Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genet Med ; 25(11): 100925, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37422716

RESUMEN

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.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Adulto , Niño , Humanos , Hipertensión Arterial Pulmonar/genética , Mutación , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Transporte de Membrana/genética , Receptores de Activinas Tipo II/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Morfogenéticas Óseas/genética
2.
J Med Genet ; 59(9): 906-911, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34493544

RESUMEN

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.


Asunto(s)
Hipertensión Arterial Pulmonar , Adenosina Trifosfatasas/genética , Adulto , Preescolar , Hipertensión Pulmonar Primaria Familiar/genética , Heterocigoto , Homocigoto , Humanos , Proteínas de Transporte de Membrana/genética , Morbilidad
3.
Am J Respir Crit Care Med ; 201(5): 575-585, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31661308

RESUMEN

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.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Factor 2 de Diferenciación de Crecimiento/genética , Hipertensión Arterial Pulmonar/genética , Adulto , Proteínas Morfogenéticas Óseas/metabolismo , Estudios de Casos y Controles , Variaciones en el Número de Copia de ADN , Femenino , Factor 2 de Diferenciación de Crecimiento/metabolismo , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Transporte de Proteínas , Hipertensión Arterial Pulmonar/metabolismo , Factores Sexuales
4.
Hum Mutat ; 36(12): 1113-27, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26387786

RESUMEN

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.


Asunto(s)
Hipertensión Pulmonar/genética , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/química , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Asesoramiento Genético , Predisposición Genética a la Enfermedad , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/metabolismo , Familia de Multigenes , Mutación , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
5.
Nat Genet ; 38(11): 1242-4, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17013395

RESUMEN

Idiopathic congenital nystagmus is characterized by involuntary, periodic, predominantly horizontal oscillations of both eyes. We identified 22 mutations in FRMD7 in 26 families with X-linked idiopathic congenital nystagmus. Screening of 42 singleton cases of idiopathic congenital nystagmus (28 male, 14 females) yielded three mutations (7%). We found restricted expression of FRMD7 in human embryonic brain and developing neural retina, suggesting a specific role in the control of eye movement and gaze stability.


Asunto(s)
Proteínas del Citoesqueleto/genética , Genes Ligados a X , Proteínas de la Membrana/genética , Nistagmo Congénito/genética , Encéfalo/embriología , Encéfalo/metabolismo , Mapeo Cromosómico , Cromosomas Humanos X , Proteínas del Citoesqueleto/fisiología , Movimientos Oculares/genética , Movimientos Oculares/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Ligamiento Genético , Humanos , Masculino , Proteínas de la Membrana/fisiología , Mutación/fisiología , Linaje , Retina/metabolismo
6.
Am J Hum Genet ; 88(5): 574-85, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565291

RESUMEN

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.


Asunto(s)
Displasia Ectodérmica/genética , Proteínas Activadoras de GTPasa/genética , Mutación , Actinas/metabolismo , Adhesión Celular , Movimiento Celular , Polaridad Celular , Proliferación Celular , Mapeo Cromosómico , Citoesqueleto/metabolismo , Análisis Mutacional de ADN , Displasia Ectodérmica/embriología , Femenino , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Deformidades Congénitas de las Extremidades/embriología , Deformidades Congénitas de las Extremidades/genética , Masculino , Dermatosis del Cuero Cabelludo/congénito , Dermatosis del Cuero Cabelludo/embriología , Dermatosis del Cuero Cabelludo/genética , Transducción de Señal , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo
7.
Hum Mutat ; 32(2): 231-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21280149

RESUMEN

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.


Asunto(s)
Desarrollo Óseo , Matriz Extracelular/metabolismo , Aparato de Golgi/metabolismo , Proteínas/metabolismo , Animales , Células Cultivadas , Condrogénesis , Citoplasma/metabolismo , Enanismo/metabolismo , Fibroblastos/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Células HeLa , Humanos , Discapacidad Intelectual/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Mutación , Osteocondrodisplasias/congénito , Osteocondrodisplasias/metabolismo , Piel/citología , Técnicas del Sistema de Dos Híbridos , Pez Cebra/embriología
8.
Hum Mutat ; 32(12): 1385-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21898662

RESUMEN

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.


Asunto(s)
Hipertensión Pulmonar/genética , Transducción de Señal/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Estudios de Cohortes , Hipertensión Pulmonar Primaria Familiar , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Análisis de Secuencia de ADN , Proteína Smad1/genética , Proteína Smad8/genética
9.
Circulation ; 122(9): 920-7, 2010 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-20713898

RESUMEN

BACKGROUND: Inflammation is a feature of pulmonary arterial hypertension (PAH), and increased circulating levels of cytokines are reported in patients with PAH. However, to date, no information exists on the significance of elevated cytokines or their potential as biomarkers. We sought to determine the levels of a range of cytokines in PAH and to examine their impact on survival and relationship to hemodynamic indexes. METHODS AND RESULTS: We measured levels of serum cytokines (tumor necrosis factor-alpha, interferon-gamma and interleukin-1beta, -2, -4, -5, -6, -8, -10, -12p70, and -13) using ELISAs in idiopathic and heritable PAH patients (n=60). Concurrent clinical data included hemodynamics, 6-minute walk distance, and survival time from sampling to death or transplantation. Healthy volunteers served as control subjects (n=21). PAH patients had significantly higher levels of interleukin-1beta, -2, -4, -6, -8, -10, and -12p70 and tumor necrosis factor-alpha compared with healthy control subjects. Kaplan-Meier analysis showed that levels of interleukin-6, 8, 10, and 12p70 predicted survival in patients. For example, 5-year survival with interleukin-6 levels of >9 pg/mL was 30% compared with 63% for patients with levels < or = 9 pg/mL (P=0.008). In this PAH cohort, cytokine levels were superior to traditional markers of prognosis such as 6-minute walk distance and hemodynamics. CONCLUSIONS: This study illustrates dysregulation of a broad range of inflammatory mediators in idiopathic and familial PAH and demonstrates that cytokine levels have a previously unrecognized impact on patient survival. They may prove to be useful biomarkers and provide insight into the contribution of inflammation in PAH.


Asunto(s)
Biomarcadores/sangre , Citocinas/sangre , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/mortalidad , Inflamación/diagnóstico , Inflamación/mortalidad , Adulto , Anciano , Presión Sanguínea , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hipertensión Pulmonar/genética , Inflamación/genética , Mediadores de Inflamación/sangre , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Análisis de Supervivencia , Resistencia Vascular
10.
Genes (Basel) ; 12(11)2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34828405

RESUMEN

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


Asunto(s)
Predisposición Genética a la Enfermedad , Hipertensión Pulmonar/genética , Humanos , Hipertensión Pulmonar/diagnóstico
11.
Circulation ; 119(13): 1747-57, 2009 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-19307479

RESUMEN

BACKGROUND: This large, prospective, multicentric study was performed to analyze the distribution of tricuspid regurgitation velocity (TRV) values during exercise and hypoxia in relatives of patients with idiopathic and familial pulmonary arterial hypertension (PAH) and in healthy control subjects. We tested the hypothesis that relatives of idiopathic/familial PAH patients display an enhanced frequency of hypertensive TRV response to stress and that this response is associated with mutations in the bone morphogenetic protein receptor II (BMPR2) gene. METHODS AND RESULTS: TRV was estimated by Doppler echocardiography during supine bicycle exercise in normoxia and during 120 minutes of normobaric hypoxia (FIO(2)=12%; approximately 4500 m) in 291 relatives of 109 PAH patients and in 191 age-matched control subjects. Mean maximal TRVs were significantly higher in PAH relatives during both exercise and hypoxia. During exercise, 10% of control subjects but 31.6% of relatives (P<0.0001) exceeded the 90% quantile of mean maximal TRV seen in control subjects. Hypoxia revealed hypertensive TRV in 26% of relatives (P=0.0029). Among control subjects, TRV at rest was not related to age, sex, body mass index, systemic blood pressure, smoking status, or heart rate. Within kindreds identified as harboring deleterious mutations of the BMPR2 gene, a hypertensive TRV response occurred significantly more often compared with those without detected mutations. CONCLUSIONS: Pulmonary hypertensive response to exercise and hypoxia in idiopathic/familial PAH relatives appears as a genetic trait with familial clustering, being correlated to but not caused by a BMPR2 mutation. The suitability of this trait to predict manifest PAH development should be addressed in long-term follow-up studies.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Ecocardiografía Doppler , Hipertensión Pulmonar/diagnóstico por imagen , Hipertensión Pulmonar/genética , Insuficiencia de la Válvula Tricúspide/diagnóstico por imagen , Insuficiencia de la Válvula Tricúspide/genética , Adulto , Presión Sanguínea/fisiología , Europa (Continente) , Ejercicio Físico/fisiología , Prueba de Esfuerzo , Familia , Femenino , Frecuencia Cardíaca/fisiología , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Hipertensión Pulmonar/fisiopatología , Hipoxia/genética , Hipoxia/fisiopatología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Descanso/fisiología , Insuficiencia de la Válvula Tricúspide/fisiopatología , Adulto Joven
12.
Nat Rev Cardiol ; 17(2): 85-95, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31406341

RESUMEN

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.


Asunto(s)
Presión Arterial/genética , Variación Genética , Hipertensión Arterial Pulmonar/genética , Arteria Pulmonar/fisiopatología , Animales , Predisposición Genética a la Enfermedad , Humanos , Fenotipo , Pronóstico , Hipertensión Arterial Pulmonar/fisiopatología , Hipertensión Arterial Pulmonar/terapia , Factores de Riesgo , Remodelación Vascular/genética
13.
Genes (Basel) ; 11(11)2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33187088

RESUMEN

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.


Asunto(s)
Hipertensión Arterial Pulmonar/genética , Adenosina Trifosfatasas/genética , Proteínas Morfogenéticas Óseas/genética , Niño , Preescolar , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Lactante , Linfocinas/genética , Masculino , Proteínas de Transporte de Membrana/genética , Mutación , Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Riesgo , Proteína Smad8/genética , Receptores de Sulfonilureas/genética , Secuenciación del Exoma/métodos
14.
Artículo en Inglés | MEDLINE | ID: mdl-33320693

RESUMEN

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.

16.
Nat Commun ; 9(1): 1416, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29650961

RESUMEN

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.


Asunto(s)
Adenosina Trifosfatasas/química , Acuaporina 1/química , Hipertensión Pulmonar Primaria Familiar/genética , Factores de Diferenciación de Crecimiento/química , Proteínas de Transporte de Membrana/química , Mutación , Factores de Transcripción SOXF/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adulto , Acuaporina 1/genética , Acuaporina 1/metabolismo , Secuencia de Bases , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Estudios de Casos y Controles , Hipertensión Pulmonar Primaria Familiar/diagnóstico , Hipertensión Pulmonar Primaria Familiar/metabolismo , Hipertensión Pulmonar Primaria Familiar/patología , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Modelos Moleculares , Pronóstico , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Secuenciación Completa del Genoma
17.
Hum Mutat ; 27(2): 121-32, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16429395

RESUMEN

Pulmonary arterial hypertension (PAH) is clinically characterized by a sustained elevation in mean pulmonary artery pressure leading to significant morbidity and mortality. The disorder is typically sporadic, and in such cases the term idiopathic PAH (IPAH) is used. However, cases that occur within families (familial PAH (FPAH)) display similar clinical and histopathological features, suggesting a common etiology. Heterozygous mutations of a type II member of the TGF-beta cell signaling superfamily known as BMPR2 on chromosome 2q33 have been identified in many kindreds with FPAH, yet display both reduced penetrance and sex bias. This report presents the compilation of data for 144 distinct mutations that alter the coding sequence of the BMPR2 gene identified in 210 independent PAH subjects. This large data set characterizes the extent of sequence variation and reveals that the majority (71%) of mutations in FPAH and IPAH comprise nonsense, frameshift, and splice-site defects, and gene rearrangements. These predict premature termination of the transcript with likely loss through the process of nonsense-mediated decay (NMD). A total of 44 missense mutations were identified that substitute amino acid residues at highly conserved sites within recognized functional domains of the mature receptor. We assess this category of mutations in the context of their heterogeneous effects on cell signaling when assayed by in vitro cell-based systems. Disease-causing mutation hot-spots within BMPR2 are summarized. Taken together, these observations are likely to aid in the development of targeted mutation detection strategies relevant for patient management. Finally, we examine the age- and sex-dependent reduced penetrance of BMPR2 mutations by reviewing bmpr2 animal models and the requirement for additional genetic and/or environmental modifiers of disease. In conclusion, these data provide compelling genetic evidence that haploinsufficiency is the predominant molecular mechanism underlying disease predisposition, and support the concept of a critical threshold of signaling activity below which disease may be precipitated.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Mutación , Arteria Pulmonar/patología , Receptores de Factores de Crecimiento Transformadores beta/genética , Mapeo Cromosómico , Heterocigoto , Humanos , Modelos Biológicos , Mutación Missense , Polimorfismo Genético , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal
18.
Circulation ; 111(5): 607-13, 2005 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-15699281

RESUMEN

BACKGROUND: Primary pulmonary arterial hypertension (PAH) is a potentially devastating condition resulting from occlusion of the pulmonary arterioles by the formation of vascular lesions. Heterozygous mutations in the gene encoding the bone morphogenetic protein receptor type II (BMPR2) have been identified in both familial (FPAH) and idiopathic PAH. Mutant alleles are typically of low penetrance, indicating that other factors are required for the onset of PAH. Previous reports have suggested that the characteristic plexiform lesions in affected lungs are akin to neoplasia, showing monoclonal expansion and microsatellite instability. We hypothesized that in patients with germline mutations, BMPR2 might behave as a classic tumor suppressor gene, with somatic loss of the wild-type allele contributing to disease progression. METHODS AND RESULTS: To test this hypothesis, plexiform and concentric vascular lesions were serially microdissected from lung explant tissue derived from 7 FPAH cases. DNA was analyzed for loss of heterozygosity at BMPR2 and for microsatellite instability (MSI) at 5 loci. MSI was detected in 1 of 37 lesions at a single locus, BAT-26, whereas heterozygosity at BMPR2 was retained at all informative loci. We also describe a FPAH patient carrying biallelic constitutional missense mutations of BMPR2 who manifested disease at a stage and manner similar to heterozygous patients. CONCLUSIONS: Taken together, these data demonstrate that MSI is uncommon in FPAH and suggest that somatic loss of the remaining wild-type BMPR2 allele in heterozygous mutation carriers likely does not play a significant role in modulating the onset or progression of FPAH.


Asunto(s)
Predisposición Genética a la Enfermedad , Hipertensión Pulmonar/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética , Adulto , Alelos , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Análisis Mutacional de ADN , Progresión de la Enfermedad , Mutación de Línea Germinal , Humanos , Hipertensión Pulmonar/patología , Pérdida de Heterocigocidad , Pulmón/cirugía , Microdisección , Repeticiones de Microsatélite , Mutación Missense
19.
Nat Med ; 21(7): 777-85, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26076038

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Endoteliales/metabolismo , Factor 2 de Diferenciación de Crecimiento/farmacología , Hipertensión Pulmonar/patología , Arteria Pulmonar/patología , Envejecimiento/patología , Animales , Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Densitometría , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Immunoblotting , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones Endogámicos C57BL , Monocrotalina , Fosforilación/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Sístole/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
20.
Circ Cardiovasc Genet ; 8(4): 572-581, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25963545

RESUMEN

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.


Asunto(s)
Displasia Ectodérmica/genética , Predisposición Genética a la Enfermedad/genética , Haploinsuficiencia , Cardiopatías Congénitas/genética , Deformidades Congénitas de las Extremidades/genética , Receptor Notch1/genética , Dermatosis del Cuero Cabelludo/congénito , Adolescente , Adulto , Secuencia de Bases , Niño , Exoma/genética , Salud de la Familia , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Linaje , Estructura Terciaria de Proteína , Receptor Notch1/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dermatosis del Cuero Cabelludo/genética , Análisis de Secuencia de ADN/métodos , Transducción de Señal/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA