Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jul 29.
Published in final edited form as: J Am Coll Cardiol. 2009 Jun 30;54(1 0):S32–S42. doi: 10.1016/j.jacc.2009.04.015

Genetics and Genomics of Pulmonary Arterial Hypertension

Rajiv D Machado *, Oliver Eickelberg , Greg Elliott , Mark W Geraci §, Masayuki Hanaoka , James E Loyd , John H Newman , John A Phillips III #, Florent Soubrier **, Richard C Trembath ††, Wendy K Chung ‡‡
PMCID: PMC3725550  NIHMSID: NIHMS154654  PMID: 19555857

Abstract

Pulmonary arterial hypertension (PAH) is a rare disorder that may be hereditable (HPAH), idiopathic (IPAH), or associated with either drug-toxin exposures or other medical conditions. Familial cases have long been recognised and are usually due to mutations in Bone Morphogenetic Protein Receptor type 2 gene (BMPR2), or, much less commonly, 2 other members of the transforming growth factor-beta superfamily, Activin-like Kinase-Type I (ALK1) and Endoglin (ENG), which are associated with hereditary hemorrhagic telangiectasia. In addition, approximately 20% of patients with IPAH carry mutations in BMPR2. We provide a summary of BMPR2 mutations associated with IPAH/HPAH, most of which are unique to each family and are presumed to result in loss of function. We review the finding of missense variants and variants of unknown significance in BMPR2 in IPAH/HPAH, fenfluramine exposure, and PAH associated with congenital heart disease. Clinical testing for BMPR2 mutations is available and may be offered to HPAH and IPAH patients but should be preceded by genetic counselling, since lifetime penetrance is only 10%–20%, and there are currently no known effective preventative measures. Identification of a familial mutation can be valuable in reproductive planning and identifying family members who are not mutation carriers and thus will not require lifelong surveillance. With advances in genomic technology and with international collaborative efforts, genome-wide association studies will be conducted to identify additional genes for HPAH, genetic modifiers for BMPR2 penetrance, and genetic susceptibility to IPAH. In addition, collaborative studies of BMPR2 mutation carriers should enable identification of environmental modifiers, biomarkers for disease development and progression, and surrogate markers for efficacy end points in clinical drug development, thereby providing an invaluable resource for trials of PAH prevention.

Keywords: Pulmonary Arterial Hypertension, Genetics, Genomics

Clinical and Molecular Genetic Features of Pulmonary Arterial Hypertension

Pulmonary arterial hypertension (PAH) is a rare disorder with an estimated incidence of approximately 2 cases per million per year (1,2). It is characterized by a sustained increase in mean pulmonary artery pressure (>25 mm Hg at rest or 30 mm Hg with exercise), normal pulmonary capillary wedge pressure, and increased pulmonary vascular resistance. For adults, mean age at presentation ranges from 36 to 50 years, although individuals of any age can be affected (2,3). Prior to the advent of modern therapies, life expectancy for adults with idiopathic PAH (IPAH) was <3 years from diagnosis; for children, it was <10 months (4).

Pulmonary arterial hypertension may be heritable (HPAH), idiopathic, or associated with drug or toxin exposures (fenfluramine derivatives or toxic oil syndrome), or other medical conditions, including connective tissue diseases, human immunodeficiency virus infection, congenital heart disease, and portal hypertension. Familial cases have long been recognized (5), and in 2000, Bone Morphogenetic Receptor Protein 2 (BMPR2) was identified following linkage analysis (6-8) as the gene responsible for more than 70% of HPAH and approximately 20% of IPAH cases (9-12). Crude indirect estimates of the population carrier frequency for BMPR2 mutations lie in the frequency range of 0.001 to 0.01% (13). Two further receptor members of the transforming growth factor-beta (TGF-β) cell signalling superfamily are also recognized as uncommon causes of HPAH. Heterozygous mutations in Activin-like Kinase-Type I (ALK1) (14) and Endoglin (ENG) (15) cause hereditary hemorrhagic telangiectasia (HHT) and may rarely lead directly to the development of PAH.

Heritable PAH is inherited as an autosomal dominant trait with incomplete penetrance and an estimated lifetime risk of 10%–20% (16). The disease is more frequent in women, with a ratio of at least 1.7:1 women to men (2,17,18). Both incomplete penetrance and the significantly skewed gender ratio suggest interactions between BMPR2 disease mutations and environmental exposures that may include hormones, together with a role for modifying genes. The latest classification scheme now replaces the term familial PAH with HPAH at least in part to recognize the fact that up to 20% 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 in the prospective National Institutes of Health registry (18). A family history of PAH may go unrecognized in IPAH cases with BMPR2 mutations, as a consequence of either incomplete penetrance or de novo (spontaneous) mutations. Heterozygous BMPR2 sequence variants have been identified in a small subset of patients with PAH associated with relatively brief exposure to fenfluramine (13,19) or with congenital heart disease (20), 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 been identified in PAH associated with the scleroderma-spectrum disease or with human immunodeficiency virus (21,22).

HPAH and IPAH have a similar clinical course. HPAH is associated with a slightly younger age of onset and a slightly more severe hemodynamic impairment at diagnosis, but with similar survival (23). Patients with PAH and disease-causing 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 (23-25).

Genetic Anticipation

Families with BMPR2 mutations have been reported to have genetic anticipation, or earlier age of diagnosis in subsequent generations (17). 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. The question of genetic anticipation can be better addressed in future registries in which all patients with HPAH and IPAH can be genetically characterized and unbiased family studies can be performed.

The TGF-β Family and PAH

The TGF-β 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. Displaying high evolutionary conservation across species, TGF-β members are segregated into several subfamilies, notably the prototypic TGF-β ligands, receptors, and accessory molecules, activins, and the largest of these groups, the bone morphogenetic proteins (26). The implication of BMPR2, ALK-1, and ENG 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 (27).

BMPR-II Structure and Signal Transduction

The 4 functional domains of TGF-β type II receptors are typically highly conserved across the family. They consist of an N-terminal ligand binding domain, a single transmembrane region, a serine/threonine kinase, and a cytoplasmic tail domain. Particular to BMPR-II, however, is the presence of an exceptionally long postkinase cytoplasmic domain, primarily encoded by exon 12 of the gene. A second isoform, generated by alternative splicing of exon 12, is expressed ubiquitously at the mRNA level, although the in vivo function of the mature polypeptide remains enigmatic (28).

Signal transduction routed through the TGF-β pathway has been extensively interrogated over the last 2 decades; in contrast, BMP signalling remains less well described. The paradigmatic BMP pathway is a phosphorylation relay of signalling intermediaries initiated at the cell surface and culminating in the nucleus (Figure 1) (29). A heterotetrameric complex consisting of type I receptors, for example ALK1, BMPR1A or BMPR1B, and BMPR-II, amalgamate to bind extracellular dimeric ligand. These interactions promote close receptor species proximity and activation of the type I receptor by the constitutive kinase BMPR-II. The type I receptors, in turn, bind and phosphorylate members of the receptor R Smad family, namely SMAD1/5 or 8. When activated, the affinity of the R-Smads for a nuclear chaperone, Smad-4, common across the TGF-β system, is augmented. These complex-bound signalling molecules shuttle to the nucleus, where, in concert with specific nuclear cofactors and repressors, they influence regulation of a limited set of target genes (26). Within the framework of an essentially uncomplicated canonical pathway, the specificity and flexibility of signalling outcomes is tightly maintained by the myriad, cell-specific levels of regulation extending from sequestration of extracellular ligand to inhibitive competition of type I receptors by nonfunctional decoy receptors and extensive cross-talk with other signalling pathways including p38MAPK, the cytoskeleton-associated proteins Tctex-1 and LIMK-1 ( 26,30-32), and c-src and RACK-1 (33,34).

Figure 1.

Figure 1

Upon binding ligand, the type II receptor phosphorylates a type-I partner, which may be BMPR1A/B or upon activation by the ligands BMP9/10, ALK-1. This leads to a propagation of cytoplasmic signal transduction through the R-Smads 1/5/8, or, independently, via the p38MAPKs. In association with Smad4 the R-Smads translocate to the nucleus and, in complex with transcriptional cofactors, modulate the expression of a target set of genes.

The burgeoning understanding of TGF-β receptor structure and function has been an invaluable aid in determining the likely impact of BMPR2 mutations in PAH. Conversely, and of equivalent importance, analysis of the spectrum and pattern of these defects, in conjunction with directed functional studies, has confirmed and better resolved those regions of the receptor necessary for normal physiologic activity.

Major Genetic Risk Factors in PAH

Through the compilation of previously reported mutation data, combined with an ongoing global collaboration targeted at conducting systematic and comprehensive screening of patient samples, a combined total of 298 BMPR2 mutations have been identified (35). Of these, 88 are novel (Table 1) (36-39). These mutations were identified in independent 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 risen on unrelated genetic backgrounds, suggesting that genetic founder events are uncommon (35). Indeed, approximately 41% (122/298) of all recorded BMPR2 mutations are small deletions or insertions at sites of low complexity sequence or as a consequence of C>T transitions, presumably resulting from the relatively common process of spontaneous cytosine deamination. Thus, despite the fact that PAH typically presents as a late-onset disease, ancestral mutations in this gene are at best rare, possibly because of a deleterious effect on reproductive fitness. In addition, we describe uncommon genetic susceptibilities, including ALK-1 defects in PAH and PAH associated with HHT.

Table 1.

Pathogenic BMPR2 mutations in PAH

Location Mutation
category
Domain Nucleotide
change
Amino Acid
change
Frequency in
this study
Comment Reference
5′UTR Missense c.*-944/5GC>AT 1 H Aldred et al (36)
5′UTR and
Exon 1
Deletion c.?_-540_76+?del 1 H This analysis
Exon 1 Missense c.28C>T p.T10W 1 I Baloira et al (37)
Exon 1 Nonsense c.71C>A p.A24E 1 I This analysis
Intron 1 Splice-site c.76+5G>A 1 I This analysis
Exon 2 Nonsense ECD c.124C>T p.Q42X 1 I Fujiwara et al (38)
Exon 2 Frameshift ECD c.186insTACC p.G63fsX1 1 H This analysis
Exon 2 Frameshift ECD c.189_207delins14 p.S64EfsX32 1 ASD / PAH This analysis
Exon 2 Deletion ECD c.189-209del21 p.del6470(STCYGLW) 1 H This analysis
Exon 2 Nonsense ECD c.201T>G p.Y67X 1 H This analysis
Exon 2 Missense ECD c.247G>A p.G83R 1 I This analysis
Exons 1-4 Deletion ECD c.1-?_419+?del 1 I This analysis
Exons 1-8 Deletion ECD c.1-?_1128+?del 1 I This analysis
Intron 2 Splice-site ECD c.247+1delCAAGTG 1 H This analysis
Intron 2 Splice-site ECD c.247+1_+4 delGTAA p.C84_S140del 1 I This analysis
Intron 2 Splice-site ECD c.248-2A>G 1 H This analysis
Intron 2 Splice-site ECD c. 248-5 delTATAGGinsAC 1 H This analysis
Exon 3 Missense ECD c.248G>A p.G83E 1 H This analysis
Exon 3 Missense ECD c.250T>C p.C84R 1 H This analysis
Exon 3 Missense ECD c.250T>G p.C84G 1 I This analysis
Exon 3 Frameshift ECD c.261insA p.87fsX9 1 H This analysis
Exon 3 Missense ECD c.295T>C p.C99R 1 H This analysis
Exon 3 Missense ECD c.296G>T p.C99F 1 I This analysis
Exon 3 Missense ECD c.296G>A p.C99Y 1 H This analysis
Exon 3 Nonsense ECD c.339C>A p.Y113X 1 I This analysis
Exon 3 Nonsense ECD c.339C>G p.Y113X 1 I Fujiwara et al (38)
Exon 3 Missense ECD c.350G>A p.C117Y 1 H This analysis
Exon 3 Nonsense ECD c.354-355TA>AG p.C118X 1 H This analysis
Exon 3 Missense ECD c.367T>C p.C123R 1 I Fujiwara et al (38)
Exon 3 Missense ECD c.370A>G p.N124D 1 I This analysis
Exon 3 Missense ECD c.377A>G p.N126S 1 I This analysis
Exon 2-3 Deletion ECD c.77-?_418+?del 5 H,H,H,H,I This analysis
Exon 3 Deletion ECD c.248-?_418+?del 2 H,H This analysis
Intron 3 Splice-site ECD c.418+1G>C 1 H This analysis
Intron 3 Splice-site ECD c.418+5G>A 2 H This analysis
Exons 4-5 Deletion ECD c.419-?_621+?del 2 H,I This analysis
Exon 5 Nonsense TM c.583G>T p.E195X 1 PVOD This analysis
Exon 5 Missense TM c.604A>T p.N202Y 1 PVOD This analysis
Exon 6 Frameshift KD c.612delA p.L204fsX5 1 H This analysis
Exon 6 Nonsense KD c.631C>T p.R211X 2 H This analysis
Exon 6 Frameshift KD c.660insG p.G220fsX4 1 NK This analysis
Exon 6 Frameshift KD c.690-691delAGinsT p.K239fsX21 1 H This analysis
Exon 6 Missense KD c.794A>G p.E265G 1 I This analysis
Exon 6 Missense KD c.797G>C p.R266T 1 NK This analysis
Exon 6 Missense KD c.806G>T p.A268V 1 I Baloira et al (37)
Intron 6 Splice-site KD c.852+1G>C 1 I This analysis
Intron 6 Splice-site KD c.853-1G>C 1 H This analysis
Exon 7 Nonsense KD c.928A>T p.R310X 1 I This analysis
Exon 7 Missense KD c.932G>A p.G311E 1 H This analysis
Exon 8 Missense KD c.1019T>C p.L340P 1 H This analysis
Exon 8 Nonsense KD c.1189-1190delTG p.C347X 1 H This analysis
Exon 8 Missense KD c.1042G>A p.V348E 1 H This analysis
Exon 8 Frameshift KD c.1095delC p.R365fsX8 1 H This analysis
Exon 8 Frameshift KD c.1099-1103delGGGGA p.E368fsX1 1 H This analysis
Exon 8 Deletion KD c.968-?_1129+?del 1 H This analysis
Intron 8 Splice-site KD c.968-5A>G 1 H This analysis
Exon 9 Missense KD c.1157A>T p.E386V 1 H Fu et al (39)
Exon 9 Missense KD c.1171G>A p.A391T 1 I This analysis
Exon 9 Nonsense KD c.1207C>T p.Q403X 1 H Fujiwara et al (38)
Exon 9 Frameshift KD c.1214delA p.D405fsX6 1 H This analysis
Exon 9 Nonsense KD c.1248-1251delATTT p.F417X 1 H This analysis
Exon 9 Frameshift KD c.1271delTCCCAGinsCGGAGA p.F424fsX10 1 H This analysis
Intron 9 Splice-site KD c.1276+1G>A 1 H This analysis
Intron 9 Splice-site KD c.1277-9A>G 1 H This analysis
Exon 10 Nonsense KD c.1297C>T p.Q433X 2 I,NK This analysis
Exon 10 Frameshift KD c.1392delA p.A465fsX9 1 H This analysis
Exon 11 Missense KD c.1460A>T p.D487V 1 H This analysis
Exon 11 Missense KD c.1472G>A p.R491Q 1 I This analysis
Exon 12 Missense CD c.1598A>G p.H533R 1 I This analysis
Exon 12 Frameshift CD c.2410-2413delGTCA p.V804fsX1 1 H This analysis
Exon 12 Frameshift CD c.2441-2442delAC p.H814fsX2 1 H This analysis
Exon 12 Frameshift CD c.2504insA p.T835fsX6 1 PAH+collagen
disease
This analysis
Exon 12 Frameshift CD c.2609del T p.L870fsX1 1 I This analysis
Exon 12 Nonsense CD c.2626C>T p.Q876X 1 H This analysis
Exon 12 Nonsense CD c.2695C>T p.R899X 2 I This analysis
Exon 12 Nonsense CD c.2789C>G p.S930X 2 I This analysis
Exon 13 Missense CD c.2945A>G p.L982R 1 H This analysis

Numbering is based on +1 of the initiation methionine.

UTR = untranslated region; ECD = extracellular domain; TM = transmembrane; KD = kinase domain; CD = cytoplasmic domain; F = familial ; I = idiopathic; NK = not known; PVOD = pulmonary veno-occlusive disease; ASD = atrial-septal defect.

Mutation in BMPR2 Constitutes the Primary Genetic Risk in PAH

A wide range of mutation detection methodologies have been employed to screen patients for point mutations and large gene rearrangements, among them direct sequencing, melting curve analysis, denaturing high-performance liquid chromatography, Southern blotting, and multiplex ligation-dependent probe amplification. To date, mutations have been recorded in over 70% of subjects with 1 or more affected relatives, while in idiopathic cases, mutation detection rates between centers range from 10%–40% (12,19,36,40). Of interest, mutations of TGF-β superfamily members have also been observed in associated forms of PAH, albeit at far lower frequencies (35). The spectrum of BMPR2 mutation in hereditary and spontaneous disease is analogous, comprising all major classes of mutations. Mutations predicting premature truncation of the polypeptide chain are the most prevalent (203/298, 68%). Although there is another putative PAH locus at 2q32 that was mapped in part by using stress echocardiography to detect asymptomatic obligate carriers (41), no gene has yet been identified from this interval. Thus, to date, mutations in BMPR2 remain the primary genetic susceptibility for PAH.

Truncating Mutations of BMPR2 in Heritable and Idiopathic PAH

Mutations predicted to introduce premature truncation codons to the BMPR2 open reading frame encompass nonsense (85/298, 29%), frame-shift (73/298, 24%), splice-site (26/298; 9%), and gene duplications/deletions (19/298, 6%). An exception of note is a double substitution of 2 consecutive bases (GC>AT) identified in a large PAH kindred 944 base pairs upstream of the translation start site (36). Bioinformatic assessment indicated that the mutation, within the context of flanking sequence, would likely generate the formation of an aberrant translational start signal, consequently leading to incorporation of a premature stop codon in the first exon of the gene. Allele-specific polymerase chain reaction assays utilising variant and cell-based studies to examine activation of the nonsense-mediated decay (NMD) surveillance machinery have confirmed both degradation and loss of the mutant harbouring transcript. Indeed, both conventional studies on patient cell lines and novel in vitro technologies demonstrate that the majority of mutations in this class are rapidly removed from the cell via the NMD pathway (36,42). Along with the observation of large proximal and whole deletions of BMPR2, these findings now firmly substantiate haploinsufficiency as the predominant molecular mechanism underlying BMPR2 predisposition to hereditary and idiopathic forms of disease (11,13,43).

Distribution and Impact of Missense Mutations Across Conserved Functional Domains of BMPR2

Missense mutations of BMPR2 in HPAH/IPAH cluster mainly in regions of the gene-encoding receptor domains indispensable to signalling activity and are confined to exons 2, 3, 6-9, 11, and 12. The extracellular ligand-binding domain of BMPR-II adopts a precisely folded conformation, exquisitely dependent on the formation of 5 disulphide bridges by 10 cysteine residues dispersed across exons 2 and 3, invariant in the majority of type II receptors (44). Amino acid substitutions in this domain are common, particularly at the cysteine residues, with 17/22 independent cysteine mutations affecting 8 of the 10 conserved residues. Subcellular analysis of all tested cysteine mutant constructs demonstrates substantial cytosolic retention, likely to be due to a profound loss of conformational integrity (45,46).

The BMPR-II catalytic domain shares the fundamental structural and functional characteristics of members of the extensive eukaryotic protein kinase superfamily. The kinase region is compartmentalized into 12 subdomains of variable importance to the processes of adenosine triphosphate binding, substrate recognition, and phosphate group transfer (47). However, dispersed across the subdomains are 12 critical, highly conserved, amino acid residues. Missense mutations are typically restricted to regions crucial to kinase activity, as best exemplified by substitution of the native arginine residue at the invariant position 491, a site of frequent and recurrent mutation in PAH. Disruption of the key structural interaction between this residue and a glutamic acid at position 386 effectively renders the receptor kinase inactive. While noncysteine substitutions traffic normally to the cell surface, all kinase mutations occurring in these catalytically important domains display a near complete abolition of signalling through the Smad pathway (45,46).

Variants of Unknown Significance in PAH

Amino acid substitutions are seldom observed in the cytoplasmic domain of BMPR-II (7/298; 2%), in contrast to truncating mutations, which account for approximately 15% of the overall mutation load in PAH. Furthermore, missense mutations in this region are functionally distinct from classical PAH-causing defects, as they are associated with normal levels of Smad activation (45,46). Instead, in transient transfection-based assays, disease-specific substitutions of the cytoplasmic tail appear to constitutively activate p38MAPK and impede phosphorylation of the dynein light chain Tctex-1 (32,46). Thus, this domain of the receptor is significant in relaying receptor signal through Smad-independent pathways. The relevance of this finding to disease pathogenesis is currently under investigation.

The mutation spectrum in PAH associated with other conditions or acknowledged risk factors, for example, fenfluramine use, is distinct from the mutation spectrum in hereditary/idiopathic disease (35). PAH patients with fenfluramine exposure exclusively harbor missense mutations, in comparison to only one third of patients with HPAH. Moreover, the substitutions occur at positions not recognized as having a major impact on receptor function. For example, none of the 5 reported variants in the extracellular domain observed in patients with PAH and congenital heart disease or associated with appetite suppressant intake alter cysteine residues, and 2 residues altered in exon 5 encode a receptor region of no known functional significance (13,20,47). Significantly, a recent in vitro study of this mutation series has indicated that the signaling capacity of receptors harboring atypical variation approaches physiologic levels (42). Akin to the cytoplasmic tail substitutions, the relationship of these variants to the etiology of PAH is difficult to assess. A possible interpretation may be that these alleles produce PAH in the setting of additional genetic or environmental risk factors.

Rare Disease Alleles Underlying PAH

HHT is an autosomal dominant vascular disorder characterized by the appearance of cutaneous telangiectasias and arteriovenous malformations. The disease is caused by pathogenic mutations of either the TGF-β type I receptor ALK-I or accessory receptor endoglin (ENG) (Table 2A) (15,27,48,49), and, rarely, Smad 4, which is also associated with juvenile polyposis. A small proportion of HHT patients have PAH that is clinically and histopathologically indistinguishable from other heritable forms of PAH, while others have PAH due to pulmonary arteriovenous fistulas (27). 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 (50,51). The majority of these defects comprise missense mutations and cluster in functional domains of the receptor, namely the kinase domain and NANDOR box. In rare instances mutations of ALK-1 (n=9) appear to cause IPAH or HPAH without HHT (Table 2B) (27,38,48). Of interest, 4 mutations are confined to 2 discrete positions (amino acids 479 and 484) of the kinase domain, suggesting that the native residues, both arginines, may be important for maintenance of the pulmonary architecture (45). However, an important caveat is that these patients usually present with early-onset disease and may go on to develop HHT at a later stage (38,48).

Table 2A.

ENG mutations underlying PAH

Location Domain PAH classification Nucleotide
change
Amino acid
change
Reference
Exon 5 ECD PAH+HHT c.682-686del p.S228fsX102 Harrison et al (27)
Exon 10 ECD PAH+HHT c.1334delT p.M445fsX44 Harrison et al (27)
Exon 11 ECD PAH+HHT+dexfenfluramine c.del410G p.Q470fsX20 Chaouat et al (15)
Intron 12 ECD PAH+HHT c.1429-
22T>C
Harrison et al (48)
Mache et al (49)

Table 2B.

ALK-1 mutations underlying PAH

Location Domain PAH
classification
Nucleotide
change
Amino acid
change
Age at
onset
Reference
Exon 2 ECD IPAH c.430C>T p.R144X NK This analysis
Exon 5 ECD IPAH c.536A>C p.D179A 51 years Harrison et al (27)
Exon 7 KD HPAH c.936C>G p.H312Q 14 years Fujiwara et al (38)
Exon 8 KD IPAH c.1142T>C p.L381P 9 years Fujiwara et al(38)
Exon 9 KD HPAH c.1270C>A p.P424T 7 years Fujiwara et al(38)
Exon 10 KD IPAH c.1436G>A p.R479Q 7 years Fujiwara et al (38)
Exon 10 KD IPAH c.1436G>C p.R479P NK This analysis
Exon 10 KD IPAH c.1451G>A p.R484Q 18 months Harrison et al (27)
Exon 10 KD HPAH c.1451G>A p.R484Q 2 years Fujiwara et al (38)

Numbering is based on +1 of the initiation methionine of ALK1.

ECD = extracellular domain; KD = kinase domain.

Genetic Testing for PAH

Clinical genetic testing is available for PAH for BMPR2, ALK1, and ENG. In most cases, genetic analysis will commence with analysis of BMPR2 unless there are specific clinical symptoms or family history to suggest HHT, such as mucocutaneous telangiectasias, recurrent epistaxis, gastrointestinal bleeding, or arteriovenous malformations in the pulmonary, hepatic, gastrointestinal, or cerebral circulations. Genetic testing may be offered to any individual with a family history of PAH or IPAH (without other known affected family members), and physicians may have a duty to inform these patients of the possibility that PAH could develop in other family members. It may be necessary to go back to stored blood or deoxyribonucleic acid (DNA) if the only affected family member is deceased and if those materials are available for testing.

Evaluation of BMPR2 should begin with full sequencing of all 13 exons including splice junctions. If no mutation is identified by sequence analysis, further characterization for genomic deletions and rearrangements should be evaluated by an appropriate methodology, such as multiplex ligation-dependent probe amplification. Using these combined approaches, approximately 70% of HPAH patients will have identified mutations in BMPR2 (36). Clinical genetic testing is available in North America and Europe, with the current cost of testing ranging from approximately US $1000–$3000 to analyze the first member of a family. Testing other family members for a family-specific mutation is US $300–$500. Genetic testing should involve pretest and post-test genetic counseling, ideally with a genetic counselor experienced in pulmonary hypertension. 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 serial evaluation for detection of PAH.

The most common reasons that persons pursue genetic testing are to inform their children of their hereditary predisposition or to make informed decisions about family planning (52). In the past, many patients opted not to pursue genetic testing because of anxiety regarding genetic discrimination. Recognition of these concerns has led a number of countries to introduce either voluntary or legal codes to protect individuals requesting genetic counseling and formal testing. For example, in the United States, the Genetic Information Nondiscrimination Act, passed in May 2008, protects members of both individual and group health insurance plans from discrimination in coverage or cost of health insurance coverage and also protects against discrimination in employment based upon a genetic predisposition (53). Genetic testing of children should be performed with caution, because of the potentially significant psychological impact on a child, particularly overt anxiety for the future development of a potentially fatal disease in the absence of currently known effective disease-prevention strategies.

Clinical Monitoring of Individuals at Risk

Clinical monitoring of patients with a family history of PAH or carriers of the BMPR2 mutation has not been evaluated rigorously. Consideration has been given to annual clinical examination, echocardiogram, stress echocardiography, Doppler echocardiography during supine bicycle exercise, and right heart catheterization at rest and with exercise. The 1998 World Pulmonary Hypertension Conference suggested that first-degree relatives of known HPAH patients should be screened annually using clinical examination and echocardiography. It is hoped that with regular surveillance, individuals can be diagnosed earlier in their disease and benefit from early treatment. Although there are currently no data to suggest that early diagnosis will improve the outcome, such studies are in progress (54,55).

REPRODUCTIVE PLANNING

Prenatal testing is available for BMPR2 mutations if a familial mutation has been identified. For a prospective parent carrying a known BMPR2 mutation, there is a 50% risk of transmitting this mutation to any offspring (56). Prenatal testing can be performed by chorionic villus sampling as early as the 10th week of pregnancy. However, partly in consequence of the reduced penetrance of PAH disease mutations, in our experience very few families have pursued this option. Additionally, preimplantation genetic diagnosis is available, whereby families use in vitro fertilization, and genetic testing is performed on the embryo prior to embryo transfer. Only embryos without the familial BMPR2 mutation are transferred to the uterus. This option may prove more appealing, particularly to women who are carriers of the BMPR2 mutation and have been advised not to pursue a pregnancy because of the increased risk of developing PAH symptoms themselves. In such cases, in vitro fertilization is already required for surrogacy. However, preimplantation genetic diagnosis with in vitro fertilization is costly, may not be covered by insurance in the US, and is not available in all European countries. Preimplantation genetic diagnosis has some associated diagnostic errors (57).

Identifying Novel, Highly Penetrant Genes for HPAH

A small percentage of HPAH families have multiple affected individuals but do not have identified mutations in BMPR2, despite full sequencing of exons and splice junctions and testing for genomic alterations (9-12). Although many of these families have limited numbers of living affected family members available for research, HPAH in these families may be due to locus or allelic heterogeneity that could be identified by sequencing of candidate genes, or in rare cases, analyzed by linkage if sufficient numbers of affected family members are available. As our ability to cost effectively sequence and interpret greater amounts of DNA grows, we should be able to analyze the BMPR2 mutation-negative HPAH cases for mutations in the whole BMPR2 genomic locus and for other physiologic candidate genes.

Genetic Modifiers of Risk for PAH

The complex clinical features of PAH, including variable age of disease onset both within and between families and sex-dependent penetrance, imply the existence of additional factors capable of modulating disease susceptibility (58). Moreover, the likely existence of environmental modifiers is highlighted by our observation of at least 7 pairs of monozygotic twins discordant for disease (Trembath RC, personal communication). To date, a number of studies examining the contribution of variations in candidate genes considered to play a biological role in the etiology of PAH have been conducted (40,59-68). However, all the case/control analyses suffer from critically small sample cohorts that would be unlikely to identify genetic modifiers of moderate effect size. Further, validation through independent replication studies, a crucial second stage of association analyses, has been lacking in most of the studies performed to date (Table 3) (35,59-64,66-68).

Table 3.

Candidate gene association studies conducted in PAH

Ge
ne
PAH classification Patient
sample
(n)
Control
sample
(n)
Association
(Patient/control
freq.)
Significance
(p value)
Reference
ACE Not specified 60 158 DD genotype
(0.45/0.28)
(0.01) Abraham et al(59)
Not specified 51 200 No association N/A Hoeper et al (63)
Hypoxic 48 30 I allele (0.67/0.38) 0.003 Aldashev et al(60)
5-HTT PAH +
app.suppressants
89 84 II genotype
(0.65/0.27)
<0.001 Eddahibi et al (62)
IPAH/CTEPH 74/35 Unknown No association N/A Koehler et al (64)
HPAH/IPAH/APAH 133/259/136 253 No association N/A Machado et al (35)
HPAH/IPAH 166/83 125 II genotype with
early onset in
FPAH
<0.02 Willers et al (67)
Endoglin PAH+systemic
sclerosis
23 140 6bINS (0.11/0.24) <0.01 Wipff et al (68)
PGIS Chronic
thromboembolic
PH
90 144 No association N/A Amano et al (61)
Kv1.5 IPAH NO
Respond
ers
42
NO non-
responder
s
37
SNP4 a allele;
SNP17 a allele
(0.07/0.23;
0.016/0.06)
0.01/0.05 Remillard et al (66)

ACE = angiotensin converting enzyme; 5-HTT = 5-hydroxy-tryptamine (serotonin) transporter; PGIS = prostacyclin synthase gene; Kv1.5 = potassium channel subunit 1.5; NO = nitric oxide; I = insertion; D = deletion.

Genome-wide Association Analysis in IPAH

Driven by the HapMap project (69) and the development of genotyping platforms facilitating analysis of several hundred thousand independent loci, an understanding of the human haplotypic architecture of sequence variation and the identification of loci conferring even modest risks for disease are now feasible. To achieve the power required to conduct a genome-wide association (GWA) study and to detect signals of modest genotypic relative risk (GRR), large numbers of cases and controls and international multicenter collaboration will be required (70,71).

GWA Study Design in IPAH

A staged approach will be taken, using a common genotyping platform and concurrent screens. This will provide a dataset of >1000 cases and ≤3000 controls. All patients will be screened for mutations in BMPR2 and ALK-1, and mutation carriers will be removed from the analysis. Data from the 2 screens may be combined in a meta-analysis (72). Based on power calculation simulations, these numbers are considered sufficient to detect variants of moderate effect size (GRR 1.3). The second stage of the study will be independent replication by a GWA performed on a combined cohort of North American IPAH cases and controls. Again, the data sets will be combined in a meta-analysis, and loci displaying significant association across the 3 studies will be chosen for further interrogation. In this way, the false-positive report rate is anticipated to be limited and the likelihood of true associations with IPAH increased (73).

Publicly available data from the HapMap consortium on the extent of linkage disequilibrium in regions displaying positive signals will be used to fine-map associated loci and establish a panel of tagging single nucleotide polymorphisms. Where strong association is identified with low frequency haplotypes, candidate genes will be resequenced to capture rare, potentially causal variants. All potentially functional single nucleotide polymorphisms will subsequently be analyzed in all patient and control cohorts. A range of well established statistical tools, including conditional logistic regression, will be used to identify those variants most likely to be true susceptibility alleles.

Modifiers of BMPR2 Mutation in Heritable Disease

The major PAH centers worldwide have access to at least 250 kindreds with identified BMPR2 mutations. Risk alleles from the IPAH GWA study will be assessed in affected probands from these families by comparison to age- and sex-matched unaffected mutation carriers to determine whether, in the framework of BMPR2 mutation, they act as modifiers in familial disease as well as IPAH.

Future Research

Identification of a substantial number of genetically at-risk individuals offers the potential to develop an extremely valuable resource for future studies, including assessment of the natural history of PAH, development of biomarkers for disease onset and progression, the identification of environmental and genetic modifiers, and the opportunity to test methods for primary prevention. Biomarker development will benefit from advances in expression profiling, proteomics, and metabolomics, by longitudinally testing individuals at increased genetic risk. Interestingly, the largest number of genetically at-risk individuals is within families of patients currently diagnosed with IPAH, given the relatively higher prevalence of this form of PAH. Through clinical genetic testing programs or by enrollment in research programs, such kindreds will provide an invaluable resource for studying PAH and potentially identifying preventive measures. Clearly these types of ambitious studies will require large numbers of patients as well as collaboration among multiple centers around the world. However, such a cohort of asymptomatic BMPR2 mutation carriers is likely to provide one of the most powerful resources for an in-depth understanding of PAH pathogenesis and prospects for prevention.

Acknowledgements

This manuscript is dedicated to the memory of Jane H. Morse, MD, to acknowledge her invaluable contribution to the genetics of PH. The authors would like to thank all the patients and families who have generously contributed to this research, and Josue Martinez and Grace Lee for assistance with manuscript preparation.

Funding provided by NHLBI 060056, Deseret Foundation, Intermountain Medical Center, NIH PO1 HL072058, GCRC RR000095, German Research Foundation (DFG) Collaborative Research Center 547 (O.E.), the Excellence cluster “Cardiopulmonary System” (ECCPS), European Commission under the 6th Framework Programme (Contract No: LSHM-CT-2005-018725, PULMOTENSION), and BHF - FS/07/036.

Abbreviations and Acronyms

ALK1

activin-like kinase type I

BMPR2

bone morphogenetic protein receptor type 2 gene

DHPLC

denaturing high-performance liquid chromatography

DNA

deoxyribonucleic acid

ENG

endoglin

EUK

eukaryotic protein kinase

GRR

genotypic relative risk

GWA

genome-wide association

HHT

hereditary hemorrhagic telangiectasia

HPAH

heritable PAH

IPAH

idiopathic pulmonary arterial hypertension

MLPA

multiplex ligation-dependent probe amplification

NMD

nonsense-mediated decay

PAH

pulmonary arterial hypertension

PTC

premature truncation codon

TGF-β

transforming growth factor-beta

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest Dr. Elliott is employed by Intermountain Healthcare. Intermountain Healthcare has filed a patent based on Dr. Elliott’s invention for the use of BMPR2 mutation analysis to assess vasoreactivity in pulmonary arterial hypertension.

The other authors have reported that they have no conflicts of interest to declare.

References

  • 1.Gaine SP, Rubin LJ. Primary pulmonary hypertension. Lancet. 1998;352:719–25. doi: 10.1016/S0140-6736(98)02111-4. [DOI] [PubMed] [Google Scholar]
  • 2.Humbert M, Sitbon O, Chaouat A, et al. Pulmonary arterial hypertension in France: results from a national registry. Am J Respir Crit Care Med. 2006;173:1023–30. doi: 10.1164/rccm.200510-1668OC. [DOI] [PubMed] [Google Scholar]
  • 3.D’Alonzo GE, Barst RJ, Ayres SM, et al. Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. ann Intern Med. 1991;115:343–9. doi: 10.7326/0003-4819-115-5-343. [DOI] [PubMed] [Google Scholar]
  • 4.Runo JR, Loyd JE. Primary pulmonary hypertension. Lancet. 2003;361:1533–44. doi: 10.1016/S0140-6736(03)13167-4. [DOI] [PubMed] [Google Scholar]
  • 5.Dresdale DT, Michtom RJ, Schultz M. Recent studies in primary pulmonary hypertension including pharmacodynamic observations on pulmonary vascular resistance. Bull N Y Acad Med. 1954;30:195–207. [PMC free article] [PubMed] [Google Scholar]
  • 6.Deng Z, Morse JH, Slager SL, et al. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Genet. 2000;67:737–44. doi: 10.1086/303059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lane KB, Machado RD, Pauciulo MW, et al. Heterozygous germline mutations in BMPR2, encoding a TGF-β receptor, cause familial primary pulmonary hypertension. The International PPH Consortium. Nat Genet. 2000;26:81–4. doi: 10.1038/79226. [DOI] [PubMed] [Google Scholar]
  • 8.Newman JH, Wheeler L, Lane KB, et al. Mutation in the gene for bone morphogenetic protein receptor II as a cause of primary pulmonary hypertension in a large kindred. N Engl J Med. 2001;345:319–24. doi: 10.1056/NEJM200108023450502. [DOI] [PubMed] [Google Scholar]
  • 9.Aldred MA, Vijayakrishnan J, James V, et al. BMPR2 gene rearrangements account for a significant proportion of mutations in familial and idiopathic pulmonary arterial hypertension. Hum Mutat. 2006;27:212–3. doi: 10.1002/humu.9398. [DOI] [PubMed] [Google Scholar]
  • 10.Cogan JD, Vnencak-Jones CL, Phillips JA, 3rd, et al. Gross BMPR2 gene rearrangements constitute a new cause for primary pulmonary hypertension. Genet Med. 2005;7:169–74. doi: 10.1097/01.gim.0000156525.09595.e9. [DOI] [PubMed] [Google Scholar]
  • 11.Cogan JD, Pauciulo MW, Batchman AP, et al. High frequency of BMPR2 exonic deletions/duplications in familial pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;174:590–8. doi: 10.1164/rccm.200602-165OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Thomson JR, Machado RD, Pauciulo MW, et al. Sporadic primary pulmonary hypertension is associated with germline mutations of the gene encoding BMPR-II, a receptor member of the TGF-β family. J Med Genet. 2000;37:741–5. doi: 10.1136/jmg.37.10.741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Humbert M, Deng Z, Simonneau G, et al. BMPR2 germline mutations in pulmonary hypertension associated with fenfluramine derivatives. Eur Respir J. 2002;20:518–23. doi: 10.1183/09031936.02.01762002. [DOI] [PubMed] [Google Scholar]
  • 14.Trembath RC, Thomson JR, Machado RD, et al. Clinical and molecular genetic features of pulmonary hypertension in patients with hereditary hemorrhagic telangiectasia. N Engl J Med. 2001;345:325–34. doi: 10.1056/NEJM200108023450503. [DOI] [PubMed] [Google Scholar]
  • 15.Chaouat A, Coulet F, Favre C, et al. Endoglin germline mutation in a patient with hereditary haemorrhagic telangiectasia and dexfenfluramine associated pulmonary arterial hypertension. Thorax. 2004;59:446–8. doi: 10.1136/thx.2003.11890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sztrymf B, Yäici A, Girerd B, Humbert M. Genes and pulmonary arterial hypertension. Respiration. 2007;74:123–32. doi: 10.1159/000098818. [DOI] [PubMed] [Google Scholar]
  • 17.Loyd JE, Butler MG, Foroud TM, Conneally PM, Phillips JA, 3rd, Newman JH. Genetic anticipation and abnormal gender ratio at birth in familial primary pulmonary hypertension. Am J Respir Crit Care Med. 1995;152:93–7. doi: 10.1164/ajrccm.152.1.7599869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rich S, Dantzker DR, Ayres SM, et al. Primary pulmonary hypertension: a national prospective study. Ann Intern Med. 1987;107:216–23. doi: 10.7326/0003-4819-107-2-216. [DOI] [PubMed] [Google Scholar]
  • 19.Souza R, Humbert M, Sztrymf B, et al. Pulmonary arterial hypertension associated with fenfluramine exposure: report of 109 cases. Eur Respir J. 2008;31:343–8. doi: 10.1183/09031936.00104807. [DOI] [PubMed] [Google Scholar]
  • 20.Roberts KE, McElroy JJ, Wong WP, et al. BMPR2 mutations in pulmonary arterial hypertension with congenital heart disease. Eur Respir J. 2004;24:371–4. doi: 10.1183/09031936.04.00018604. [DOI] [PubMed] [Google Scholar]
  • 21.Nunes H, Humbert M, Sitbon O, et al. Prognostic factors for survival in human immunodeficiency virus-associated pulmonary arterial hypertension. Am J Respir Crit Care Med. 2003;167:1433–9. doi: 10.1164/rccm.200204-330OC. [DOI] [PubMed] [Google Scholar]
  • 22.Tew MB, Arnett FC, Reveille JD, Tan FK. Mutations of bone morphogenetic protein receptor type II are not found in patients with pulmonary hypertension and underlying connective tissue diseases. Arthritis Rheum. 2002;46:2829–30. doi: 10.1002/art.10487. [DOI] [PubMed] [Google Scholar]
  • 23.Sztrymf B, Coulet F, Girerd B, et al. Clinical outcomes of pulmonary arterial hypertension in carriers of BMPR2 mutation. Am J Respir Crit Care Med. 2008;177:1377–83. doi: 10.1164/rccm.200712-1807OC. [DOI] [PubMed] [Google Scholar]
  • 24.Elliott CG, Glissmeyer EW, Havlena GT, et al. Relationship of BMPR2 mutations to vasoreactivity in pulmonary arterial hypertension. Circulation. 2006;113:2509–15. doi: 10.1161/CIRCULATIONAHA.105.601930. [DOI] [PubMed] [Google Scholar]
  • 25.Rosenzweig EB, Morse JH, Knowles JA, et al. Clinical implications of determining BMPR2 mutation status in a large cohort of children and adults with pulmonary arterial hypertension. J Heart Lung Transplant. 2008;27:668–74. doi: 10.1016/j.healun.2008.02.009. [DOI] [PubMed] [Google Scholar]
  • 26.Shi Y, Massagué J. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell. 2003;113:685–700. doi: 10.1016/s0092-8674(03)00432-x. [DOI] [PubMed] [Google Scholar]
  • 27.Harrison RE, Flanagan JA, Sankelo M, et al. Molecular and functional analysis identifies ALK-1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic telangiectasia. J Med Genet. 2003;40:865–71. doi: 10.1136/jmg.40.12.865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Liu F, Ventura F, Doody J, Massagué J. Human type II receptor for bone morphogenic proteins (BMPs): extension of the two-kinase receptor model to the BMPs. Mol Cell Biol. 1995 Jul;15(7):3479–86. doi: 10.1128/mcb.15.7.3479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Machado RD, Aldred MA, James V, et al. Mutations of the TGF-β type II receptor BMPR2 in pulmonary arterial hypertension. Hum Mutat. 2006;27:121–32. doi: 10.1002/humu.20285. [DOI] [PubMed] [Google Scholar]
  • 30.Adachi-Yamada T, Nakamura M, Irie K, et al. p38 mitogen-activated protein kinase can be involved in transforming growth factor β superfamily signal transduction in Drosophila wing morphogenesis. Mol Cell Biol. 1999;19:2322–9. doi: 10.1128/mcb.19.3.2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Foletta VC, Lim MA, Soosairaiah J, et al. Direct signaling by the BMP type II receptor via the cytoskeletal regulator LIMK1. J Cell Biol. 2003;162:1089–98. doi: 10.1083/jcb.200212060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Machado RD, Rudarakanchana N, Atkinson C, et al. Functional interaction between BMPR-II and Tctex-1, a light chain of Dynein, is isoform-specific and disrupted by mutations underlying primary pulmonary hypertension. Hum Mol Genet. 2003;12:3277–86. doi: 10.1093/hmg/ddg365. [DOI] [PubMed] [Google Scholar]
  • 33.Wong WK, Knowles JA, Morse JH. Bone morphogenetic protein receptor type II C-terminus interacts with c-Src: implication for a role in pulmonary arterial hypertension. Am J Respir Cell Mol Biol. 2005;33:438–46. doi: 10.1165/rcmb.2005-0103OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zakrzewicz A, Hecker M, Marsh LM, et al. Receptor for activated C-kinase 1, a novel interaction partner of type II bone morphogenetic protein receptor, regulates smooth muscle cell proliferation in pulmonary arterial hypertension. Circulation. 2007;115:2957–68. doi: 10.1161/CIRCULATIONAHA.106.670026. [DOI] [PubMed] [Google Scholar]
  • 35.Machado RD, Aldred MA, James V, et al. Mutations of the TGF-β type II receptor BMPR2 in pulmonary arterial hypertension. Hum Mutat. 2006;27:121–32. doi: 10.1002/humu.20285. [DOI] [PubMed] [Google Scholar]
  • 36.Aldred MA, Machado RD, James V, Morrell NW, Trembath RC. Characterization of the BMPR2 5′-untranslated region and a novel mutation in pulmonary hypertension. Am J Respir Crit Care Med. 2007;176:819–24. doi: 10.1164/rccm.200701-164OC. [DOI] [PubMed] [Google Scholar]
  • 37.Baloira A, Vilariño C, Leiro V, Valverde D. Mutations in the gene encoding bone morphogenetic protein receptor 2 in patients with idiopathic pulmonary arterial hypertension. Arch Bronconeumol. 2008;44:29–34. doi: 10.1016/s1579-2129(08)60006-3. [DOI] [PubMed] [Google Scholar]
  • 38.Fujiwara M, Yagi H, Matsuoka R, et al. Implications of mutations of activin receptor-like kinase 1 gene (ALK1) in addition to bone morphogenetic protein receptor II gene (BMPR2) in children with pulmonary arterial hypertension. Circ J. 2008;72:127–33. doi: 10.1253/circj.72.127. [DOI] [PubMed] [Google Scholar]
  • 39.Fu LJ, Zhou AQ, Huang MR, et al. A novel mutation in the BMPR2 gene in familial pulmonary arterial hypertension. Chin Med J (Engl) 2008;121:399–404. [PubMed] [Google Scholar]
  • 40.Machado RD, Koehler R, Glissmeyer E, et al. Genetic association of the serotonin transporter in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;173:793–7. doi: 10.1164/rccm.200509-1365OC. [DOI] [PubMed] [Google Scholar]
  • 41.Janssen B, Rindermann M, Barth U, et al. Linkage analysis in a large family with primary pulmonary hypertension: genetic heterogeneity and a second primary pulmonary hypertension locus on 2q31-32. Chest. 2002;121:54S–6S. doi: 10.1378/chest.121.3_suppl.54s. [DOI] [PubMed] [Google Scholar]
  • 42.Nasim MT, Ghouri A, Patel B, et al. Stoichiometric imbalance in the receptor complex contributes to dysfunctional BMPR-II mediated signalling in pulmonary arterial hypertension. Hum Mol Genet. 2008;17:1683–94. doi: 10.1093/hmg/ddn059. [DOI] [PubMed] [Google Scholar]
  • 43.Machado RD, Pauciulo MW, Thomson JR, et al. BMPR2 haploinsufficiency as the inherited molecular mechanism for primary pulmonary hypertension. Am J Hum Genet. 2001;68:92–102. doi: 10.1086/316947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Greenwald J, Fischer WH, Vale WW, Choe S. Three-finger toxin fold for the extracellular ligand-binding domain of the type II activin receptor serine kinase. Nat Struct Biol. 1999;6:18–22. doi: 10.1038/4887. [DOI] [PubMed] [Google Scholar]
  • 45.Nishihara A, Watabe T, Imamura T, Miyazono K. Functional heterogeneity of bone morphogenetic protein receptor-II mutants found in patients with primary pulmonary hypertension. Mol Biol Cell. 2002;13:3055–63. doi: 10.1091/mbc.E02-02-0063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rudarakanchana N, Flanagan JA, Chen H, et al. Functional analysis of bone morphogenetic protein type II receptor mutations underlying primary pulmonary hypertension. Hum Mol Genet. 2002;11:1517–25. doi: 10.1093/hmg/11.13.1517. [DOI] [PubMed] [Google Scholar]
  • 47.Hanks SK, Hunter T. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J. 1995;9:576–96. [PubMed] [Google Scholar]
  • 48.Harrison RE, Berger R, Haworth SG, et al. Transforming growth factor-β receptor mutations and pulmonary arterial hypertension in childhood. Circulation. 2005;111:435–41. doi: 10.1161/01.CIR.0000153798.78540.87. [DOI] [PubMed] [Google Scholar]
  • 49.Mache CJ, Gamillscheg A, Popper HH, Haworth SG. Early-life pulmonary arterial hypertension with subsequent development of diffuse pulmonary arteriovenous malformations in hereditary haemorrhagic telangiectasia type 1. Thorax. 2008;63:85–6. doi: 10.1136/thx.2007.076109. [DOI] [PubMed] [Google Scholar]
  • 50.Abdalla SA, Cymerman U, Rushlow D, et al. Novel mutations and polymorphisms in genes causing hereditary hemorrhagic telangiectasia. Hum Mutat. 2005;25:320–1. doi: 10.1002/humu.9312. [DOI] [PubMed] [Google Scholar]
  • 51.Prigoda NL, Savas S, Abdalla SA, et al. Hereditary haemorrhagic telangiectasia: mutation detection, test sensitivity and novel mutations. J Med Genet. 2006;43:722–8. doi: 10.1136/jmg.2006.042606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jones DL, Sandberg JC, Rosenthal MJ, Saunders RC, Hannig VL, Clayton EW. What patients and their relatives think about testing for BMPR2. J Genet Couns. 2008;17:452–8. doi: 10.1007/s10897-008-9172-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. [Accessed October 27, 2008];Genetic Information Nondiscrimination Act. http://thomas.loc.gov/cgi-bin/query/D?c110:6:./temp/~c110jLNYmP.
  • 54.Newman JH, Trembath RC, Morse JA, et al. Genetic basis of pulmonary arterial hypertension: current understanding and future directions. J Am Coll Cardiol. 2004;43:33S–9S. doi: 10.1016/j.jacc.2004.02.028. [DOI] [PubMed] [Google Scholar]
  • 55.McGoon M, Gutterman D, Steen V, et al. Screening, early detection, and diagnosis of pulmonary arterial hypertension: ACCP evidence-based clinical practice guidelines. Chest. 2004;126:14S–34S. doi: 10.1378/chest.126.1_suppl.14S. [DOI] [PubMed] [Google Scholar]
  • 56.Deng Z, Morse JH, Slager SL, et al. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Genet. 2000;67:737–44. doi: 10.1086/303059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Harper JC, Boelaert K, Geraedts J, et al. ESHRE PGD Consortium data collection V: cycles from January to December 2002 with pregnancy follow-up to October 2003. Hum Reprod. 2006;21:3–21. doi: 10.1093/humrep/dei292. [DOI] [PubMed] [Google Scholar]
  • 58.Grünig E, Mereles D, Arnold K, et al. Primary pulmonary hypertension is predominantly a hereditary disease. Chest. 2002;121:81S–2S. doi: 10.1378/chest.121.3_suppl.81s. [DOI] [PubMed] [Google Scholar]
  • 59.Abraham WT, Raynolds MV, Badesch DB, et al. Angiotensin-converting enzyme DD genotype in patients with primary pulmonary hypertension: increased frequency and association with preserved haemodynamics. J Renin Angiotensin Aldosterone Syst. 2003;4:27–30. doi: 10.3317/jraas.2003.003. [DOI] [PubMed] [Google Scholar]
  • 60.Aldashev AA, Sarybaev AS, Sydykov AS, et al. Characterization of high-altitude pulmonary hypertension in the Kyrgyz: association with angiotensin-converting enzyme genotype. Am J Respir Crit Care Med. 2002;166:1396–402. doi: 10.1164/rccm.200204-345OC. [DOI] [PubMed] [Google Scholar]
  • 61.Amano S, Tatsumi K, Tanabe N, et al. Polymorphism of the promoter region of prostacyclin synthase gene in chronic thromboembolic pulmonary hypertension. Respirology. 2004;9:184–9. doi: 10.1111/j.1440-1843.2004.00568.x. [DOI] [PubMed] [Google Scholar]
  • 62.Eddahibi S, Humbert M, Fadel E, et al. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J Clin Invest. 2001;108:1141–50. doi: 10.1172/JCI12805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hoeper MM, Tacacs A, Stellmacher U, Lichtinghagen R. Lack of association between angiotensin converting enzyme (ACE) genotype, serum ACE activity, and haemodynamics in patients with primary pulmonary hypertension. Heart. 2003;89:445–6. doi: 10.1136/heart.89.4.445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Koehler R, Olschewski H, Hoeper M, Janssen B, Grünig E. Serotonin transporter gene polymorphism in a cohort of German patients with idiopathic pulmonary arterial hypertension or chronic thromboembolic pulmonary hypertension. Chest. 2005;128:619S. doi: 10.1378/chest.128.6_suppl.619S. [DOI] [PubMed] [Google Scholar]
  • 65.Phillips JA, 3rd, Poling JS, Phillips CA, et al. Synergistic heterozygosity for TGFbeta1 SNPs and BMPR2 mutations modulates the age at diagnosis and penetrance of familial pulmonary arterial hypertension. Genet Med. 2008;10:359–65. doi: 10.1097/GIM.0b013e318172dcdf. [DOI] [PubMed] [Google Scholar]
  • 66.Remillard CV, Tigno DD, Platoshyn O, et al. Function of Kv1.5 channels and genetic variations of KCNA5 in patients with idiopathic pulmonary arterial hypertension. Am J Physiol Cell Physiol. 2007;292:1837–53. doi: 10.1152/ajpcell.00405.2006. [DOI] [PubMed] [Google Scholar]
  • 67.Willers ED, Newman JH, Loyd JE, et al. Serotonin transporter polymorphisms in familial and idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;173:798–802. doi: 10.1164/rccm.200509-1361OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wipff J, Kahan A, Hachulla E, et al. Association between an endoglin gene polymorphism and systemic sclerosis-related pulmonary arterial hypertension. Rheumatology (Oxford) 2007;46:622–5. doi: 10.1093/rheumatology/kel378. [DOI] [PubMed] [Google Scholar]
  • 69. [Accessed October 27, 2008];International HapMap project. http://www.hapmap.org.
  • 70.Hattersley AT, McCarthy MI. What makes a good genetic association study? Lancet. 2005;366:1315–23. doi: 10.1016/S0140-6736(05)67531-9. [DOI] [PubMed] [Google Scholar]
  • 71.Hirschhorn JN, Daly MJ. Genome-wide association studies for common diseases and complex traits. Nat Rev Genet. 2005;6:95–108. doi: 10.1038/nrg1521. [DOI] [PubMed] [Google Scholar]
  • 72.Lohmueller KE, Pearce CL, Pike M, Lander ES, Hirschhorn JN. Meta-analysis of genetic association studies supports a contribution of common variants to susceptibility to common disease. Nat Genet. 2003;33:177–82. doi: 10.1038/ng1071. [DOI] [PubMed] [Google Scholar]
  • 73.Wacholder S, Chanock S, Garcia-Closas M, El ghormli L, Rothman N. Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J Natl Cancer Inst. 2004;96:434–42. doi: 10.1093/jnci/djh075. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES