAH have
been identified utilizing smaller families than required for
linkage (Austin etal. 2012; Ma etal. 2013). In addition to
identifying rare, monogenic causes of PAH, genome-wide
association study (GWAS) has been successfully applied to
identify common variants predisposing to PAH (Germain
etal. 2013).

BMPR2 andthe TGF family inHPAH


Two laboratories independently mapped the first HPAH
locus, PPH1, to chromosome 2q31-32 (Morse etal. 1997;
Nichols etal. 1997). Fine mapping of the originally large
27cM PPH1 locus narrowed the minimal genetic region
to ~3cM (Deng etal. 2000a, b) thus enabling nucleic acid
sequencing of candidate genes. Two groups independently
and simultaneously identified bone morphogenetic protein
receptor II gene (BMPR2) as the PPH1 gene (Deng etal.
2000a, b; International PPH Consortium etal. 2000).
A mutation in BMPR2 can be identified in ~70% of
HPAH and in ~1040% of IPAH cases (Aldred etal. 2006;
Cogan etal. 2005, 2006; Thomson etal. 2000). BMPR2 is
a member of the transforming growth factor-beta (TGF-)
receptor superfamily.
To date, mutations have been recorded in over 70% of
subjects with one or more affected relatives (Aldred etal.
2007; Machado etal. 2006; Souza etal. 2008; Thomson
etal. 2000). A total of 298 BMPR2 unique mutations have
been identified so far (Machado etal. 2009). The spectrum
of BMPR2 mutations in hereditary and spontaneous disease
is analogous, comprising all major classes of loss of function mutations. Mutations also include larger deletions or
re-arrangements affecting one or more exons or the entire
gene and are not readily detectable by sequencing methods. These mutations were identified in probands, including
those with a known PAH family history, sporadic onset of
disease, and PAH associated with other disorders. All mutations studied to date appear to have independently arisen,

13

Hum Genet (2014) 133:471479

suggesting that genetic founder events are uncommon


(Machado etal. 2006).
The TGF- superfamily comprises a large series of
cytokine growth factors that control a host of cellular functions, among them proliferation, migration, differentiation,
apoptosis, and extracellular matrix secretion and deposition. TGF- members are segregated into several subfamilies, which include the prototypic TGF- ligands, receptors,
and accessory molecules, activins, and the largest of these
groups, the bone morphogenetic proteins (Shi and Massague 2003). The implication of BMPR2, ALK-1, ENG,
SMAD4, and SMAD8 as causal factors in hereditary and
associated forms of PAH has emphasized the critical importance of this pathway to the integrity of the pulmonary
vasculature (Harrison etal. 2003) (Fig.1). Heterozygous
germline mutations in activin-like kinase-type I (ALK1)
(Trembath etal. 2001) and Endoglin (ENG) (Chaouat etal.
2004) cause hereditary hemorrhagic telangiectasia (HHT)
and may rarely lead to the development of PAH. HHT is
an autosomal dominant vascular disorder characterized by
the appearance of cutaneous telangiectasias and arteriovenous malformations. Mutations in the SMAD4 gene have
been linked to HHT as well as juvenile polyposis and HHT
(Nasim etal. 2011). A small proportion of HHT patients
have PAH that is clinically and histopathologically indistinguishable from other heritable forms of PAH, while others
have PAH associated with pulmonary arteriovenous fistulas
(Harrison etal. 2003). The underlying causative factor in
these patients is, typically, mutations of ALK-1. Up to 20%
(16/83) of all detected mutations in ALK-1 are associated
with the development of PAH, and, of these, 81% (13/16)
are consistently observed with PAH (Abdalla etal. 2005;
Prigoda etal. 2006). In rare instances, mutations of ALK-1
appear to cause IPAH or HPAH without HHT (Greenwald
etal. 1999; Tew etal. 2002; Trembath etal. 2001).
SMAD8 has also been recently recognized as a possible
cause of PAH (Shintani etal. 2009). One Japanese family
was discovered to have a nonsense mutation in this gene,
leading to a truncated protein (Shintani etal. 2009). Moreover, another BMP receptor BMPR1B (ALK6) is rarely associated with PAH, and two variants in BMPR1B were found
in IPAH patients (Chida etal. 2012).
Mutations in caveolin 1 (CAV1) are a rare cause of PAH,
and two frameshift mutations were identified in both familial cases (frequency 1/62) and IPAH (frequency 1/198)
(Austin etal. 2012). The expression of CAV1 is necessary
for the formation of caveolae, which are 50100nm diameter plasma membrane invaginations and cytoplasmic vesicles presented in numerous cell types including endothelial
and smooth muscle cells of the systemic and pulmonary
vasculature, and type I alveolar cells of the lung. Caveolae are rich in cell surface receptors critical to initiation
of a cellular signaling cascade. CAV1 mutations are loss of

Hum Genet (2014) 133:471479

473

Fig.1Molecular pathogenesis of hereditary PAH. BMP binds to


BMPR2. Upon ligand binding, the type II receptor phosphorylates a
type I receptor, including ALK1, ALK2, ALK3 or ALK6. This leads
to phosphorylation of Smad1/5/8 and phosphorylation of Smad4 with
translocation of the phosphorylated Smads to the nucleus to modulate
the expression of target genes. Upon TGF ligand binding, the TGF
type II receptor phosphorylates a type I receptor, ALK5. This leads to
phosphorylation of Smad2/3which phosphorylate Smad4 and translocate to the nucleus. Endoglin is an accessory membrane glycoprotein that interacts with signaling receptor complexes for the BMP and
TGF- superfamily. Caveolin-1 normally dampens BMP signaling by
inhibiting receptors or their signaling downstream to prevent vascular

proliferation (Maniatis etal. 2008). Lack of caveolin-1 causes activation of STAT3 and ERK1/2 signaling (Mathew etal. 2004), activation
of Ras/p42/44/MAP kinase and upregulation of cyclin D1 (Razani
etal. 2001; Schwencke etal. 2006). Caveolin-1 functions as a tonic
inhibitor of eNOS to facilitate NO mediated relaxation (Razani etal.
2001). Caveolin-1 modifies TGF beta signaling at the plasma membrane which may provide a mechanistic link between CAV1 and
BMPR2 mutations in the pathogenesis of PAH (Austin etal. 2012).
KCNK3 is a potassium channel protein in pulmonary artery smooth
muscle cells. Activation of K+ channel causes K+ efflux, membrane
hyper polarization, and vasodilatation

function mutations resulting in haploinsufficiency. CAV1


directly down regulates Smad2/3 phosphorylation and loss
of CAV1 enhanced Smad2/3 signaling which has been
observed in the lungs of patients with PAH and other types
of pulmonary hypertension, providing a mechanistic link
to the TGF receptor superfamily abnormalities (Phillips
etal. 2008).
The genetics of PAH are complex due to incomplete
penetrance. Almost all hereditary forms of PAH are autosomal dominantly inherited. The majority of families with
HPAH have mutations in BMPR2. The penetrance of the
BMPR2 mutation is low with an estimated lifetime risk
of 20% (Loyd etal. 1995; Rich etal. 1987; Sztrymf etal.
2007). There is an influence of gender on the development
of PAH. Female mutation carriers are about 2.5-fold more
likely to develop FPAH than those of males (Loyd 2002)
with a female to male ratio of 1.7:1 (Larkin etal. 2012).
Some have hypothesized a second genetic hit within the
pulmonary vasculature including epigenetic changes in
Superoxide dismutase 2 (Archer etal. 2010) or somatic
chromosomal abnormalities and quasineoplastic cells

leading to endothelial cell proliferation (Cogan etal. 2006).


One BMPR2 carrier has been shown to have a loss of chromosome 13 in these cells associated with loss of SMAD-8
(Aldred etal. 2010). Alterations in BMPR2 isoform ratios
may provide an explanation of the reduced penetrance
among BMPR2 mutation carriers (Cogan etal. 2012). The
disease is more frequent in adult women, with a ratio of
ranges from 2:1 to 4:1 women to men (Frost etal. 2011;
Humbert etal. 2006). Both incomplete penetrance and the
significantly skewed gender ratio only after puberty suggest
that the BMPR2 mutation alone may not be sufficient to
cause PAH. A second hit which may include other genetic
and/or environmental modifiers of BMPR2 may be necessary to induce development of PAH, indicted by a linkage
study that BMPR2 and the 3q22 locus interact epistatically
(Rodriguez-Murillo etal. 2010). Strong candidates for
modifiers may include sex hormones with altered androgen
to estrogen balance, given the preference of female gender
in PAH and the observation that the majority of women
with PAH are postmenopausal (Scorza etal. 2002; Zamanian etal. 2009). Heterozygous BMPR2 sequence variants

13


474

have been identified in a small subset of patients with PAH


associated with (relatively brief) exposure to fenfluramine
(Humbert etal. 2002; Souza etal. 2008), congenital heart
disease (Roberts etal. 2004), and veno-occlusive disease
(Montani etal. 2008; Runo etal. 2003) raising the question as to whether such factors represent disease triggers in
the face of inherited susceptibility in some patients. In contrast, BMPR2 mutations have not previously been identified
in modestly sized series of PAH associated with the scleroderma-spectrum disease, with human immunodeficiency
virus (Nunes etal. 2003; Tew etal. 2002), Down syndrome
(Greenwood and Nadas 1976), neurofibromatosis 1 (Stewart etal. 2007), Gaucher disease (Lo etal. 2011) or autoimmune polyendocrine syndrome (Alghamdi etal. 2010).

HPAH due toKCNK3 mutations


Mutations in the potassium channel, KCNK3, are a rare
cause of both familial and IPAH (Ma etal. 2013). KCNK3
is a member of the two-pore domain potassium channels
expressed in pulmonary artery smooth muscle cells and
thought to be responsible for setting the resting membrane
potential, responding to hypoxia, and regulating pulmonary
vascular tone (Gurney etal. 2003; Hartness etal. 2001;
Nagaraj etal. 2013; Olschewski etal. 2006; Osipenko
etal. 1997). We identified a novel heterozygous missense
variant c.608 G>A, p.G203D in KCNK3 in a familial PAH
case. We then independently identified five additional heterozygous missense variants (E182K, T8K, Y192C, G97R,
V221L) in 2 of 82 unrelated familial PAH and 3 of 230 idiopathic PAH patients. All six rare variants are novel and
located in highly conserved domains and predicted to be
damaging. Electrophysiological patch clamp studies of the
mutant channels demonstrated that all six missense mutations result in loss-of-function at physiological pH (Ma
etal. 2013). Application of the phospholipase A2 inhibitor
ONO-RS-082 was capable of rescuing channel activity in
some but not all disease associated mutants. This suggests
a potential novel mechanism for therapeutic intervention
by pharmacologically increasing currents through KCNK3
in patients with pulmonary arterial hypertension (Ma etal.
2013).

Autosomal recessive HPAH


Recently, mutations in Eukaryotic Translation Initiation
Factor 2 Alpha Kinase 4 EIF2AK4 were identified in multiple families with pulmonary capillary hemangiomatosis
(PCH)/PAH-associated veno-occlusive disease (PVOD)
(Best etal. 2013; Eyries etal. 2013). All affected subjects
in 14 familial cases carried deleterious homozygous or

13

Hum Genet (2014) 133:471479

compound-heterozygous rare variants in EIF1AK4, consistent with autosomal recessive inheritance. Additional
mutation screening by two research teams demonstrated
EIF1AK4 mutations were presented in 5/20 (25%) and
2/10 (20%) of idiopathic cases of PCH/PVOD (Best etal.
2013; Eyries etal. 2013). The protein product of EIF2AK4
belongs to a family of kinases that regulates angiogenesis,
and the alpha subunit of this protein plays a critical role in
induction of angiogenesis, proliferation, and resistance to
apoptosis in stressful environments. EIF2AK4 was found to
interact with SMAD4, SMAD1, ALK-1, endoglin (ENG),
and transforming growth factor- receptor-2 (TGFBR2),
the same signaling network through which BMPR2 acts.
EIF2AK4 mutations are associated with both PVOD and
PCH and further elucidate the underlying molecular genetic
etiology common to the two disease classifications and suggest that perhaps the classification should be revised based
upon underlying etiology.

Genetic complexity ofmonogenic forms ofHPAH


Families with BMPR2 mutations have been reported to
have genetic anticipation, or earlier age of diagnosis in
subsequent generations (Loyd etal. 1995). However, no
systematic population-based study has been performed to
avoid the ascertainment bias that could result in the recruitment and study of families associated with earlier-onset
disease in more recent generations. Furthermore, the usual
genetic mechanisms for anticipation, including trinucleotide repeat expansions, are not present in BMPR2.
Heritable PAH and idiopathic PAH have a similar clinical course. Heritable PAH due to BMPR2 mutations is associated with an earlier age of onset (even in cases without a
family history of PAH) and a slightly more severe hemodynamic impairment at diagnosis: patients with HPAH
had higher mean pulmonary artery pressure, lower cardiac index, and higher pulmonary vascular resistance, but
interestingly had similar survival although they were more
likely to be treated with parenteral prostacyclin therapy or
lung transplant (Sztrymf etal. 2008). Both children and
adults with PAH and BMPR2 mutations are, however, less
likely to respond to acute vasodilator testing during right
heart catheterization and are unlikely to benefit from treatment with calcium channel blockade (Elliott etal. 2006;
Rosenzweig etal. 2008). Symptomatic HPAH patients carrying ALK1 mutations, most without HHT, have an earlier age of onset and more rapid disease progression than
HPAH patients with BMPR2 mutations despite responsiveness to vasodilators at the time of diagnosis (Girerd etal.
2010a, b). There has been controversy over the genotype
phenotype correlations with BMPR2 mutations. Some have
claimed that missense mutations are associated with a more

Hum Genet (2014) 133:471479

severe phenotype than truncating mutations (Austin etal.


2009), but this result has not been replicated (Girerd etal.
2010a, b). At this time, many PAH experts do not routinely
use genetic test results to guide management of the patient
with PAH although the information might suggest the need
for more aggressive therapy and/or earlier consideration of
transplant.

Common variants increase risk ofPAH


In addition to rare mutations as a monogenic cause of
HPAH, a two-stage genome-wide association study
(GWAS) was performed in idiopathic and familial PAH to
identify common variants predisposing to PAH. In a study
with a discovery and confirmation cohort with a total of 625
cases and 1,525 healthy individuals, there was a significant
association at the Cerebellin 2 (CBLN2) locus mapping to
18q22.3, with the risk allele conferring an odds ratio for
PAH of 1.97 (Germain etal. 2013). CBLN2 is expressed
in the lung, with higher expression in explanted lungs from
individuals with PAH and in endothelial cells cultured from
explanted PAH lungs. The hypothesis is that CBLN2 synthesized by endothelial cells might act on vascular SMC
proliferation in a paracrine fashion (Germain etal. 2013).

Pediatric PAH
PAH in children has a wider range of genetic etiologies
than in adults. Pediatric PAH is associated with genetic syndromes with and without congenital heart disease, vascular
disease, and hepatic disease. With all of these syndromes,
PAH is an uncommon complication although in certain
syndromes such as Down syndrome PAH is more common (Nichols etal. 1997). Genetic syndromes more commonly, but not necessarily, associated with CHD and PAH
also include DiGeorge syndrome, VACTERL syndrome,
CHARGE syndrome, Scimitar syndrome (Vida etal. 2010),
Noonan syndrome (Tinker etal. 1989), and chromosomal
anomalies associated with congenital diaphragmatic hernia. Genetic syndromes associated with PAH usually not
associated with CHD include Adams-Oliver syndrome
(Patel etal. 2004; Piazza etal. 2004), Neurofibromatosis
1 (Deng etal. 2000a, b), Long QT syndrome, hypertrophic
cardiomyopathy, Cantu syndrome (Kobayashi etal. 2010)
autoimmune polyendocrine syndrome (International PPH
Consortium etal. 2000), mitochondrial disorders including
mitochondrial encephalopathy lactic acidosis and strokelike episodes (Sproule etal. 2008) Gaucher disease (Deng
etal. 2000a, b), and glycogen storage diseases (GSDI and
GSDIII) (Lee etal. 2011). The mechanism for development

475

of PH has not been definitely demonstrated for most


genetic syndromes but could involve increased pulmonary
blood flow with left to right shunts with CHD, pulmonary
venous obstruction (Cantu syndrome) (Kobayashi etal.
2010) or production of diffusible hepatic factors increasing
the pulmonary pressures (Gaucher disease and GSD) (Lee
etal. 2011). For all of these genetic disorders a common
question is why the majority of patients with these disorders never develop PAH and what is different about those
who do.

Genetic testing forPAH


Clinical genetic testing is available for PAH for BMPR2,
ALK1, ENG, SMAD8, and CAV1 genes including both
sequencing and deletion/duplication testing. Genetic testing is available for either single genes or the entire panel
of genes. The cost of testing has recently decreased in the
US with the introduction of these gene panels, increasing
the accessibility of testing to patients. Genetic testing is
often considered in pediatric PAH to explain the etiology
of the disease and counsel family members about identifying other family members at risk and accurately determining the risk of recurrence in future children. The most
commonly cited reasons for genetic testing for PAH is to
provide information to and about children (Jones etal.
2008). Pre-implantation genetic diagnosis has been used to
avoid transmission to offspring once the familial mutation
was identified (Frydman etal. 2012).
Using the combined approaches of sequencing and deletion duplication analysis, approximately 75% of HPAH
patients will have an identified mutation in a PAH gene,
most commonly in BMPR2 (Machado etal. 2001). Clinical
genetic testing is available in North America and Europe,
with the current cost of testing usually covered by insurance with results generally available in 8weeks. Once the
mutation in a family is known, testing other family members for a family-specific mutation in US is relatively inexpensive, accurate, and fast. As a consequence of the incomplete penetrance and variable age of onset, identification of
a BMPR2 mutation may have a complex and serious psychosocial impact on the family and is often associated with
feelings of guilt in the parent who has passed on mutations
to the children. Genetic testing is most helpful when it is
able to identify members of the family who are not genetically at risk for PAH, and who can then forgo the otherwise
recommended serial evaluations to screen for development
of PAH. For at-risk family members, regular screening by
echocardiogram every 3years and awareness of disease
symptoms should enable early diagnosis and treatment
which may improve outcomes.

13


476

Conclusion
The genetics of pulmonary hypertension are complex.
While loss-of-function mutations of some genes, such as
BMPR2, have been established as major monogenetic risk
factor for PAH, their disease penetrance is modest, suggesting other genetic or environmental modifiers. The skewed
gender ratio for IPAH and HPAH after puberty suggests
that these modifiers may be gender specific and could be
related to hormonal differences. Genetic testing is available
for PAH and should be considered in families to identify
individuals who are at increased risk of developing PAH.
Positively tested individuals will undergo close clinical follow-up for PAH development and may make reproductive
decisions based upon an increased risk of recurrence.
Acknowledgments This work was supported by R01 HL060056.

References
Abdalla SA, Cymerman U, Rushlow D, Chen N, Stoeber GP, Lemire
EG, Letarte M (2005) Novel mutations and polymorphisms in
genes causing hereditary hemorrhagic telangiectasia. Hum Mutat
25(3):320321. doi:10.1002/humu.9312
Aldred MA, Vijayakrishnan J, James V, Soubrier F, Gomez-Sanchez
MA, Martensson G, Galie N, Manes A, Corris P, Simonneau G,
Humbert M, Morrell NW, Trembath RC (2006) BMPR2 gene
rearrangements account for a significant proportion of mutations
in familial and idiopathic pulmonary arterial hypertension. Hum
Mutat 27(2):212213. doi:10.1002/humu.9398
Aldred MA, Machado RD, James V, Morrell NW, Trembath RC
(2007) Characterization of the BMPR2 5-untranslated region and
a novel mutation in pulmonary hypertension. Am J Respir Crit
Care Med 176(8):819824. doi:10.1164/rccm.200701-164OC
Aldred MA, Comhair SA, Varella-Garcia M, Asosingh K, Xu W,
Noon GP, Thistlethwaite PA, Tuder RM, Erzurum SC, Geraci
MW, Coldren CD (2010) Somatic chromosome abnormalities in the lungs of patients with pulmonary arterial hypertension. Am J Respir Crit Care Med 182(9):11531160.
doi:10.1164/rccm.201003-0491OC
Alghamdi MH, Steinraths M, Panagiotopoulos C, Potts JE, Sandor
GG (2010) Primary pulmonary arterial hypertension and autoimmune polyendocrine syndrome in a pediatric patient. Pediatr Cardiol 31(6):872874. doi:10.1007/s00246-010-9704-y
Archer SL, Marsboom G, Kim GH, Zhang HJ, Toth PT, Svensson EC,
Dyck JR, Gomberg-Maitland M, Thbaud B, Husain AN, Cipriani N, Rehman J (2010) Epigenetic attenuation of mitochondrial
superoxide dismutase 2 in pulmonary arterial hypertension: a
basis for excessive cell proliferation and a new therapeutic target. Circulation 121(24):26612671. doi:10.1161/CIRCULATIO
NAHA.109.916098
Austin ED, Phillips JA, Cogan JD, Hamid R, Yu C, Stanton KC, Phillips CA, Wheeler LA, Robbins IM, Newman JH, Loyd JE (2009)
Truncating and missense BMPR2 mutations differentially affect
the severity of heritable pulmonary arterial hypertension. Respir
Res 10:87. doi:10.1186/1465-9921-10-87
Austin ED, Ma L, LeDuc C, Berman Rosenzweig E, Borczuk A, Phillips JA 3rd, 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

13

Hum Genet (2014) 133:471479


arterial hypertension. Circ Cardiovasc Genet 5(3):336343. doi:1
0.1161/CIRCGENETICS.111.961888
Best DH, Sumner KL, Austin ED, Chung WK, Brown LM, Borczuk
AC, Rosenzweig EB, Bayrak-Toydemir P, Mao R, Cahill BC,
Tazelaar HD, Leslie KO, Hemnes AR, Robbins IM, Elliott CG
(2013) EIF2AK4 Mutations in pulmonary capillary hemangiomatosis. Chest. doi:10.1378/chest.13-2366
Chaouat A, Coulet F, Favre C, Simonneau G, Weitzenblum E, Soubrier F, Humbert M (2004) Endoglin germline mutation in a
patient with hereditary haemorrhagic telangiectasia and dexfenfluramine associated pulmonary arterial hypertension. Thorax
59(5):446448. doi:10.1136/thx.2003.11890
Chida A, Shintani M, Nakayama T, Furutani Y, Hayama E, Inai K,
Saji T, Nonoyama S, Nakanishi T (2012) Missense mutations of
the BMPR1B (ALK6) gene in childhood idiopathic pulmonary
arterial hypertension. Circ J 76(6):15011508. doi:10.1253/circj
.CJ-11-1281
Cogan JD, Vnencak-Jones CL, Phillips JA 3rd, Lane KB, Wheeler
LA, Robbins IM, Garrison G, Hedges LK, Loyd JE (2005) Gross
BMPR2 gene rearrangements constitute a new cause for primary
pulmonary hypertension. Genet Med 7(3):169174
Cogan JD, Pauciulo MW, Batchman AP, Prince MA, Robbins
IM, Hedges LK, Stanton KC, Wheeler LA, Phillips JA 3rd,
Loyd JE, Nichols WC (2006) High frequency of BMPR2
exonic deletions/duplications in familial pulmonary arterial
hypertension. Am J Respir Crit Care Med 174(5):590598.
doi:10.1164/rccm.200602-165OC
Cogan J, Austin E, Hedges L, Womack B, West J, Loyd J, Hamid R
(2012) Role of BMPR2 alternative splicing in heritable pulmonary arterial hypertension penetrance. Circulation 126(15):1907
1916. doi:10.1161/CIRCULATIONAHA.112.106245
DAlonzo GE, Barst RJ, Ayres SM, Bergofsky EH, Brundage BH, Detre
KM, Fishman AP, Goldring RM, Groves BM, Kernis JT, Levy PS,
Pietra GG, Reid LM, Reeves JT, Rich S, Vreim CE, Williams GW,
Wu M (1991) Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. Ann Intern
Med 115(5):343349. doi:10.7326/0003-4819-115-5-343
Deng Z, Haghighi F, Helleby L, Vanterpool K, Horn EM, Barst RJ,
Hodge SE, Morse JH, Knowles JA (2000a) Fine mapping of
PPH1, a gene for familial primary pulmonary hypertension, to a
3-cM region on chromosome 2q33. Am J Respir Crit Care Med
161(3 Pt 1):10551059. doi:10.1164/ajrccm.161.3.9906051
Deng Z, Morse JH, Slager SL, Cuervo N, Moore KJ, Venetos G,
Kalachikov S, Cayanis E, Fischer SG, Barst RJ, Hodge SE,
Knowles JA (2000b) Familial primary pulmonary hypertension
(gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Genet 67(3):737744.
doi:10.1086/303059
Dresdale DT, Michtom RJ, Schultz M (1954) Recent studies in primary pulmonary hypertension, including pharmacodynamic
observations on pulmonary vascular resistance. Bull NY Acad
Med 30(3):195207
Elliott CG, Glissmeyer EW, Havlena GT, Carlquist J, McKinney
JT, Rich S, McGoon MD, Scholand MB, Kim M, Jensen RL,
Schmidt JW, Ward K (2006) Relationship of BMPR2 mutations to vasoreactivity in pulmonary arterial hypertension. Circulation
113(21):25092515.
doi:10.1161/CIRCULATION
AHA.105.601930
Eyries M, Montani D, Girerd B, Perret C, Leroy A, Lonjou C, Chelghoum N, Coulet F, Bonnet D, Dorfmller P, Fadel E, Sitbon
O, Simonneau G, Tregout DA, Humbert M, Soubrier F (2013)
EIF2AK4 mutations cause pulmonary veno-occlusive disease, a recessive form of pulmonary hypertension. Nat Genet.
doi:10.1038/ng.2844
Frost AE, Badesch DB, Barst RJ, Benza RL, Elliott CG, Farber HW,
Krichman A, Liou TG, Raskob GE, Wason P, Feldkircher K,

Hum Genet (2014) 133:471479


Turner M, McGoon MD (2011) The changing picture of patients
with pulmonary arterial hypertension in the United States: how
REVEAL differs from historic and non-US Contemporary Registries. Chest 139(1):128137. doi:10.1378/chest.10-0075
Frydman N, Steffann J, Girerd B, Frydman R, Munnich A, Simonneau G, Humbert M (2012) Pre-implantation genetic diagnosis
in pulmonary arterial hypertension due to BMPR2 mutation. Eur
Respir J 39(6):15341535. doi:10.1183/09031936.00185011
Gali N, Hoeper MM, Humbert M, Torbicki A, Vachiery JL, Barbera
JA, Beghetti M, Corris P, Gaine S, Gibbs JS, Gomez-Sanchez
MA, Jondeau G, Klepetko W, Opitz C, Peacock A, Rubin L,
Zellweger M, Simonneau G, ESC Committee for Practice Guidelines (CPG) (2009) Guidelines for the diagnosis and treatment
of pulmonary hypertension: the task force for the diagnosis and
treatment of pulmonary hypertension of the European Society of
Cardiology (ESC) and the European Respiratory Society (ERS),
endorsed by the International Society of Heart and Lung Transplantation (ISHLT). Eur Heart J 20:24932537. doi:10.1093/eur
heartj/ehp297
Germain M, Eyries M, Montani D, Poirier O, Girerd B, Dorfmller
P, Coulet F, Nadaud S, Maugenre S, Guignabert C, Carpentier W,
Vonk-Noordegraaf A, Lvy M, Chaouat A, Lambert JC, Bertrand
M, Dupuy AM, Letenneur L, Lathrop M, Amouyel P, de Ravel TJ,
Delcroix M, Austin ED, Robbins IM, Hemnes AR, Loyd JE, Berman-Rosenzweig E, Barst RJ, Chung WK, Simonneau G, Trgout
DA, Humbert M, Soubrier F (2013) Genome-wide association
analysis identifies a susceptibility locus for pulmonary arterial
hypertension. Nat Genet 45(5):518521. doi:10.1038/ng.2581
Girerd B, Montani D, Coulet F, Sztrymf B, Yaici A, Jas X, Tregouet
D, Reis A, Drouin-Garraud V, Fraisse A, Sitbon O, OCallaghan
DS, Simonneau G, Soubrier F, Humbert M (2010a) Clinical
outcomes of pulmonary arterial hypertension in patients carrying an ACVRL1 (ALK1) mutation. Am J Respir Crit Care Med
181(8):851861. doi:10.1164/rccm.200908-1284OC
Girerd B, Montani D, Eyries M, Yaici A, Sztrymf B, Coulet F, Sitbon O, Simonneau G, Soubrier F, Humbert M (2010b) Absence
of influence of gender and BMPR2 mutation type on clinical phenotypes of pulmonary arterial hypertension. Respir Res 11:73.
doi:10.1186/1465-9921-11-73
Greenwald J, Fischer WH, Vale WW, Choe S (1999) Three-finger
toxin fold for the extracellular ligand-binding domain of the type
II activin receptor serine kinase. Nat Struct Biol 6(1):1822.
doi:10.1038/4887
Greenwood RD, Nadas AS (1976) The clinical course of cardiac disease in Downs syndrome. Pediatrics 58(6):893897
Gurney AM, Osipenko ON, MacMillan D, McFarlane KM, Tate RJ,
Kempsill FE (2003) Two-pore domain K channel, TASK-1, in
pulmonary artery smooth muscle cells. Circ Res 93(10):957964.
doi:10.1161/01.RES.0000099883.68414.61
Harrison RE, Flanagan JA, Sankelo M, Abdalla SA, Rowell J,
Machado RD, Elliott CG, Robbins IM, Olschewski H, McLaughlin V, Gruenig E, Kermeen F, Halme M, Risnen-Sokolowski
A, Laitinen T, Morrell NW, Trembath RC (2003) Molecular
and functional analysis identifies ALK-1 as the predominant
cause of pulmonary hypertension related to hereditary haemorrhagic telangiectasia. J Med Genet 40(12):865871. doi:10.1136
/jmg.40.12.865
Hartness ME, Lewis A, Searle GJ, OKelly I, Peers C, Kemp PJ
(2001) Combined antisense and pharmacological approaches
implicate hTASK as an airway O(2) sensing K(+) channel. J Biol
Chem 276(28):2649926508. doi:10.1074/jbc.M010357200
Humbert M, Deng Z, Simonneau G, Barst RJ, Sitbon O, Wolf M,
Cuervo N, Moore KJ, Hodge SE, Knowles JA (2002) Morse JH
(2002) BMPR2 germline mutations in pulmonary hypertension
associated with fenfluramine derivatives. Eur Respir J 20(3):518
523. doi:10.1183/09031936.02.01762002

477
Humbert M, Sitbon O, Chaouat A, Bertocchi M, Habib G, Gressin
V, Yaici A, Weitzenblum E, Cordier JF, Chabot F, Dromer C,
Pison C, Reynaud-Gaubert M, Haloun A, Laurent M, Hachulla E,
Simonneau G (2006) Pulmonary arterial hypertension in France:
results from a national registry. Am J Respir Crit Care Med
173(9):10231030. doi:10.1164/rccm.200510-1668OC
Humbert M, Sitbon O, Chaouat A, Bertocchi M, Habib G, Gressin
V, Yaci A, Weitzenblum E, Cordier JF, Chabot F, Dromer C,
Pison C, Reynaud-Gaubert M, Haloun A, Laurent M, Hachulla
E, Cottin V, Degano B, Jas X, Montani D, Souza R, Simonneau
G (2010) Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern
management era. Circulation 122(2):156163. doi:10.1161/CIRC
ULATIONAHA.109.911818
International PPH Consortium, Lane KB, Machado RD, Pauciulo
MW, Thomson JR, Phillips JA 3rd, 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):8184. doi:10.1038/79226
Jones DL, Sandberg JC, Rosenthal MJ, Saunders RC, Hannig VL,
Clayton EW (2008) What patients and their relatives think about
testing for BMPR2. J Genet Couns 17(5):452458. doi:10.1007/
s10897-008-9172-1
Kobayashi D, Cook AL, Williams DA (2010) Pulmonary hypertension secondary to partial pulmonary venous obstruction in a child
with Cantu syndrome. Pediatr Pulmonol 45(7):727729. doi:10.1
002/ppul.21215
Larkin EK, Newman JH, Austin ED, Hemnes AR, Wheeler L, Robbins
IM, West JD, Phillips JA 3rd, Hamid R, Loyd JE (2012) Longitudinal analysis casts doubt on the presence of genetic anticipation
in heritable pulmonary arterial hypertension. Am J Respir Crit
Care Med 186(9):892896. doi:10.1164/rccm.201205-0886OC
Lee TM, Berman-Rosenzweig ES, Slonim AE, Chung WK
(2011) Two cases of pulmonary hypertension associated
with Type III glycogen storage disease. JIMD Rep 1:7982.
doi:10.1007/8904_2011_20
Lo SM, Liu J, Chen F, Pastores GM, Knowles J, Boxer M, Aleck K,
Mistry PK (2011) Pulmonary vascular disease in Gaucher disease: clinical spectrum, determinants of phenotype and longterm outcomes of therapy. J Inherit Metab Dis 34(3):643650.
doi:10.1007/s10545-011-9313-9
Loyd JE (2002) Parker B. Francis Lecture. Genetics and gene expression in pulmonary hypertension. Chest 121(3 Suppl):46S50S
Loyd JE, Butler MG, Foroud TM, Conneally PM, Phillips JA 3rd, Newman JH (1995) Genetic anticipation and abnormal gender ratio at
birth in familial primary pulmonary hypertension. Am J Respir Crit
Care Med 152(1):9397. doi:10.1164/ajrccm.152.1.7599869
Ma L, Roman-Campos D, Austin ED, Eyries M, Sampson K, Soubrier F, Germain M, Trgout D, Borczuk A, Rosenzweig EB,
Girerd B, Montani D, Humbert M, Loyd JE, Kass RS, Chung
WK (2013) A novel channelopathy in pulmonary arterial hypertension. N Engl J Med 369:351361
Machado RD, Pauciulo MW, Thomson JR, Lane KB, Morgan NV,
Wheeler L, Phillips JA 3rd, Newman J, Williams D, Gali N,
Manes A, McNeil K, Yacoub M, Mikhail G, Rogers P, Corris P,
Humbert M, Donnai D, Martensson G, Tranebjaerg L, Loyd JE,
Trembath RC, Nichols WC (2001) BMPR2 haploinsufficiency as
the inherited molecular mechanism for primary pulmonary hypertension. Am J Hum Genet 68(1):92102. doi:10.1086/316947
Machado RD, Koehler R, Glissmeyer E, Veal C, Suntharalingam J,
Kim M, Carlquist J, Town M, Elliott CG, Hoeper M, Fijalkowska
A, Kurzyna M, Thomson JR, Gibbs SR, Wilkins MR, Seeger W,
Morrell NW, Gruenig E, Trembath RC, Janssen B (2006) Genetic
association of the serotonin transporter in pulmonary arterial hypertension. Am J Respir Crit Care Med 173(7):793797.
doi:10.1164/rccm.200509-1365OC

13


478
Machado RD, Eickelberg O, Elliott CG, Geraci MW, Hanaoka M,
Loyd JE, Newman JH, Phillips JA 3rd, Soubrier F, Trembath
RC, Chung WK (2009) Genetics and genomics of pulmonary
arterial hypertension. J Am Coll Cardiol 54(1 Suppl):S32S42.
doi:10.1016/j.jacc.2009.04.015
Maniatis NA, Shinin V, Schraufnagel DE, Okada S, Vogel SM, Malik
AB, Minshall RD (2008) Increased pulmonary vascular resistance and defective pulmonary artery filling in caveolin-1-/mice. Am J Physiol Lung Cell Mol Physiol 294(5):L865L873.
doi:10.1152/ajplung.0 0079.2007
Mathew R, Huang J, Shah M, Patel K, Gewitz M, Sehgal PB (2004)
Disruption of endothelial-cell caveolin-1alpha/raft scaffolding during development of monocrotaline-induced pulmonary
hypertension. Circulation 110(11):14991506. doi:10.1161/01.
CIR.0000141576.39579.23
Montani D, Achouh L, Dorfmller P, Le Pavec J, Sztrymf B, Tchrakian C, Rabiller A, Haque R, Sitbon O, Jas X, Dartevelle P,
Matre S, Capron F, Musset D, Simonneau G, Humbert M (2008)
Pulmonary veno-occlusive disease: clinical, functional, radiologic, and hemodynamic characteristics and outcome of 24 cases
confirmed by histology. Medicine (Baltimore) 87(4):220233.
doi:10.1097/MD.0b013e31818193bb
Morse JH, Jones AC, Barst RJ, Hodge SE, Wilhelmsen KC, Nygaard
TG (1997) Mapping of familial primary pulmonary hypertension locus (PPH1) to chromosome 2q31-q32. Circulation
95(12):26032606. doi:10.1161/01.CIR.95.12.2603
Nagaraj C, Tang B, Blint Z, Wygrecka M, Hrzenjak A, Kwapiszewska G, Stacher E, Lindenmann J, Weir EK, Olschewski H,
Olschewski A (2013) Src tyrosine kinase is crucial for potassium channel function in human pulmonary arteries. Eur Respir J
41(1):8595. doi:10.1183/09031936.00211811
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):13851389. doi:10.1002/humu.21605
Nichols WC, Koller DL, Slovis B, Foroud T, Terry VH, Arnold ND,
Siemieniak DR, Wheeler L, Phillips JA 3rd, Newman JH, Conneally PM, Ginsburg D, Loyd JE (1997) Localization of the gene
for familial primary pulmonary hypertension to chromosome
2q31-32. Nat Genet 15(3):277280. doi:10.1038/ng0397-277
Nunes H, Humbert M, Sitbon O, Morse JH, Deng Z, Knowles
JA, Le Gall C, Parent F, Garcia G, Herv P, Barst RJ, Simonneau G (2003) Prognostic factors for survival in human immunodeficiency virus-associated pulmonary arterial hypertension. Am J Respir Crit Care Med 167(10):14331439.
doi:10.1164/rccm.200204-330OC
Olschewski A, Li Y, Tang B, Hanze J, Eul B, Bohle RM, Wilhelm J,
Morty RE, Brau ME, Weir EK, Kwapiszewska G, Klepetko W,
Seeger W, Olschewski H (2006) Impact of TASK-1 in human pulmonary artery smooth muscle cells. Circ Res 98(8):10721080.
doi:10.1161/01.RES.0000219677.12988.e9
Osipenko ON, Evans AM, Gurney AM (1997) Regulation of the
resting potential of rabbit pulmonary artery myocytes by a
low threshold, O2-sensing potassium current. Br J Pharmacol
120(8):14611470. doi:10.1038/sj.bjp.0701075
Patel MS, Taylor GP, Bharya S, Al-Sannaa N, Adatia I, Chitayat
D, Suzanne Lewis ME, Human DG (2004) Abnormal pericyte
recruitment as a cause for pulmonary hypertension in AdamsOliver syndrome. Am J Med Genet A 129A(3):294299. doi:10
.1002/ajmg.a.30221
Phillips JA 3rd, Poling JS, Phillips CA, Stanton KC, Austin ED,
Cogan JD, Wheeler L, Yu C, Newman JH, Dietz HC, Loyd JE
(2008) Synergistic heterozygosity for TGFbeta1 SNPs and

13

Hum Genet (2014) 133:471479


BMPR2 mutations modulates the age at diagnosis and penetrance
of familial pulmonary arterial hypertension. Genet Med 10:359
365. doi:10.1097/GIM.0b013e318172dcdf
Piazza AJ, Blackston D, Sola A (2004) A case of Adams-Oliver
syndrome with associated brain and pulmonary involvement:
further evidence of vascular pathology? Am J Med Genet A
130A(2):172175. doi:10.1002/ajmg.a.30210
Prigoda NL, Savas S, Abdalla SA, Piovesan B, Rushlow D, Vandezande K, Zhang E, Ozcelik H, Gallie BL, Letarte M (2006)
Hereditary haemorrhagic telangiectasia: mutation detection, test
sensitivity and novel mutations. J Med Genet 43(9):722728. doi:
10.1136/jmg.2006.042606
Razani B, Engelman JA, Wang XB, Schubert W, Zhang XL, Marks
CB, Macaluso F, Russell RG, Li M, Pestell RG, Di Vizio D,
Hou H Jr, Kneitz B, Lagaud G, Christ GJ, Edelmann W, Lisanti
MP (2001) Caveolin-1 null mice are viable but show evidence
of hyperproliferative and vascular abnormalities. J Biol Chem
276(41):3812138138. doi:10.1074/jbc.M105408200
Rich S, Dantzker DR, Ayres SM, Bergofsky EH, Brundage BH, Detre
KM, Fishman AP, Goldring RM, Groves BM, Koerner SK, Levy
PC, Reid LM, Vreim CE, Williams GW (1987) Primary pulmonary hypertension. A national prospective study. Ann Intern Med
107(2):216223. doi:10.7326/0003-4819-107-2-216
Roberts KE, McElroy JJ, Wong WP, Yen E, Widlitz A, Barst RJ,
Knowles JA, Morse JH (2004) BMPR2 mutations in pulmonary
arterial hypertension with congenital heart disease. Eur Respir J
24(3):371374. doi:10.1183/09031936.04.00018604
Rodriguez-Murillo L, Subaran R, Stewart WC, Pramanik S, Marathe S, Barst RJ, Chung WK, Greenberg DA (2010) Novel
loci interacting epistatically with bone morphogenetic protein receptor 2 cause familial pulmonary arterial hypertension. J Heart Lung Transplant 29(2):174180. doi:10.1016/
j.healun.2009.08.022
Rosenzweig EB, Morse JH, Knowles JA, Chada KK, Khan AM,
Roberts KE, McElroy JJ, Juskiw NK, Mallory NC, Rich S, Diamond B, Barst RJ (2008) Clinical implications of determining
BMPR2 mutation status in a large cohort of children and adults
with pulmonary arterial hypertension. J Heart Lung Transplant
27(6):668674. doi:10.1016/j.healun.2008.02.009
Runo JR, Vnencak-Jones CL, Prince M, Loyd JE, Wheeler L, Robbins IM, Lane KB, Newman JH, Johnson J, Nichols WC,
Phillips JA 3rd (2003) Pulmonary veno-occlusive disease
caused by an inherited mutation in bone morphogenetic protein receptor II. Am J Respir Crit Care Med 167(6):889894.
doi:10.1164/rccm.200208-861OC
Schwencke C, Braun-Dullaeus RC, Wunderlich C, Strasser RH
(2006) Caveolae and caveolin in transmembrane signaling:
implications for human disease. Cardiovasc Res 70(1):4249.
doi:10.1016/j.cardiores.2005.11.029
Scorza R, Caronni M, Bazzi S, Nador F, Beretta L, Antonioli R,
Origgi L, Ponti A, Marchini M, Vanoli M (2002) Post-menopause
is the main risk factor for developing isolated pulmonary hypertension in systemic sclerosis. Ann NY Acad Sci 966:238246.
doi:10.1111/j.1749-6632.2002.tb04221.x
Shi Y, Massague J (2003) Mechanisms of TGF-beta signaling from
cell membrane to the nucleus. Cell 113(6):685700. doi:10.1016/
S0092-8674(03)00432-X
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(5):331337. doi:10.1136/
jmg.2008.062703
Souza R, Humbert M, Sztrymf B, Jas X, Yaci A, Le Pavec J, Parent F, Herv P, Soubrier F, Sitbon O, Simonneau G (2008) Pulmonary arterial hypertension associated with fenfluramine
exposure: report of 109 cases. Eur Respir J 31(2):343348.
doi:10.1183/09031936.00104807

Hum Genet (2014) 133:471479


Sproule DM, Dyme J, Coku J, de Vinck D, Rosenzweig E, Chung
WK, De Vivo DC (2008) Pulmonary artery hypertension in
a child with MELAS due to a point mutation of the mitochondrial tRNA((Leu)) gene (m.3243A>G). J Inherit Metab Dis.
doi:10.1007/s10545-007-0735-3
Stewart DR, Cogan JD, Kramer MR, Miller WT Jr, Christiansen LE,
Pauciulo MW, Messiaen LM, Tu GS, Thompson WH, Pyeritz RE,
Ryu JH, Nichols WC, Kodama M, Meyrick BO, Ross DJ (2007)
Is pulmonary arterial hypertension in neurofibromatosis type 1
secondary to a plexogenic arteriopathy? Chest 132(3):798808.
doi:10.1378/chest.06-3017
Sztrymf B, Yaci A, Girerd B, Humbert M (2007) Genes and pulmonary arterial hypertension. Respiration 74(2):123132.
doi:10.1159/000098818
Sztrymf B, Coulet F, Girerd B, Yaici A, Jais X, Sitbon O, Montani
D, Souza R, Simonneau G, Soubrier F, Humbert M (2008) Clinical outcomes of pulmonary arterial hypertension in carriers of
BMPR2 mutation. Am J Respir Crit Care Med 177(12):1377
1383. doi:10.1164/rccm.200712-1807OC
Tew MB, Arnett FC, Reveille JD, Tan FK (2002) Mutations of bone
morphogenetic protein receptor type II are not found in patients
with pulmonary hypertension and underlying connective tissue diseases. Arthritis Rheum 46(10):28292830. doi:10.1002/
art.10487
Thomson JR, Machado RD, Pauciulo MW, Morgan NV, Humbert M,
Elliott GC, Ward K, Yacoub M, Mikhail G, Rogers P, Newman J,
Wheeler L, Higenbottam T, Gibbs JS, Egan J, Crozier A, Peacock

479
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. doi:10.1136/jmg.37.10.741
Tinker AN, Uren N, Schofield J (1989) Severe pulmonary hypertension in Ullrich-Noonan syndrome. Br Heart J 62(1):7477
Trembath RC, Thomson JR, Machado RD, Morgan NV, Atkinson C,
Winship I, Simonneau G, Galie N, Loyd JE, Humbert M, Nichols WC, Morrell NW, Berg J, Manes A, McGaughran J, Pauciulo
M, Wheeler L (2001) Clinical and molecular genetic features of
pulmonary hypertension in patients with hereditary hemorrhagic
telangiectasia. N Engl J Med 345(5):325334. doi:0.1056/N
EJM200108023450503
Vida VL, Padalino MA, Boccuzzo G, Tarja E, Berggren H, Carrel T,
Ciek S, Crupi G, Di Carlo D, Di Donato R, Fragata J, Hazekamp M, Hraska V, Maruszewski B, Metras D, Pozzi M, Pretre R,
Rubay J, Sairanen H, Sarris G, Schreiber C, Meyns B, Tlaskal T,
Urban A, Thiene G, Stellin G (2010) Scimitar syndrome: a European Congenital Heart Surgeons Association (ECHSA) multicentric study. Circulation 122(12):11591166. doi:10.1161/CIRCUL
ATIONAHA.109.926204
Zamanian RT, Hansmann G, Snook S, Lilienfeld D, Rappaport KM,
Reaven GM, Rabinovitch M, Doyle RL (2009) Insulin resistance
in pulmonary arterial hypertension. Eur Respir J 33(2):318324.
doi:10.1183/09031936.00000508

13

Anda mungkin juga menyukai