Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Genet Med ; 25(11): 100925, 2023 11.
Article in English | MEDLINE | ID: mdl-37422716

ABSTRACT

PURPOSE: Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. Genetic testing is currently recommended for adults diagnosed with heritable, idiopathic, anorexigen-, hereditary hemorrhagic telangiectasia-, and congenital heart disease-associated PAH, PAH with overt features of venous/capillary involvement, and all children diagnosed with PAH. Variants in at least 27 genes have putative evidence for PAH causality. Rigorous assessment of the evidence is needed to inform genetic testing. METHODS: An international panel of experts in PAH applied a semi-quantitative scoring system developed by the NIH Clinical Genome Resource to classify the relative strength of evidence supporting PAH gene-disease relationships based on genetic and experimental evidence. RESULTS: Twelve genes (BMPR2, ACVRL1, ATP13A3, CAV1, EIF2AK4, ENG, GDF2, KCNK3, KDR, SMAD9, SOX17, and TBX4) were classified as having definitive evidence and 3 genes (ABCC8, GGCX, and TET2) with moderate evidence. Six genes (AQP1, BMP10, FBLN2, KLF2, KLK1, and PDGFD) were classified as having limited evidence for causal effects of variants. TOPBP1 was classified as having no known PAH relationship. Five genes (BMPR1A, BMPR1B, NOTCH3, SMAD1, and SMAD4) were disputed because of a paucity of genetic evidence over time. CONCLUSION: We recommend that genetic testing includes all genes with definitive evidence and that caution be taken in the interpretation of variants identified in genes with moderate or limited evidence. Genes with no known evidence for PAH or disputed genes should not be included in genetic testing.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Adult , Child , Humans , Pulmonary Arterial Hypertension/genetics , Mutation , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/genetics , Genetic Predisposition to Disease , Genetic Testing , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Adenosine Triphosphatases/genetics , Membrane Transport Proteins/genetics , Activin Receptors, Type II/genetics , Protein Serine-Threonine Kinases/genetics , Bone Morphogenetic Proteins/genetics
2.
J Med Genet ; 59(9): 906-911, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34493544

ABSTRACT

BACKGROUND: The molecular genetic basis of pulmonary arterial hypertension (PAH) is heterogeneous, with at least 26 genes displaying putative evidence for disease causality. Heterozygous variants in the ATP13A3 gene were recently identified as a new cause of adult-onset PAH. However, the contribution of ATP13A3 risk alleles to child-onset PAH remains largely unexplored. METHODS AND RESULTS: We report three families with a novel, autosomal recessive form of childhood-onset PAH due to biallelic ATP13A3 variants. Disease onset ranged from birth to 2.5 years and was characterised by high mortality. Using genome sequencing of parent-offspring trios, we identified a homozygous missense variant in one case, which was subsequently confirmed to cosegregate with disease in an affected sibling. Independently, compound heterozygous variants in ATP13A3 were identified in two affected siblings and in an unrelated third family. The variants included three loss of function variants (two frameshift, one nonsense) and two highly conserved missense substitutions located in the catalytic phosphorylation domain. The children were largely refractory to treatment and four died in early childhood. All parents were heterozygous for the variants and asymptomatic. CONCLUSION: Our findings support biallelic predicted deleterious ATP13A3 variants in autosomal recessive, childhood-onset PAH, indicating likely semidominant dose-dependent inheritance for this gene.


Subject(s)
Pulmonary Arterial Hypertension , Adenosine Triphosphatases/genetics , Adult , Child, Preschool , Familial Primary Pulmonary Hypertension/genetics , Heterozygote , Homozygote , Humans , Membrane Transport Proteins/genetics , Morbidity
3.
Genes (Basel) ; 12(11)2021 11 16.
Article in English | MEDLINE | ID: mdl-34828405

ABSTRACT

Pulmonary arterial hypertension (PAH) is a highly heterogeneous disorder with a complex, multifactorial aetiology [...].


Subject(s)
Genetic Predisposition to Disease , Hypertension, Pulmonary/genetics , Humans , Hypertension, Pulmonary/diagnosis
4.
Article in English | MEDLINE | ID: mdl-33320693

ABSTRACT

Background - Approximately 25% of patients with pulmonary arterial hypertension (PAH) have been found to harbor rare mutations in disease-causing genes. To identify missing heritability in PAH we integrated deep phenotyping with whole-genome sequencing data using Bayesian statistics. Methods - We analyzed 13,037 participants enrolled in the NIHR BioResource - Rare Diseases (NBR) study, of which 1,148 were recruited to the PAH domain. To test for genetic associations between genes and selected phenotypes of pulmonary hypertension (PH), we used the Bayesian rare-variant association method BeviMed. Results - Heterozygous, high impact, likely loss-of-function variants in the Kinase Insert Domain Receptor (KDR) gene were strongly associated with significantly reduced transfer coefficient for carbon monoxide (KCO, posterior probability (PP)=0.989) and older age at diagnosis (PP=0.912). We also provide evidence for familial segregation of a rare nonsense KDR variant with these phenotypes. On computed tomographic imaging of the lungs, a range of parenchymal abnormalities were observed in the five patients harboring these predicted deleterious variants in KDR. Four additional PAH cases with rare likely loss-of-function variants in KDR were independently identified in the US PAH Biobank cohort with similar phenotypic characteristics. Conclusions - The Bayesian inference approach allowed us to independently validate KDR, which encodes for the Vascular Endothelial Growth Factor Receptor 2 (VEGFR2), as a novel PAH candidate gene. Furthermore, this approach specifically associated high impact likely loss-of-function variants in the genetically constrained gene with distinct phenotypes. These findings provide evidence for KDR being a clinically actionable PAH gene and further support the central role of the vascular endothelium in the pathobiology of PAH.

5.
Genes (Basel) ; 11(11)2020 11 11.
Article in English | MEDLINE | ID: mdl-33187088

ABSTRACT

Pulmonary arterial hypertension (PAH) describes a rare, progressive vascular disease caused by the obstruction of pulmonary arterioles, typically resulting in right heart failure. Whilst PAH most often manifests in adulthood, paediatric disease is considered to be a distinct entity with increased morbidity and often an unexplained resistance to current therapies. Recent genetic studies have substantially increased our understanding of PAH pathogenesis, providing opportunities for molecular diagnosis and presymptomatic genetic testing in families. However, the genetic architecture of childhood-onset PAH remains relatively poorly characterised. We sought to investigate a previously unsolved paediatric cohort (n = 18) using whole exome sequencing to improve the molecular diagnosis of childhood-onset PAH. Through a targeted investigation of 26 candidate genes, we applied a rigorous variant filtering methodology to enrich for rare, likely pathogenic variants. This analysis led to the detection of novel PAH risk alleles in five genes, including the first identification of a heterozygous ATP13A3 mutation in childhood-onset disease. In addition, we provide the first independent validation of BMP10 and PDGFD as genetic risk factors for PAH. These data provide a molecular diagnosis in 28% of paediatric cases, reflecting the increased genetic burden in childhood-onset disease and highlighting the importance of next-generation sequencing approaches to diagnostic surveillance.


Subject(s)
Pulmonary Arterial Hypertension/genetics , Adenosine Triphosphatases/genetics , Bone Morphogenetic Proteins/genetics , Child , Child, Preschool , Cohort Studies , Female , Genetic Predisposition to Disease/genetics , Humans , Infant , Lymphokines/genetics , Male , Membrane Transport Proteins/genetics , Mutation , Platelet-Derived Growth Factor/genetics , Risk Factors , Smad8 Protein/genetics , Sulfonylurea Receptors/genetics , Exome Sequencing/methods
6.
Nat Rev Cardiol ; 17(2): 85-95, 2020 02.
Article in English | MEDLINE | ID: mdl-31406341

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare, progressive disorder typified by occlusion of the pulmonary arterioles owing to endothelial dysfunction and uncontrolled proliferation of pulmonary artery smooth muscle cells and fibroblasts. Vascular occlusion can lead to increased pressure in the pulmonary arteries, often resulting in right ventricular failure with shortness of breath and syncope. Since the identification of BMPR2, which encodes a receptor in the transforming growth factor-ß superfamily, the development of high-throughput sequencing approaches to identify novel causal genes has substantially advanced our understanding of the molecular genetics of PAH. In the past 6 years, additional pathways involved in PAH susceptibility have been described through the identification of deleterious genetic variants in potassium channels (KCNK3 and ABCC8) and transcription factors (TBX4 and SOX17), among others. Although familial PAH most often has an autosomal-dominant pattern of inheritance, cases of incomplete penetrance and evidence of genetic heterogeneity support a model of PAH as a Mendelian disorder with complex disease features. In this Review, we outline the latest advances in the detection of rare and common genetic variants underlying PAH susceptibility and disease progression. These findings have clinical implications for lung vascular function and can help to identify mechanistic pathways amenable to pharmacological intervention.


Subject(s)
Arterial Pressure/genetics , Genetic Variation , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/physiopathology , Animals , Genetic Predisposition to Disease , Humans , Phenotype , Prognosis , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/therapy , Risk Factors , Vascular Remodeling/genetics
7.
Am J Respir Crit Care Med ; 201(5): 575-585, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31661308

ABSTRACT

Rationale: Recently, rare heterozygous mutations in GDF2 were identified in patients with pulmonary arterial hypertension (PAH). GDF2 encodes the circulating BMP (bone morphogenetic protein) type 9, which is a ligand for the BMP2 receptor.Objectives: Here we determined the functional impact of GDF2 mutations and characterized plasma BMP9 and BMP10 levels in patients with idiopathic PAH.Methods: Missense BMP9 mutant proteins were expressed in vitro and the impact on BMP9 protein processing and secretion, endothelial signaling, and functional activity was assessed. Plasma BMP9 and BMP10 levels and activity were assayed in patients with PAH with GDF2 variants and in control subjects. Levels were also measured in a larger cohort of control subjects (n = 120) and patients with idiopathic PAH (n = 260).Measurements and Main Results: We identified a novel rare variation at the GDF2 and BMP10 loci, including copy number variation. In vitro, BMP9 missense proteins demonstrated impaired cellular processing and secretion. Patients with PAH who carried these mutations exhibited reduced plasma levels of BMP9 and reduced BMP activity. Unexpectedly, plasma BMP10 levels were also markedly reduced in these individuals. Although overall BMP9 and BMP10 levels did not differ between patients with PAH and control subjects, BMP10 levels were lower in PAH females. A subset of patients with PAH had markedly reduced plasma levels of BMP9 and BMP10 in the absence of GDF2 mutations.Conclusions: Our findings demonstrate that GDF2 mutations result in BMP9 loss of function and are likely causal. These mutations lead to reduced circulating levels of both BMP9 and BMP10. These findings support therapeutic strategies to enhance BMP9 or BMP10 signaling in PAH.


Subject(s)
Bone Morphogenetic Proteins/genetics , Growth Differentiation Factor 2/genetics , Pulmonary Arterial Hypertension/genetics , Adult , Bone Morphogenetic Proteins/metabolism , Case-Control Studies , DNA Copy Number Variations , Female , Growth Differentiation Factor 2/metabolism , Heterozygote , Humans , Male , Middle Aged , Mutation, Missense , Protein Transport , Pulmonary Arterial Hypertension/metabolism , Sex Factors
8.
Nat Commun ; 9(1): 1416, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29650961

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare disorder with a poor prognosis. Deleterious variation within components of the transforming growth factor-ß pathway, particularly the bone morphogenetic protein type 2 receptor (BMPR2), underlies most heritable forms of PAH. To identify the missing heritability we perform whole-genome sequencing in 1038 PAH index cases and 6385 PAH-negative control subjects. Case-control analyses reveal significant overrepresentation of rare variants in ATP13A3, AQP1 and SOX17, and provide independent validation of a critical role for GDF2 in PAH. We demonstrate familial segregation of mutations in SOX17 and AQP1 with PAH. Mutations in GDF2, encoding a BMPR2 ligand, lead to reduced secretion from transfected cells. In addition, we identify pathogenic mutations in the majority of previously reported PAH genes, and provide evidence for further putative genes. Taken together these findings contribute new insights into the molecular basis of PAH and indicate unexplored pathways for therapeutic intervention.


Subject(s)
Adenosine Triphosphatases/chemistry , Aquaporin 1/chemistry , Familial Primary Pulmonary Hypertension/genetics , Growth Differentiation Factors/chemistry , Membrane Transport Proteins/chemistry , Mutation , SOXF Transcription Factors/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adult , Aquaporin 1/genetics , Aquaporin 1/metabolism , Base Sequence , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Case-Control Studies , Familial Primary Pulmonary Hypertension/diagnosis , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/pathology , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Growth Differentiation Factor 2 , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , HEK293 Cells , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Prognosis , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Whole Genome Sequencing
9.
Circulation ; 136(21): 2022-2033, 2017 11 21.
Article in English | MEDLINE | ID: mdl-28972005

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a rare disease with an emerging genetic basis. Heterozygous mutations in the gene encoding the bone morphogenetic protein receptor type 2 (BMPR2) are the commonest genetic cause of PAH, whereas biallelic mutations in the eukaryotic translation initiation factor 2 alpha kinase 4 gene (EIF2AK4) are described in pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis. Here, we determine the frequency of these mutations and define the genotype-phenotype characteristics in a large cohort of patients diagnosed clinically with PAH. METHODS: Whole-genome sequencing was performed on DNA from patients with idiopathic and heritable PAH and with pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis recruited to the National Institute of Health Research BioResource-Rare Diseases study. Heterozygous variants in BMPR2 and biallelic EIF2AK4 variants with a minor allele frequency of <1:10 000 in control data sets and predicted to be deleterious (by combined annotation-dependent depletion, PolyPhen-2, and sorting intolerant from tolerant predictions) were identified as potentially causal. Phenotype data from the time of diagnosis were also captured. RESULTS: Eight hundred sixty-four patients with idiopathic or heritable PAH and 16 with pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis were recruited. Mutations in BMPR2 were identified in 130 patients (14.8%). Biallelic mutations in EIF2AK4 were identified in 5 patients with a clinical diagnosis of pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis. Furthermore, 9 patients with a clinical diagnosis of PAH carried biallelic EIF2AK4 mutations. These patients had a reduced transfer coefficient for carbon monoxide (Kco; 33% [interquartile range, 30%-35%] predicted) and younger age at diagnosis (29 years; interquartile range, 23-38 years) and more interlobular septal thickening and mediastinal lymphadenopathy on computed tomography of the chest compared with patients with PAH without EIF2AK4 mutations. However, radiological assessment alone could not accurately identify biallelic EIF2AK4 mutation carriers. Patients with PAH with biallelic EIF2AK4 mutations had a shorter survival. CONCLUSIONS: Biallelic EIF2AK4 mutations are found in patients classified clinically as having idiopathic and heritable PAH. These patients cannot be identified reliably by computed tomography, but a low Kco and a young age at diagnosis suggests the underlying molecular diagnosis. Genetic testing can identify these misclassified patients, allowing appropriate management and early referral for lung transplantation.


Subject(s)
Arterial Pressure/genetics , Hypertension, Pulmonary/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Pulmonary Artery/physiopathology , Adult , Aged , Bone Morphogenetic Protein Receptors, Type II/genetics , DNA Mutational Analysis , Europe , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Heredity , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Pedigree , Phenotype , Predictive Value of Tests , Prospective Studies , Retrospective Studies , Risk Factors , Tomography, X-Ray Computed , Young Adult
10.
Hum Mutat ; 36(12): 1113-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26387786

ABSTRACT

Pulmonary arterial hypertension (PAH) is an often fatal disorder resulting from several causes including heterogeneous genetic defects. While mutations in the bone morphogenetic protein receptor type II (BMPR2) gene are the single most common causal factor for hereditary cases, pathogenic mutations have been observed in approximately 25% of idiopathic PAH patients without a prior family history of disease. Additional defects of the transforming growth factor beta pathway have been implicated in disease pathogenesis. Specifically, studies have confirmed activin A receptor type II-like 1 (ACVRL1), endoglin (ENG), and members of the SMAD family as contributing to PAH both with and without associated clinical phenotypes. Most recently, next-generation sequencing has identified novel, rare genetic variation implicated in the PAH disease spectrum. Of importance, several identified genetic factors converge on related pathways and provide significant insight into the development, maintenance, and pathogenetic transformation of the pulmonary vascular bed. Together, these analyses represent the largest comprehensive compilation of BMPR2 and associated genetic risk factors for PAH, comprising known and novel variation. Additionally, with the inclusion of an allelic series of locus-specific variation in BMPR2, these data provide a key resource in data interpretation and development of contemporary therapeutic and diagnostic tools.


Subject(s)
Hypertension, Pulmonary/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/chemistry , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Disease Models, Animal , Genetic Association Studies , Genetic Counseling , Genetic Predisposition to Disease , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/metabolism , Multigene Family , Mutation , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
11.
Nat Med ; 21(7): 777-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26076038

ABSTRACT

Genetic evidence implicates the loss of bone morphogenetic protein type II receptor (BMPR-II) signaling in the endothelium as an initiating factor in pulmonary arterial hypertension (PAH). However, selective targeting of this signaling pathway using BMP ligands has not yet been explored as a therapeutic strategy. Here, we identify BMP9 as the preferred ligand for preventing apoptosis and enhancing monolayer integrity in both pulmonary arterial endothelial cells and blood outgrowth endothelial cells from subjects with PAH who bear mutations in the gene encoding BMPR-II, BMPR2. Mice bearing a heterozygous knock-in allele of a human BMPR2 mutation, R899X, which we generated as an animal model of PAH caused by BMPR-II deficiency, spontaneously developed PAH. Administration of BMP9 reversed established PAH in these mice, as well as in two other experimental PAH models, in which PAH develops in response to either monocrotaline or VEGF receptor inhibition combined with chronic hypoxia. These results demonstrate the promise of direct enhancement of endothelial BMP signaling as a new therapeutic strategy for PAH.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Endothelial Cells/metabolism , Growth Differentiation Factor 2/pharmacology , Hypertension, Pulmonary/pathology , Pulmonary Artery/pathology , Aging/pathology , Animals , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Densitometry , Endothelial Cells/drug effects , Endothelial Cells/pathology , Gene Expression Profiling , Gene Knock-In Techniques , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Immunoblotting , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Monocrotaline , Phosphorylation/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Systole/drug effects , Transcription, Genetic/drug effects
12.
Circ Cardiovasc Genet ; 8(4): 572-581, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25963545

ABSTRACT

BACKGROUND: Adams-Oliver syndrome (AOS) is a rare disorder characterized by congenital limb defects and scalp cutis aplasia. In a proportion of cases, notable cardiac involvement is also apparent. Despite recent advances in the understanding of the genetic basis of AOS, for the majority of affected subjects, the underlying molecular defect remains unresolved. This study aimed to identify novel genetic determinants of AOS. METHODS AND RESULTS: Whole-exome sequencing was performed for 12 probands, each with a clinical diagnosis of AOS. Analyses led to the identification of novel heterozygous truncating NOTCH1 mutations (c.1649dupA and c.6049_6050delTC) in 2 kindreds in which AOS was segregating as an autosomal dominant trait. Screening a cohort of 52 unrelated AOS subjects, we detected 8 additional unique NOTCH1 mutations, including 3 de novo amino acid substitutions, all within the ligand-binding domain. Congenital heart anomalies were noted in 47% (8/17) of NOTCH1-positive probands and affected family members. In leukocyte-derived RNA from subjects harboring NOTCH1 extracellular domain mutations, we observed significant reduction of NOTCH1 expression, suggesting instability and degradation of mutant mRNA transcripts by the cellular machinery. Transient transfection of mutagenized NOTCH1 missense constructs also revealed significant reduction in gene expression. Mutant NOTCH1 expression was associated with downregulation of the Notch target genes HEY1 and HES1, indicating that NOTCH1-related AOS arises through dysregulation of the Notch signaling pathway. CONCLUSIONS: These findings highlight a key role for NOTCH1 across a range of developmental anomalies that include cardiac defects and implicate NOTCH1 haploinsufficiency as a likely molecular mechanism for this group of disorders.


Subject(s)
Ectodermal Dysplasia/genetics , Genetic Predisposition to Disease/genetics , Haploinsufficiency , Heart Defects, Congenital/genetics , Limb Deformities, Congenital/genetics , Receptor, Notch1/genetics , Scalp Dermatoses/congenital , Adolescent , Adult , Base Sequence , Child , Exome/genetics , Family Health , Female , Gene Expression , Humans , Male , Middle Aged , Models, Molecular , Pedigree , Protein Structure, Tertiary , Receptor, Notch1/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Scalp Dermatoses/genetics , Sequence Analysis, DNA/methods , Signal Transduction/genetics , Young Adult
13.
Turk Kardiyol Dern Ars ; 42 Suppl 1: 17-28, 2014 Oct.
Article in Turkish | MEDLINE | ID: mdl-25697031

ABSTRACT

Major discoveries have been obtained within the last decade in the field of hereditary predisposition to pulmonary arterial hypertension (PAH). Among them, the identification of bone morphogenetic protein receptor type 2 (BMPR2) as the major predisposing gene and activin A receptor type II-like kinase-1 (ACVRL1, also known as ALK1) as the major gene when PAH is associated with hereditary hemorrhagic telangiectasia. The mutation detection rate for the known genes is approximately 75% in familial PAH, but the mutation shortfall remains unexplained even after careful molecular investigation of these genes. To identify additional genetic variants predisposing to PAH, investigators harnessed the power of next-generation sequencing to successfully identify additional genes that will be described in this report. Furthermore; common genetic predisposing factors for PAH can be identified by genome-wide association studies and are detailed in this paper. The careful study of families and routine genetic diagnosis facilitated natural-history studies based on large registries of PAH patients to be set up in different countries. These longitudinal or cross-sectional studies permitted the clinical characterization of PAH in mutation carriers to be accurately described. The availability of molecular genetic diagnosis has opened up a new field for patient care, including genetic counseling for a severe disease, taking into account that the major predisposing gene has a highly variable penetrance between families. Molecular information can be drawn from the genomic study of affected tissues in PAH, in particular, pulmonary vascular tissues and cells, to gain insight into the mechanisms leading to the development of the disease. High-throughput genomic techniques, on the basis of next-generation sequencing, now allow the accurate quantification and analysis of ribonucleic acid, species, including micro-ribonucleic acids, and allow for a genome-wide investigation of epigenetic or regulatory mechanisms, which include deoxyribonucleic acid methylation, histone methylation, and acetylation, or transcription factor binding. (J Am Coll Cardiol 2013;62:D13-21) a 2013 by the American College of Cardiology Foundation.

14.
J Am Coll Cardiol ; 62(25 Suppl): D13-21, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24355637

ABSTRACT

Major discoveries have been obtained within the last decade in the field of hereditary predisposition to pulmonary arterial hypertension (PAH). Among them, the identification of bone morphogenetic protein receptor type 2 (BMPR2) as the major predisposing gene and activin A receptor type II-like kinase-1 (ACVRL1, also known as ALK1) as the major gene when PAH is associated with hereditary hemorrhagic telangiectasia. The mutation detection rate for the known genes is approximately 75% in familial PAH, but the mutation shortfall remains unexplained even after careful molecular investigation of these genes. To identify additional genetic variants predisposing to PAH, investigators harnessed the power of next-generation sequencing to successfully identify additional genes that will be described in this report. Furthermore, common genetic predisposing factors for PAH can be identified by genome-wide association studies and are detailed in this paper. The careful study of families and routine genetic diagnosis facilitated natural history studies based on large registries of PAH patients to be set up in different countries. These longitudinal or cross-sectional studies permitted the clinical characterization of PAH in mutation carriers to be accurately described. The availability of molecular genetic diagnosis has opened up a new field for patient care, including genetic counseling for a severe disease, taking into account that the major predisposing gene has a highly variable penetrance between families. Molecular information can be drawn from the genomic study of affected tissues in PAH, in particular, pulmonary vascular tissues and cells, to gain insight into the mechanisms leading to the development of the disease. High-throughput genomic techniques, on the basis of next-generation sequencing, now allow the accurate quantification and analysis of ribonucleic acid, species, including micro-ribonucleic acids, and allow for a genome-wide investigation of epigenetic or regulatory mechanisms, which include deoxyribonucleic acid methylation, histone methylation, and acetylation, or transcription factor binding.


Subject(s)
Genetic Predisposition to Disease/genetics , Genomics , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Familial Primary Pulmonary Hypertension , Genetic Predisposition to Disease/epidemiology , Genome-Wide Association Study/trends , Genomics/trends , Humans , Hypertension, Pulmonary/epidemiology , Mutation/genetics
16.
Circulation ; 126(9): 1099-109, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22832786

ABSTRACT

BACKGROUND: Beyond their role as innate immune effectors, natural killer (NK) cells are emerging as important regulators of angiogenesis and vascular remodeling. Pulmonary arterial hypertension (PAH) is characterized by severe pulmonary vascular remodeling and has long been associated with immune dysfunction. Despite this association, a role for NK cells in disease pathology has not yet been described. METHODS AND RESULTS: Analysis of whole blood lymphocytes and isolated NK cells from PAH patients revealed an expansion of the functionally defective CD56(-)/CD16(+) NK subset that was not observed in patients with chronic thromboembolic pulmonary hypertension. NK cells from PAH patients also displayed decreased levels of the activating receptor NKp46 and the killer immunoglobulin-like receptors 2DL1/S1 and 3DL1, reduced secretion of the cytokine macrophage inflammatory protein-1ß, and a significant impairment in cytolytic function associated with decreased killer immunoglobulin-like receptor 3DL1 expression. Genotyping patients (n=222) and controls (n=191) for killer immunoglobulin-like receptor gene polymorphisms did not explain these observations. Rather, we show that NK cells from PAH patients exhibit increased responsiveness to transforming growth factor-ß, which specifically downregulates disease-associated killer immunoglobulin-like receptors. NK cell number and cytotoxicity were similarly decreased in the monocrotaline rat and chronic hypoxia mouse models of PAH, accompanied by reduced production of interferon-γ in NK cells from hypoxic mice. NK cells from PAH patients also produced elevated quantities of matrix metalloproteinase 9, consistent with a capacity to influence vascular remodeling. CONCLUSIONS: Our work is the first to identify an impairment of NK cells in PAH and suggests a novel and substantive role for innate immunity in the pathobiology of this disease.


Subject(s)
Hypertension, Pulmonary/immunology , Killer Cells, Natural/immunology , Adult , Aged , Animals , CD56 Antigen/analysis , Chemokine CCL4/metabolism , Cytotoxicity, Immunologic/drug effects , Extracellular Matrix/metabolism , Female , GPI-Linked Proteins/analysis , Genotype , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Immunophenotyping , Killer Cells, Natural/chemistry , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Male , Matrix Metalloproteinase 9/analysis , Mice , Mice, Inbred C57BL , Middle Aged , Natural Cytotoxicity Triggering Receptor 1 , Pulmonary Embolism/complications , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, IgG/analysis , Receptors, KIR2DL1/biosynthesis , Receptors, KIR2DL1/genetics , Receptors, KIR3DL1/biosynthesis , Receptors, KIR3DL1/genetics , Receptors, KIR3DS1/biosynthesis , Receptors, KIR3DS1/genetics , Transforming Growth Factor beta/pharmacology
17.
Scientifica (Cairo) ; 2012: 106576, 2012.
Article in English | MEDLINE | ID: mdl-24278664

ABSTRACT

Pulmonary arterial hypertension (PAH) is an incurable disorder clinically characterised by a sustained elevation of mean arterial pressure in the absence of systemic involvement. As the adult circulation is a low pressure, low resistance system, PAH represents a reversal to a foetal state. The small pulmonary arteries of patients exhibit luminal occlusion resultant from the uncontrolled growth of endothelial and smooth muscle cells. This vascular remodelling is comprised of hallmark defects, most notably the plexiform lesion. PAH may be familial in nature but the majority of patients present with spontaneous disease or PAH associated with other complications. In this paper, the molecular genetic basis of the disorder is discussed in detail ranging from the original identification of the major genetic contributant to PAH and moving on to current next-generation technologies that have led to the rapid identification of additional genetic risk factors. The impact of identified mutations on the cell is examined, particularly, the determination of pathways disrupted in disease and critical to pulmonary vascular maintenance. Finally, the application of research in this area to the design and development of novel treatment options for patients is addressed along with the future directions PAH research is progressing towards.

18.
Hum Mutat ; 32(12): 1385-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21898662

ABSTRACT

Heterozygous germline mutations of BMPR2 contribute to familial clustering of pulmonary arterial hypertension (PAH). To further explore the genetic basis of PAH in isolated cases, we undertook a candidate gene analysis to identify potentially deleterious variation. Members of the bone morphogenetic protein (BMP) pathway, namely SMAD1, SMAD4, SMAD5, and SMAD9, were screened by direct sequencing for gene defects. Four variants were identified in SMADs 1, 4, and 9 among a cohort of 324 PAH cases, each not detected in a substantial control population. Of three amino acid substitutions identified, two demonstrated reduced signaling activity in vitro. A putative splice site mutation in SMAD4 resulted in moderate transcript loss due to compromised splicing efficiency. These results demonstrate the role of BMPR2 mutation in the pathogenesis of PAH and indicate that variation within the SMAD family represents an infrequent cause of the disease.


Subject(s)
Hypertension, Pulmonary/genetics , Signal Transduction/genetics , Bone Morphogenetic Protein Receptors, Type II/genetics , Cohort Studies , Familial Primary Pulmonary Hypertension , Female , Gene Expression Regulation , Humans , Male , Sequence Analysis, DNA , Smad1 Protein/genetics , Smad8 Protein/genetics
19.
Am J Hum Genet ; 88(5): 574-85, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21565291

ABSTRACT

Regulation of cell proliferation and motility is essential for normal development. The Rho family of GTPases plays a critical role in the control of cell polarity and migration by effecting the cytoskeleton, membrane trafficking, and cell adhesion. We investigated a recognized developmental disorder, Adams-Oliver syndrome (AOS), characterized by the combination of aplasia cutis congenita (ACC) and terminal transverse limb defects (TTLD). Through a genome-wide linkage analysis, we detected a locus for autosomal-dominant ACC-TTLD on 3q generating a maximum LOD score of 4.93 at marker rs1464311. Candidate-gene- and exome-based sequencing led to the identification of independent premature truncating mutations in the terminal exon of the Rho GTPase-activating protein 31 gene, ARHGAP31, which encodes a Cdc42/Rac1 regulatory protein. Mutant transcripts are stable and increase ARHGAP31 activity in vitro through a gain-of-function mechanism. Constitutively active ARHGAP31 mutations result in a loss of available active Cdc42 and consequently disrupt actin cytoskeletal structures. Arhgap31 expression in the mouse is substantially restricted to the terminal limb buds and craniofacial processes during early development; these locations closely mirror the sites of impaired organogenesis that characterize this syndrome. These data identify the requirement for regulated Cdc42 and/or Rac1 signaling processes during early human development.


Subject(s)
Ectodermal Dysplasia/genetics , GTPase-Activating Proteins/genetics , Mutation , Actins/metabolism , Cell Adhesion , Cell Movement , Cell Polarity , Cell Proliferation , Chromosome Mapping , Cytoskeleton/metabolism , DNA Mutational Analysis , Ectodermal Dysplasia/embryology , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Limb Deformities, Congenital/embryology , Limb Deformities, Congenital/genetics , Male , Scalp Dermatoses/congenital , Scalp Dermatoses/embryology , Scalp Dermatoses/genetics , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
20.
Hum Mutat ; 32(2): 231-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21280149

ABSTRACT

Dyggve-Melchior-Clausen syndrome (DMC), a severe autosomal recessive skeletal disorder with mental retardation, is caused by mutation of the gene encoding Dymeclin (DYM). Employing patient fibroblasts with mutations characterized at the genomic and, for the first time, transcript level, we identified profound disruption of Golgi organization as a pathogenic feature, resolved by transfection of heterologous wild-type Dymeclin. Collagen targeting appeared defective in DMC cells leading to near complete absence of cell surface collagen fibers. DMC cells have an elevated apoptotic index (P< 0.01) likely due to a stress response contingent upon Golgi-related trafficking defects. We performed spatiotemporal mapping of Dymeclin expression in zebrafish embryos and identified high levels of transcript in brain and cartilage during early development. Finally, in a chondrocyte cDNA library, we identified two novel secretion pathway proteins as Dymeclin interacting partners: GOLM1 and PPIB. Together these data identify the role of Dymeclin in secretory pathways essential to endochondral bone formation during early development.


Subject(s)
Bone Development , Extracellular Matrix/metabolism , Golgi Apparatus/metabolism , Proteins/metabolism , Animals , Cells, Cultured , Chondrogenesis , Cytoplasm/metabolism , Dwarfism/metabolism , Fibroblasts/metabolism , Genetic Diseases, X-Linked/metabolism , HeLa Cells , Humans , Intellectual Disability/metabolism , Intracellular Signaling Peptides and Proteins , Mutation , Osteochondrodysplasias/congenital , Osteochondrodysplasias/metabolism , Skin/cytology , Two-Hybrid System Techniques , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...