1
|
D'Alto M, Badagliacca R, Airò E, Ameri P, Argiento P, Garascia A, Lombardi CM, Mulè M, Raineri C, Scelsi L, Vizza CD, Ghio S. Gaps in evidence in the management of patients with intermediate-risk pulmonary arterial hypertension: Considerations following the ESC/ERS 2022 guidelines. Vascul Pharmacol 2024; 155:107374. [PMID: 38642596 DOI: 10.1016/j.vph.2024.107374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024]
Abstract
A comprehensive evaluation of risk, using multiple indices, is necessary to provide reliable prognostic information and guide therapy in pulmonary arterial hypertension (PAH). The current ESC/ERS guidelines suggest using a three-strata model for incident (newly diagnosed) patients and a four-strata model for prevalent patients with PAH. The four-strata model serves as a fundamental risk-stratification tool and relies on a minimal dataset of indicators that must be considered during follow-up. Nevertheless, there are still areas of vagueness and ambiguity when classifying and managing patients in the intermediate-risk category. For these patients, considerations should include right heart imaging, hemodynamics, as well as individual factors such as age, sex, genetic profile, disease type, comorbidities, and kidney function. The aim of this report is to present case studies, with a specific focus on patients ultimately classified as intermediate risk. We aim to emphasize the challenges and complexities encountered in the realms of diagnosis, classification, and treatment for these particular patients.
Collapse
Affiliation(s)
- Michele D'Alto
- Department of Cardiology, Monaldi Hospital, Naples, Italy
| | - Roberto Badagliacca
- Department of Scienze Cliniche Internistiche, Anestesiologiche e Cardiologiche, Università "La Sapienza", Roma, Italy
| | - Edoardo Airò
- UO Pneumologia - Fondazione Toscana "G.Monasterio" - CNR/Regione Toscana, Pisa, Italy
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genova, Italy; Cardiac, Thoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paola Argiento
- U.O.C. Cardiologia pediatrica e UTIC, A.O.R.N. dei colli - Ospedale Monaldi, Napoli, Italy
| | - Andrea Garascia
- Cardiologia 2 Insufficienza Cardiaca e Trapianti, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Carlo Mario Lombardi
- Institute of Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Massimiliano Mulè
- Clinical Cardiology and Heart Failure Unit, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Claudia Raineri
- Department of Cardiology, Città della salute- Molinette Hospital, Turin, Italy
| | - Laura Scelsi
- Fondazione I.R.C.C.S. Policlinico San Matteo Pavia, Pavia, Italy
| | - Carmine Dario Vizza
- Department of Scienze Cliniche Internistiche, Anestesiologiche e Cardiologiche, Università "La Sapienza", Roma, Italy
| | - Stefano Ghio
- UOC Cardiologia 1, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy.
| |
Collapse
|
2
|
Boucetta H, Zhang L, Sosnik A, He W. Pulmonary arterial hypertension nanotherapeutics: New pharmacological targets and drug delivery strategies. J Control Release 2024; 365:236-258. [PMID: 37972767 DOI: 10.1016/j.jconrel.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare, serious, and incurable disease characterized by high lung pressure. PAH-approved drugs based on conventional pathways are still not exhibiting favorable therapeutic outcomes. Drawbacks like short half-lives, toxicity, and teratogenicity hamper effectiveness, clinical conventionality, and long-term safety. Hence, approaches like repurposing drugs targeting various and new pharmacological cascades and/or loaded in non-toxic/efficient nanocarrier systems are being investigated lately. This review summarizes the status of conventional, repurposed, either in vitro, in vivo, and/or in clinical trials of PAH treatment. In-depth description, discussion, and classification of the new pharmacological targets and nanomedicine strategies with a description of all the nanocarriers that showed promising efficiency in delivering drugs are discussed. Ultimately, an illustration of the different nucleic acids tailored and nanoencapsulated within different types of nanocarriers to restore the pathways affected by this disease is presented.
Collapse
Affiliation(s)
- Hamza Boucetta
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
3
|
Montani D, Eichstaedt CA, Belge C, Chung WK, Gräf S, Grünig E, Humbert M, Quarck R, Tenorio-Castano JA, Soubrier F, Trembath RC, Morrell NW. [Genetic counselling and testing in pulmonary arterial hypertension - A consensus statement on behalf of the International Consortium for Genetic Studies in PAH - French version]. Rev Mal Respir 2023; 40:838-852. [PMID: 37923650 DOI: 10.1016/j.rmr.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/11/2023] [Indexed: 11/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease that can be caused by (likely) pathogenic germline genomic variants. In addition to the most prevalent disease gene, BMPR2 (bone morphogenetic protein receptor 2), several genes, some belonging to distinct functional classes, are also now known to predispose to the development of PAH. As a consequence, specialist and non-specialist clinicians and healthcare professionals are increasingly faced with a range of questions regarding the need for, approaches to and benefits/risks of genetic testing for PAH patients and/or related family members. We provide a consensus-based approach to recommendations for genetic counselling and assessment of current best practice for disease gene testing. We provide a framework and the type of information to be provided to patients and relatives through the process of genetic counselling, and describe the presently known disease causal genes to be analysed. Benefits of including molecular genetic testing within the management protocol of patients with PAH include the identification of individuals misclassified by other diagnostic approaches, the optimisation of phenotypic characterisation for aggregation of outcome data, including in clinical trials, and importantly through cascade screening, the detection of healthy causal variant carriers, to whom regular assessment should be offered.
Collapse
Affiliation(s)
- D Montani
- French Referral Center for Pulmonary Hypertension, Pulmonary Department, hôpital de Bicêtre, AP-HP, université Paris-Saclay, Le Kremlin-Bicêtre, France; Inserm UMR_S999, hôpital Marie-Lannelongue, Le Plessis-Robinson, France.
| | - C A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg, Allemagne; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Allemagne; Laboratory for Molecular Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Allemagne
| | - C Belge
- Department of Chronic Diseases & Metabolism (CHROMETA), Clinical Department of Respiratory Diseases, University Hospitals, Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), University of Leuven, 3000 Leuven, Belgique
| | - W K Chung
- Department of Pediatrics, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, États-Unis
| | - S Gräf
- Department of Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge Biomedical Campus, Cambridge CB2 0BB, Royaume-Uni; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, Royaume-Uni; NIHR BioResource, for Translational Research - Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, Royaume-Uni
| | - E Grünig
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg, Allemagne; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Allemagne
| | - M Humbert
- French Referral Center for Pulmonary Hypertension, Pulmonary Department, hôpital de Bicêtre, AP-HP, université Paris-Saclay, Le Kremlin-Bicêtre, France; Inserm UMR_S999, hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - R Quarck
- Department of Chronic Diseases & Metabolism (CHROMETA), Clinical Department of Respiratory Diseases, University Hospitals, Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), University of Leuven, 3000 Leuven, Belgique
| | - J A Tenorio-Castano
- INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario La Paz, Madrid, Espagne; CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Espagne; ITHACA, European Reference Network, Brussels, Belgique
| | - F Soubrier
- Département de génétique, Inserm UMR_S1166, AP-HP, hôpital Pitié-Salpêtrière, Institute for Cardio-metabolism and Nutrition (ICAN), Sorbonne université, Paris, France
| | - R C Trembath
- Department of Medical & Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, Royaume-Uni
| | - N W Morrell
- Department of Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge Biomedical Campus, Cambridge CB2 0BB, Royaume-Uni; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, Royaume-Uni
| |
Collapse
|
4
|
Eichstaedt CA, Belge C, Chung WK, Gräf S, Grünig E, Montani D, Quarck R, Tenorio-Castano JA, Soubrier F, Trembath RC, Morrell NW. Genetic counselling and testing in pulmonary arterial hypertension: a consensus statement on behalf of the International Consortium for Genetic Studies in PAH. Eur Respir J 2023; 61:2201471. [PMID: 36302552 PMCID: PMC9947314 DOI: 10.1183/13993003.01471-2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease that can be caused by (likely) pathogenic germline genomic variants. In addition to the most prevalent disease gene, BMPR2 (bone morphogenetic protein receptor 2), several genes, some belonging to distinct functional classes, are also now known to predispose to the development of PAH. As a consequence, specialist and non-specialist clinicians and healthcare professionals are increasingly faced with a range of questions regarding the need for, approaches to and benefits/risks of genetic testing for PAH patients and/or related family members. We provide a consensus-based approach to recommendations for genetic counselling and assessment of current best practice for disease gene testing. We provide a framework and the type of information to be provided to patients and relatives through the process of genetic counselling, and describe the presently known disease causal genes to be analysed. Benefits of including molecular genetic testing within the management protocol of patients with PAH include the identification of individuals misclassified by other diagnostic approaches, the optimisation of phenotypic characterisation for aggregation of outcome data, including in clinical trials, and importantly through cascade screening, the detection of healthy causal variant carriers, to whom regular assessment should be offered.
Collapse
Affiliation(s)
- Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Catharina Belge
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), Clinical Department of Respiratory Diseases, University Hospitals, University of Leuven, Leuven, Belgium
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefan Gräf
- Department of Medicine, Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- NIHR BioResource for Translational Research - Rare Diseases, University of Cambridge, Cambridge, UK
| | - Ekkehard Grünig
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - David Montani
- Université Paris-Saclay, AP-HP, French Referral Center for Pulmonary Hypertension, Pulmonary Department, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
- INSERM UMR_S999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Rozenn Quarck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), Clinical Department of Respiratory Diseases, University Hospitals, University of Leuven, Leuven, Belgium
| | - Jair A Tenorio-Castano
- INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER (Centro de Investigación Biomédica en Red de Enfermedades Raras), Madrid, Spain
- ITHACA, European Reference Network, Brussels, Belgium
| | - Florent Soubrier
- Sorbonne Université, AP-HP, Département de Génétique, INSERM UMR_S1166, Sorbonne Université, Institute for Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Richard C Trembath
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Nicholas W Morrell
- Department of Medicine, Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Upton P, Richards S, Bates A, Niederhoffer KY, Morrell NW, Christian S. A rare homozygous missense GDF2 (BMP9) mutation causing PAH in siblings: Does BMP10 status contribute? Am J Med Genet A 2023; 191:228-233. [PMID: 36259599 PMCID: PMC10092753 DOI: 10.1002/ajmg.a.62996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by pathological remodeling of the pulmonary vasculature causing elevated pulmonary artery pressures and ultimately, right ventricular failure from chronic pressure overload. Heterozygous pathogenic GDF2 (encoding bone morphogenetic protein 9 (BMP9)) variants account for some (>1%) adult PAH cases. Only three pediatric PAH cases, harboring homozygous or compound heterozygous variants, are reported to date. Ultra-rare pathogenic GDF2 variants are reported in hereditary hemorrhagic telangiectasia and overlapping disorders characterized by telangiectasias and arteriovenous malformations (AVMs). Here, we present two siblings with PAH homozygous for a GDF2 mutation that impairs BMP9 proprotein processing and reduces growth factor domain availability. We confirm an absence of measurable plasma BMP9 whereas BMP10 levels are detectable and serum-dependent endothelial BMP activity is evident. This contrasts with the absence of activity which we reported in two children with homozygous pathogenic GDF2 nonsense variants, one with PAH and one with pulmonary AVMs, both with telangiectasias, suggesting loss of BMP10 and endothelial BMP activity in the latter may precipitate telangiectasia development. An absence of phenotype in related heterozygous GDF2 variant carriers suggests incomplete penetrance in PAH and AVM-related diseases, indicating that additional somatic and/or genetic modifiers may be necessary for disease precipitation.
Collapse
Affiliation(s)
- Paul Upton
- Department of Medicine, Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Susan Richards
- Pediatric Pulmonary Hypertension Service, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Angela Bates
- Pediatric Pulmonary Hypertension Service, Stollery Children's Hospital, Edmonton, Alberta, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Y Niederhoffer
- Department of Medical Genetics, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Nicholas W Morrell
- Department of Medicine, Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Susan Christian
- Pediatric Pulmonary Hypertension Service, Stollery Children's Hospital, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Dai L, Du L. Genes in pediatric pulmonary arterial hypertension and the most promising BMPR2 gene therapy. Front Genet 2022; 13:961848. [PMID: 36506323 PMCID: PMC9730536 DOI: 10.3389/fgene.2022.961848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare but progressive and lethal vascular disease of diverse etiologies, mainly caused by proliferation of endothelial cells, smooth muscle cells in the pulmonary artery, and fibroblasts, which ultimately leads to right-heart hypertrophy and cardiac failure. Recent genetic studies of childhood-onset PAH report that there is a greater genetic burden in children than in adults. Since the first-identified pathogenic gene of PAH, BMPR2, which encodes bone morphogenetic protein receptor 2, a receptor in the transforming growth factor-β superfamily, was discovered, novel causal genes have been identified and substantially sharpened our insights into the molecular genetics of childhood-onset PAH. Currently, some newly identified deleterious genetic variants in additional genes implicated in childhood-onset PAH, such as potassium channels (KCNK3) and transcription factors (TBX4 and SOX17), have been reported and have greatly updated our understanding of the disease mechanism. In this review, we summarized and discussed the advances of genetic variants underlying childhood-onset PAH susceptibility and potential mechanism, and the most promising BMPR2 gene therapy and gene delivery approaches to treat childhood-onset PAH in the future.
Collapse
|
7
|
Liu D, Xu F, Gao Q, Zhai Z. Case report: Pulmonary arterial hypertension in ENG-related hereditary hemorrhagic telangiectasia. Front Cardiovasc Med 2022; 9:1020762. [DOI: 10.3389/fcvm.2022.1020762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022] Open
Abstract
A young adult woman presented with exertional dyspnea and she had had recurrent epistaxis for more than 10 years. On physical examination, cyanosis was noted on the lips, and telangiectasias were seen on the oral mucosa and fingertips. Routine investigations revealed iron deficiency anemia and slightly elevated bilirubin. The result of right heart catheterization was indicative of pulmonary arterial hypertension (PAH). Pulmonary angiography showed arteriovenous malformations of the left upper pulmonary artery, and anterior cerebral artery malformation was seen in cranial computed tomographic angiogram. Genetic testing revealed that she and her three daughters carried heterozygous variant of ENG c.1195-1196del p.Arg399GlyfsTer2, which is characterized by pulmonary and cerebral arteriovenous malformations. In addition, our patient had pulmonary hypertension (PH) that is commonly associated with ACVRL1 mutations, revealing her phenotype was not consistent with isolated ENG genetic mutations. Here, we report a case with hereditary hemorrhagic telangiectasia (HHT) combined with PAH, which is associated with interesting differential diagnosis and etiological analysis. We have discussed the relationship between PH and HHT and the characteristics of PAH in HHT patients.
Collapse
|
8
|
Bisserier M, Mathiyalagan P, Zhang S, Elmastour F, Dorfmüller P, Humbert M, David G, Tarzami S, Weber T, Perros F, Sassi Y, Sahoo S, Hadri L. Regulation of the Methylation and Expression Levels of the BMPR2 Gene by SIN3a as a Novel Therapeutic Mechanism in Pulmonary Arterial Hypertension. Circulation 2021; 144:52-73. [PMID: 34078089 PMCID: PMC8293289 DOI: 10.1161/circulationaha.120.047978] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 03/17/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Epigenetic mechanisms are critical in the pathogenesis of pulmonary arterial hypertension (PAH). Previous studies have suggested that hypermethylation of the BMPR2 (bone morphogenetic protein receptor type 2) promoter is associated with BMPR2 downregulation and progression of PAH. Here, we investigated for the first time the role of SIN3a (switch-independent 3a), a transcriptional regulator, in the epigenetic mechanisms underlying hypermethylation of BMPR2 in the pathogenesis of PAH. METHODS We used lung samples from PAH patients and non-PAH controls, preclinical mouse and rat PAH models, and human pulmonary arterial smooth muscle cells. Expression of SIN3a was modulated using a lentiviral vector or a siRNA in vitro and a specific adeno-associated virus serotype 1 or a lentivirus encoding for human SIN3a in vivo. RESULTS SIN3a is a known transcriptional regulator; however, its role in cardiovascular diseases, especially PAH, is unknown. It is interesting that we detected a dysregulation of SIN3 expression in patients and in rodent models, which is strongly associated with decreased BMPR2 expression. SIN3a is known to regulate epigenetic changes. Therefore, we tested its role in the regulation of BMPR2 and found that BMPR2 is regulated by SIN3a. It is interesting that SIN3a overexpression inhibited human pulmonary arterial smooth muscle cells proliferation and upregulated BMPR2 expression by preventing the methylation of the BMPR2 promoter region. RNA-sequencing analysis suggested that SIN3a downregulated the expression of DNA and histone methyltransferases such as DNMT1 (DNA methyltransferase 1) and EZH2 (enhancer of zeste 2 polycomb repressive complex 2) while promoting the expression of the DNA demethylase TET1 (ten-eleven translocation methylcytosine dioxygenase 1). Mechanistically, SIN3a promoted BMPR2 expression by decreasing CTCF (CCCTC-binding factor) binding to the BMPR2 promoter. Last, we identified intratracheal delivery of adeno-associated virus serotype human SIN3a to be a beneficial therapeutic approach in PAH by attenuating pulmonary vascular and right ventricle remodeling, decreasing right ventricle systolic pressure and mean pulmonary arterial pressure, and restoring BMPR2 expression in rodent models of PAH. CONCLUSIONS All together, our study unveiled the protective and beneficial role of SIN3a in pulmonary hypertension. We also identified a novel and distinct molecular mechanism by which SIN3a regulates BMPR2 in human pulmonary arterial smooth muscle cells. Our study also identified lung-targeted SIN3a gene therapy using adeno-associated virus serotype 1 as a new promising therapeutic strategy for treating patients with PAH.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prabhu Mathiyalagan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shihong Zhang
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Firas Elmastour
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Dorfmüller
- Hôpital Marie Lannelongue, Department of Pathology, Le Plessis Robinson, France
| | - Marc Humbert
- Université Paris-Sud, and Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France
- Service de Pneumologie et Soins Intensifs Respiratoires and INSERM U999, Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, Paris, France
| | - Gregory David
- New York University School of Medicine, New York, NY, USA
| | - Sima Tarzami
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington DC, USA
| | - Thomas Weber
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Yassine Sassi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susmita Sahoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Aldalaan AM, Ramzan K, Saleemi SA, Weheba I, Alquait L, Abdelsayed A, Alzubi F, Zaytoun H, Alharbi N, Al-Owain M, Imtiaz F. Genetic basis of pulmonary arterial hypertension: a prospective study from a highly inbred population. Pulm Circ 2021; 11:20458940211032057. [PMID: 34377436 PMCID: PMC8323432 DOI: 10.1177/20458940211032057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH), whether idiopathic PAH (IPAH), heritable PAH, or associated with other conditions, is a rare and potentially lethal disease characterized by progressive vascular changes. To date, there is limited data on the genetic basis of PAH in the Arab region, and none from Saudi Arabian patients. This study aims to identify genetic variations and to evaluate the frequency of risk genes associated to PAH, in Saudi Arabian patients. Adult PAH patients, diagnosed with IPAH and pulmonary veno-occlusive disease, of Saudi Arabian origin, were enrolled in this study. Forty-eight patients were subjected to whole-exome sequencing, with screening of 26 genes suggested to be associated with the disease. The median age at diagnosis was 29.5 years of age, with females accounting for 89.5% of our cohort population. Overall, we identified variations in nine genes previously associated with PAH, in 16 patients. Fourteen of these variants have not been described before. Plausible deleterious variants in risk genes were identified in 33.3% (n = 16/48) of our entire cohort and 25% of these cases carried variants in BMPR2 (n = 4/16). Our results highlight the genetic etiology of PAH in Saudi Arabia patients and provides new insights for the genetic diagnosis of familial and IPAH as well as for the identification of the biological pathways of the disease. This will enable the development of new target therapeutic strategies, for a disease with a high rate of morbidity and mortality.
Collapse
Affiliation(s)
- Abdullah M. Aldalaan
- Department of Medicine, King Faisal Specialist Hospital &
Research Center, Riyadh, Saudi Arabia
| | - Khushnooda Ramzan
- Department of Clinical Genomics, Centre of Genomic Medicine,
King Faisal Specialist Hospital & Research Center, Riyadh, Saudi
Arabia
| | - Sarfraz A. Saleemi
- Department of Clinical Genomics, Centre of Genomic Medicine,
King Faisal Specialist Hospital & Research Center, Riyadh, Saudi
Arabia
| | - Ihab Weheba
- Department of Clinical Genomics, Centre of Genomic Medicine,
King Faisal Specialist Hospital & Research Center, Riyadh, Saudi
Arabia
- National Research Centre, Cairo, Egypt
| | - Laila Alquait
- Department of Clinical Genomics, Centre of Genomic Medicine,
King Faisal Specialist Hospital & Research Center, Riyadh, Saudi
Arabia
| | - Abeer Abdelsayed
- Department of Medicine, King Faisal Specialist Hospital &
Research Center, Riyadh, Saudi Arabia
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Fatima Alzubi
- Department of Medicine, King Faisal Specialist Hospital &
Research Center, Riyadh, Saudi Arabia
| | - Hamdeia Zaytoun
- Department of Medicine, King Faisal Specialist Hospital &
Research Center, Riyadh, Saudi Arabia
| | - Nadeen Alharbi
- Department of Medicine, King Faisal Specialist Hospital &
Research Center, Riyadh, Saudi Arabia
| | - Mohammed Al-Owain
- Department of Medical Genetics, King Faisal Specialist Hospital
& Research Center, Riyadh, Saudi Arabia
| | - Faiqa Imtiaz
- Department of Clinical Genomics, Centre of Genomic Medicine,
King Faisal Specialist Hospital & Research Center, Riyadh, Saudi
Arabia
| |
Collapse
|
10
|
Mukherjee D, Konduri GG. Pediatric Pulmonary Hypertension: Definitions, Mechanisms, Diagnosis, and Treatment. Compr Physiol 2021; 11:2135-2190. [PMID: 34190343 PMCID: PMC8289457 DOI: 10.1002/cphy.c200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pediatric pulmonary hypertension (PPH) is a multifactorial disease with diverse etiologies and presenting features. Pulmonary hypertension (PH), defined as elevated pulmonary artery pressure, is the presenting feature for several pulmonary vascular diseases. It is often a hidden component of other lung diseases, such as cystic fibrosis and bronchopulmonary dysplasia. Alterations in lung development and genetic conditions are an important contributor to pediatric pulmonary hypertensive disease, which is a distinct entity from adult PH. Many of the causes of pediatric PH have prenatal onset with altered lung development due to maternal and fetal conditions. Since lung growth is altered in several conditions that lead to PPH, therapy for PPH includes both pulmonary vasodilators and strategies to restore lung growth. These strategies include optimal alveolar recruitment, maintaining physiologic blood gas tension, nutritional support, and addressing contributing factors, such as airway disease and gastroesophageal reflux. The outcome for infants and children with PH is highly variable and largely dependent on the underlying cause. The best outcomes are for neonates with persistent pulmonary hypertension (PPHN) and reversible lung diseases, while some genetic conditions such as alveolar capillary dysplasia are lethal. © 2021 American Physiological Society. Compr Physiol 11:2135-2190, 2021.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| | - Girija G. Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| |
Collapse
|
11
|
Bofarid S, Hosman AE, Mager JJ, Snijder RJ, Post MC. Pulmonary Vascular Complications in Hereditary Hemorrhagic Telangiectasia and the Underlying Pathophysiology. Int J Mol Sci 2021; 22:3471. [PMID: 33801690 PMCID: PMC8038106 DOI: 10.3390/ijms22073471] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022] Open
Abstract
In this review, we discuss the role of transforming growth factor-beta (TGF-β) in the development of pulmonary vascular disease (PVD), both pulmonary arteriovenous malformations (AVM) and pulmonary hypertension (PH), in hereditary hemorrhagic telangiectasia (HHT). HHT or Rendu-Osler-Weber disease is an autosomal dominant genetic disorder with an estimated prevalence of 1 in 5000 persons and characterized by epistaxis, telangiectasia and AVMs in more than 80% of cases, HHT is caused by a mutation in the ENG gene on chromosome 9 encoding for the protein endoglin or activin receptor-like kinase 1 (ACVRL1) gene on chromosome 12 encoding for the protein ALK-1, resulting in HHT type 1 or HHT type 2, respectively. A third disease-causing mutation has been found in the SMAD-4 gene, causing a combination of HHT and juvenile polyposis coli. All three genes play a role in the TGF-β signaling pathway that is essential in angiogenesis where it plays a pivotal role in neoangiogenesis, vessel maturation and stabilization. PH is characterized by elevated mean pulmonary arterial pressure caused by a variety of different underlying pathologies. HHT carries an additional increased risk of PH because of high cardiac output as a result of anemia and shunting through hepatic AVMs, or development of pulmonary arterial hypertension due to interference of the TGF-β pathway. HHT in combination with PH is associated with a worse prognosis due to right-sided cardiac failure. The treatment of PVD in HHT includes medical or interventional therapy.
Collapse
Affiliation(s)
- Sala Bofarid
- Department of Cardiology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands;
| | - Anna E. Hosman
- Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (A.E.H.); (J.J.M.); (R.J.S.)
| | - Johannes J. Mager
- Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (A.E.H.); (J.J.M.); (R.J.S.)
| | - Repke J. Snijder
- Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (A.E.H.); (J.J.M.); (R.J.S.)
| | - Marco C. Post
- Department of Cardiology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands;
- Department of Cardiology, University Medical Center Utrecht, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
12
|
Egom EEA, Moyou-Somo R, Essame Oyono JL, Kamgang R. Identifying Potential Mutations Responsible for Cases of Pulmonary Arterial Hypertension. APPLICATION OF CLINICAL GENETICS 2021; 14:113-124. [PMID: 33732008 PMCID: PMC7958998 DOI: 10.2147/tacg.s260755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/18/2021] [Indexed: 01/09/2023]
Abstract
Pulmonary Arterial Hypertension (PAH) is a progressive and devastating disease for which there is an escalating body of genetic and related pathophysiological information on disease pathobiology. Nevertheless, the success to date in identifying susceptibility genes, genetic variants and epigenetic processes has been limited due to PAH clinical multi-faceted variations. A number of germline gene candidates have been proposed but demonstrating consistently the association with PAH has been problematic, at least partly due to the reduced penetrance and variable expressivity. Although the data for bone morphogenetic protein receptor type 2 (BMPR2) and related genes remains undoubtedly the most extensive, recent advanced gene sequencing technologies have facilitated the discovery of further gene candidates with mutations among those with and without familial forms of PAH. An in depth understanding of the multitude of biologic variations associated with PAH may provide novel opportunities for therapeutic intervention in the coming years. This knowledge will irrevocably provide the opportunity for improved patient and family counseling as well as improved PAH diagnosis, risk assessment, and personalized treatment.
Collapse
Affiliation(s)
- Emmanuel Eroume-A Egom
- Institut du Savoir Montfort (ISM), Hôpital Montfort, Ottawa, ON, Canada.,Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon.,Reflex Medical Centre Cardiac Diagnostics, Reflex Medical Centre, Mississauga, ON, Canada
| | - Roger Moyou-Somo
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| | - Jean Louis Essame Oyono
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| | - Rene Kamgang
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| |
Collapse
|
13
|
mTOR Signaling in Pulmonary Vascular Disease: Pathogenic Role and Therapeutic Target. Int J Mol Sci 2021; 22:ijms22042144. [PMID: 33670032 PMCID: PMC7926633 DOI: 10.3390/ijms22042144] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease without a cure. The exact pathogenic mechanisms of PAH are complex and poorly understood, yet a number of abnormally expressed genes and regulatory pathways contribute to sustained vasoconstriction and vascular remodeling of the distal pulmonary arteries. Mammalian target of rapamycin (mTOR) is one of the major signaling pathways implicated in regulating cell proliferation, migration, differentiation, and protein synthesis. Here we will describe the canonical mTOR pathway, structural and functional differences between mTOR complexes 1 and 2, as well as the crosstalk with other important signaling cascades in the development of PAH. The pathogenic role of mTOR in pulmonary vascular remodeling and sustained vasoconstriction due to its contribution to proliferation, migration, phenotypic transition, and gene regulation in pulmonary artery smooth muscle and endothelial cells will be discussed. Despite the progress in our elucidation of the etiology and pathogenesis of PAH over the two last decades, there is a lack of effective therapeutic agents to treat PAH patients representing a significant unmet clinical need. In this review, we will explore the possibility and therapeutic potential to use inhibitors of mTOR signaling cascade to treat PAH.
Collapse
|
14
|
At the X-Roads of Sex and Genetics in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:genes11111371. [PMID: 33233517 PMCID: PMC7699559 DOI: 10.3390/genes11111371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
Group 1 pulmonary hypertension (pulmonary arterial hypertension; PAH) is a rare disease characterized by remodeling of the small pulmonary arteries leading to progressive elevation of pulmonary vascular resistance, ultimately leading to right ventricular failure and death. Deleterious mutations in the serine-threonine receptor bone morphogenetic protein receptor 2 (BMPR2; a central mediator of bone morphogenetic protein (BMP) signaling) and female sex are known risk factors for the development of PAH in humans. In this narrative review, we explore the complex interplay between the BMP and estrogen signaling pathways, and the potentially synergistic mechanisms by which these signaling cascades increase the risk of developing PAH. A comprehensive understanding of these tangled pathways may reveal therapeutic targets to prevent or slow the progression of PAH.
Collapse
|
15
|
Haarman MG, Kerstjens-Frederikse WS, Vissia-Kazemier TR, Breeman KTN, Timens W, Vos YJ, Roofthooft MTR, Hillege HL, Berger RMF. The Genetic Epidemiology of Pediatric Pulmonary Arterial Hypertension. J Pediatr 2020; 225:65-73.e5. [PMID: 32502478 DOI: 10.1016/j.jpeds.2020.05.051] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/27/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To describe the prevalence of pulmonary arterial hypertension (PAH)-associated gene mutations, and other genetic characteristics in a national cohort of children with PAH from the Dutch National registry and to explore genotype-phenotype associations and outcomes. STUDY DESIGN Children (n = 70) diagnosed with idiopathic PAH, heritable PAH, PAH associated with congenital heart disease with coincidental shunt (PAH-congenital heart disease group 3), PAH after closure of a cardiac shunt (PAH-congenital heart disease group 4), or PAH associated with other noncardiac conditions were enrolled. Targeted next-generation sequencing was performed on PAH-associated genes (BMPR2, ACVRL1, EIF2AK4, CAV1, ENG, KCNK3, SMAD9, and TBX4). Also, children were tested for specific genetic disorders in case of clinical suspicion. Additionally, children were tested for copy number variations. RESULTS Nineteen children (27%) had a PAH-associated gene mutation/variant: BMPR2 n = 7, TBX4 n = 8, ACVRL1 n = 1, KCNK3 n = 1, and EIF2AK4 n = 2. Twelve children (17%) had a genetic disorder with an established association with PAH (including trisomy 21 and cobalamin C deficiency). In another 16 children (23%), genetic disorders without an established association with PAH were identified (including Noonan syndrome, Beals syndrome, and various copy number variations). Survival rates differed between groups and was most favorable in TBX4 variant carriers. CONCLUSIONS Children with PAH show a high prevalence of genetic disorders, not restricted to established PAH-associated genes. Genetic architecture could play a role in risk-stratified care management in pediatric PAH.
Collapse
Affiliation(s)
- Meindina G Haarman
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands.
| | | | - Theresia R Vissia-Kazemier
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | - Karel T N Breeman
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Yvonne J Vos
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marc T R Roofthooft
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | - Hans L Hillege
- Department of Epidemiology, University Medical Center Groningen, Groningen, the Netherlands; Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
16
|
Jang AY, Kim BG, Kwon S, Seo J, Kim HK, Chang HJ, Chang SA, Cho GY, Rhee SJ, Jung HO, Kim KH, Seo HS, Kim KH, Shin J, Lee JS, Kim M, Lee YJ, Chung WJ. Prevalence and clinical features of bone morphogenetic protein receptor type 2 mutation in Korean idiopathic pulmonary arterial hypertension patients: The PILGRIM explorative cohort. PLoS One 2020; 15:e0238698. [PMID: 32966279 PMCID: PMC7510973 DOI: 10.1371/journal.pone.0238698] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 08/19/2020] [Indexed: 11/18/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a progressive chronic disease with poor outcomes. One reason for poor prognosis is the lack of understanding regarding individual variability in response to treatment. Idiopathic PAH (IPAH) patients with bone morphogenetic protein receptor type 2 (BMPR2) mutations have distinct phenotypes that are crucial for individualized therapy but evidence regarding their prevalence and clinical features in the Korean population is lacking. Therefore, the present study aimed to screen Korean IPAH patients for BMPR2 mutations and analyze their clinical phenotypes. Methods We enrolled 73 unrelated IPAH patients for BMPR2 mutation screening between March 2010 to November 2015 from 11 hospitals in Korea. Thirty-three lineal family members from 6 families of BMPR2 mutation carriers were also screened. Results Among 73 patients, 16 (22%) had BMPR2 mutations. Mutation carriers were younger (27 vs. 47 years; p = 0.02) and had a higher mean pulmonary arterial pressure (mPAP) than non-carriers (64 vs. 51 mmHg; p<0.05). Of the 16 individuals with mutations, 5 deletion, 2 splice-site, 6 nonsense, and 3 missense mutations were found, among which, 9 were newly identified mutation types. Patients less than 30 years old had more BMPR2 mutations (44 vs. 14%; p = 0.04) and a higher mPAP (64 vs. 50 mmHg; p = 0.04) compared with those equaled to or over 30 years old. There were no differences in hemodynamic profiles or the proportion of BMPR2 mutation carriers between groups according to sex. Conclusion The prevalence of BMPR2 mutations in Korean IPAH patients was 22%. Mutation carriers were younger and had a poorer hemodynamic profile compared with the non-carriers. Clinical trial registration Clinicaltrials.gov NCT01054105
Collapse
Affiliation(s)
- Albert Youngwoo Jang
- Department of Cardiovascular Medicine, Gachon University Gil Medical Center, Incheon, Korea
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
| | - Bo-Gyeong Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Sunkoo Kwon
- Department of Cardiovascular Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Jiyoung Seo
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Hyung Kwan Kim
- Division of Cardiology, Section of Cardiovascular Imaging, Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Hyuk-Jae Chang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Sung-A Chang
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Goo-Yeong Cho
- Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Korea
| | - Sang Jae Rhee
- Department of Cardiovascular Medicine, Wonkwang University Hospital, Iksan, Korea
| | - Hae Ok Jung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Kyung-Hee Kim
- Division of Cardiology, Department of Internal Medicine, Sejong General Hospital, Bucheon, Korea
| | - Hye Sun Seo
- Department of Cardiology, Soonchunhyang University Hospital, Bucheon, Korea
| | - Kye Hun Kim
- The Heart Center of Chonnam National University Hospital, Gwangju, Korea
| | - Jinho Shin
- Division of Cardiology, Department of Internal Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Jun Soo Lee
- Department of Cardiovascular Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Minsu Kim
- Department of Cardiovascular Medicine, Gachon University Gil Medical Center, Incheon, Korea
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
| | - Young Jae Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- * E-mail: (WJC); (YJL)
| | - Wook-Jin Chung
- Department of Cardiovascular Medicine, Gachon University Gil Medical Center, Incheon, Korea
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
- * E-mail: (WJC); (YJL)
| |
Collapse
|
17
|
Ge X, Zhu T, Zhang X, Liu Y, Wang Y, Zhang W. Gender differences in pulmonary arterial hypertension patients with BMPR2 mutation: a meta-analysis. Respir Res 2020; 21:44. [PMID: 32028950 PMCID: PMC7006426 DOI: 10.1186/s12931-020-1309-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To investigate the differences in the proportions of BMPR2 mutations in familial hereditary pulmonary arterial hypertension (HPAH) and idiopathic pulmonary arterial hypertension (IPAH) between males and females and the relationship between BMPR2 mutation and PAH severity. METHODS A computer was used to search the electronic Cochrane Library, PubMed/MEDLINE, and EMBASE databases for clinical trials containing information on the relationship between PAH prognosis and BMPR2 mutations through March 2019. After obtaining the data, a meta-analysis was performed using Review Manager Version 5.3 and Stata. RESULTS A meta-analysis was performed on 17 clinical trials (2198 total patients: 644 male, 1554 female). The results showed that among patients with HPAH and IPAH, the BMPR2 mutation rate is higher in male than in female patients [male group (224/644, 34.78%), female group (457/1554, 29.41%), OR = 1.30, 95% CI: 1.06~1.60, P = 0.01, I2 = 10%]. Furthermore, haemodynamic and functional parameters were more severe in IPAH and HPAH patients with BMPR2 mutations than in those without, and those with BMPR2 mutation were diagnosed at a younger age. The risk of death or transplantation was higher in PAH patients with BMPR2 mutations than in those without (OR = 2.51, 95% CI: 1.29~3.57, P = 0.003, I2 = 24%). Furthermore, the difference was significant only in male patients (OR = 5.58, 95% CI: 2.16~14.39, P = 0.0004, I2 = 0%) and not in female patients (OR = 1.41, 95% CI: 0.75~2.67, P = 0.29, I2 = 0%). CONCLUSION Among patients with HPAH and IPAH, men are more likely to have BMPR2 mutations, which may predict more severe PAH indications and prognosis.
Collapse
Affiliation(s)
- Xiaoyue Ge
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Tiantian Zhu
- Teaching and Research Office of Clinical Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xinyi Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Ye Liu
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yonglong Wang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Weifang Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
18
|
Genotypes and Phenotypes of Chinese Pediatric Patients With Idiopathic and Heritable Pulmonary Arterial Hypertension—A Single-Center Study. Can J Cardiol 2019; 35:1851-1856. [DOI: 10.1016/j.cjca.2019.07.628] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022] Open
|
19
|
Galambos C, Mullen MP, Shieh JT, Schwerk N, Kielt MJ, Ullmann N, Boldrini R, Stucin-Gantar I, Haass C, Bansal M, Agrawal PB, Johnson J, Peca D, Surace C, Cutrera R, Pauciulo MW, Nichols WC, Griese M, Ivy D, Abman SH, Austin ED, Danhaive O. Phenotype characterisation of TBX4 mutation and deletion carriers with neonatal and paediatric pulmonary hypertension. Eur Respir J 2019; 54:13993003.01965-2018. [PMID: 31151956 DOI: 10.1183/13993003.01965-2018] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/19/2019] [Indexed: 11/05/2022]
Abstract
Rare variants in the T-box transcription factor 4 gene (TBX4) have recently been recognised as an emerging cause of paediatric pulmonary hypertension (PH). Their pathophysiology and contribution to persistent pulmonary hypertension in neonates (PPHN) are unknown. We sought to define the spectrum of clinical manifestations and histopathology associated with TBX4 variants in neonates and children with PH.We assessed clinical data and lung tissue in 19 children with PH, including PPHN, carrying TBX4 rare variants identified by next-generation sequencing and copy number variation arrays.Variants included six 17q23 deletions encompassing the entire TBX4 locus and neighbouring genes, and 12 likely damaging mutations. 10 infants presented with neonatal hypoxic respiratory failure and PPHN, and were subsequently discharged home. PH was diagnosed later in infancy or childhood. Three children died and two required lung transplantation. Associated anomalies included patent ductus arteriosus, septal defects, foot anomalies and developmental disability, the latter with a higher prevalence in deletion carriers. Histology in seven infants showed abnormal distal lung development and pulmonary hypertensive remodelling.TBX4 mutations and 17q23 deletions underlie a new form of developmental lung disease manifesting with severe, often biphasic PH at birth and/or later in infancy and childhood, often associated with skeletal anomalies, cardiac defects, neurodevelopmental disability and other anomalies.
Collapse
Affiliation(s)
- Csaba Galambos
- Dept of Pathology and Laboratory Services, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA.,Contributed equally to this work as joint first authors
| | - Mary P Mullen
- Dept of Cardiology, Boston Children's Hospital and Pediatrics Harvard Medical School, Boston, MA, USA.,Contributed equally to this work as joint first authors
| | - Joseph T Shieh
- Institute for Human Genetics, Medical Genetics, University of California San Francisco, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Nicolaus Schwerk
- Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Matthew J Kielt
- Division of Pediatric Pulmonology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicola Ullmann
- Division of Pediatric Pulmonology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Renata Boldrini
- Dept of Laboratory Medicine, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Cristina Haass
- Division of Neonatology, San Pietro-Fatebenefratelli Hospital, Rome, Italy
| | - Manish Bansal
- Division of Pediatric Cardiology, University of Iowa Children's Hospital, Iowa City, IA, USA
| | - Pankaj B Agrawal
- Division of Newborn Medicine and Genetics and Genomics, Harvard School of Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Joyce Johnson
- Dept of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Donatella Peca
- Dept of Laboratory Medicine, Bambino Gesù Children's Hospital, Rome, Italy
| | - Cecilia Surace
- Dept of Laboratory Medicine, Bambino Gesù Children's Hospital, Rome, Italy
| | - Renato Cutrera
- Division of Pediatric Pulmonology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Dept of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Dept of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthias Griese
- Division of Pediatric Pulmonology, Ludwig-Maximilians-University, Munich, Germany
| | - Dunbar Ivy
- Division of Pediatric Cardiology, Dept of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Steven H Abman
- Division of Pulmonary Medicine, Dept of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Eric D Austin
- Division of Pediatric Pulmonology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Olivier Danhaive
- Division of Neonatology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA, USA .,Division of Neonatology, Catholic University of Louvain, Brussels, Belgium
| |
Collapse
|
20
|
Wang XJ, Lian TY, Jiang X, Liu SF, Li SQ, Jiang R, Wu WH, Ye J, Cheng CY, Du Y, Xu XQ, Wu Y, Peng FH, Sun K, Mao YM, Yu H, Liang C, Shyy JYJ, Zhang SY, Zhang X, Jing ZC. Germline BMP9 mutation causes idiopathic pulmonary arterial hypertension. Eur Respir J 2019; 53:13993003.01609-2018. [PMID: 30578397 DOI: 10.1183/13993003.01609-2018] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/29/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Idiopathic pulmonary arterial hypertension (IPAH) is a rare disease with high heritability. Although several predisposing genes have been linked to IPAH, the genetic aetiology remains unknown for a large number of IPAH cases. METHODS We conducted an exome-wide gene-based burden analysis on two independent case-control studies, including a total of 331 IPAH cases and 10 508 controls. Functional assessments were conducted to analyse the effects of genetic mutations on protein biosynthesis and function. RESULTS The gene encoding human bone morphogenetic protein 9 (BMP9) was identified as a novel genetic locus displaying exome-wide association with IPAH in the discovery cohort (OR 18.8; p=1.9×10-11). This association was authenticated in the independent replication cohort (p=1.0×10-5). Collectively, the rare coding mutations in BMP9 occurred in 6.7% of cases, ranking this gene second to BMPR2, comprising a combined significance of 2.7×10-19 (OR 21.2). Intriguingly, the patients with BMP9 mutations had lower plasma levels of BMP9 than those without. Functional studies showed that the BMP9 mutations led to reduced BMP9 secretion and impaired anti-apoptosis ability in pulmonary arterial endothelial cells. CONCLUSION We identify BMP9 as an IPAH culprit gene.
Collapse
Affiliation(s)
- Xiao-Jian Wang
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,These two authors contributed equally to this work
| | - Tian-Yu Lian
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,These two authors contributed equally to this work
| | - Xin Jiang
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shao-Fei Liu
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Su-Qi Li
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong Jiang
- Dept of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Hui Wu
- Dept of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jue Ye
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chun-Yan Cheng
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yao Du
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi-Qi Xu
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Wu
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fu-Hua Peng
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Sun
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Min Mao
- Dept of Respiratory Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Huan Yu
- Novogene Co., Ltd, Beijing, China
| | | | - John Y-J Shyy
- Dept of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Shu-Yang Zhang
- Dept of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Cheng Jing
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Rosenzweig EB, Abman SH, Adatia I, Beghetti M, Bonnet D, Haworth S, Ivy DD, Berger RMF. Paediatric pulmonary arterial hypertension: updates on definition, classification, diagnostics and management. Eur Respir J 2019; 53:1801916. [PMID: 30545978 PMCID: PMC6351335 DOI: 10.1183/13993003.01916-2018] [Citation(s) in RCA: 359] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022]
Abstract
Paediatric pulmonary arterial hypertension (PAH) shares common features of adult disease, but is associated with several additional disorders and challenges that require unique approaches. This article discusses recent advances, ongoing challenges and distinct approaches for the care of children with PAH, as presented by the Paediatric Task Force of the 6th World Symposium on Pulmonary Hypertension. We provide updates of the current definition, epidemiology, classification, diagnostics and treatment of paediatric PAH, and identify critical knowledge gaps. Several features of paediatric PAH including the prominence of neonatal PAH, especially in pre-term infants with developmental lung diseases, and novel genetic causes of paediatric PAH are highlighted. The use of cardiac catheterisation as a diagnostic modality and haemodynamic definitions of PAH, including acute vasoreactivity, are addressed. Updates are provided on issues related to utility of the previous classification system to reflect paediatric-specific aetiologies and approaches to medical and interventional management of PAH, including the Potts shunt. Although a lack of clinical trial data for the use of PAH-targeted therapy persists, emerging data are improving the identification of appropriate targets for goal-oriented therapy in children. Such data will likely improve future clinical trial design to enhance outcomes in paediatric PAH.
Collapse
Affiliation(s)
- Erika B Rosenzweig
- Columbia University Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - Steven H Abman
- University of Colorado, Children's Hospital Colorado, Denver, CO, USA
| | - Ian Adatia
- Glenwood Children's Heart Clinic, Edmonton, AB, Canada
| | - Maurice Beghetti
- Pediatric Cardiology Unit and Centre Universitaire de Cardiologie et Chirurgie Cardiaque Pédiatrique, University Hospitals of Geneva and Lausanne, Lausanne, Switzerland
| | - Damien Bonnet
- Hôpital Necker Enfants Malades, AP-HP, Université Paris Descartes, Paris, France
| | | | - D Dunbar Ivy
- University of Colorado, Children's Hospital Colorado, Denver, CO, USA
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Savale L, Guignabert C, Weatherald J, Humbert M. Precision medicine and personalising therapy in pulmonary hypertension: seeing the light from the dawn of a new era. Eur Respir Rev 2018; 27:27/148/180004. [PMID: 29653948 PMCID: PMC9488842 DOI: 10.1183/16000617.0004-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023] Open
Abstract
Pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH) include different cardiopulmonary disorders in which the interaction of multiple genes with environmental and behavioural factors modulates the onset and the progression of these severe conditions. Although the development of therapeutic agents that modulate abnormalities in three major pathobiological pathways for PAH has revolutionised our approach to the treatment of PAH, the long-term survival rate remains unsatisfactory. Accumulating evidence has underlined that clinical outcomes and responses to therapy in PAH are modified by multiple factors, including genetic variations, which will be different for each individual. Since precision medicine, also known as stratified medicine or personalised medicine, aims to better target intervention to the individual while maximising benefit and minimising harm, it has significant potential advantages. This article aims to assemble and discuss the different initiatives that are currently underway in the PH/PAH fields together with the opportunities and prospects for their use in the near future. Development of precision medicine strategies will be the next frontier in the evolution of PAH treatmenthttp://ow.ly/8T8730j7e36
Collapse
|
23
|
Cirulis MM, Ryan JJ. Who’s Who of Pulmonary Hypertension. Circ Genom Precis Med 2018; 11:e002116. [DOI: 10.1161/circgen.118.002116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Meghan M. Cirulis
- Division of Pulmonary Medicine (M.M.C.) and Division of Cardiovascular Medicine (J.J.R.), Department of Medicine, University of Utah, Salt Lake City
| | - John J. Ryan
- Division of Pulmonary Medicine (M.M.C.) and Division of Cardiovascular Medicine (J.J.R.), Department of Medicine, University of Utah, Salt Lake City
| |
Collapse
|
24
|
Zhu N, Gonzaga-Jauregui C, Welch C, Ma L, Qi H, King AK, Krishnan U, Rosenzweig EB, Ivy DD, Austin ED, Hamid R, Nichols WC, Pauciulo MW, Lutz KA, Sawle A, Reid JG, Overton JD, Baras A, Dewey F, Shen Y, Chung WK. Exome Sequencing in Children With Pulmonary Arterial Hypertension Demonstrates Differences Compared With Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e001887. [PMID: 29631995 PMCID: PMC5896781 DOI: 10.1161/circgen.117.001887] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/31/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a rare disease characterized by pulmonary arteriole remodeling, elevated arterial pressure and resistance, and subsequent heart failure. Compared with adult-onset disease, pediatric-onset PAH is more heterogeneous and often associated with worse prognosis. Although BMPR2 mutations underlie ≈70% of adult familial PAH (FPAH) cases, the genetic basis of PAH in children is less understood. METHODS We performed genetic analysis of 155 pediatric- and 257 adult-onset PAH patients, including both FPAH and sporadic, idiopathic PAH (IPAH). After screening for 2 common PAH risk genes, mutation-negative FPAH and all IPAH cases were evaluated by exome sequencing. RESULTS We observed similar frequencies of rare, deleterious BMPR2 mutations in pediatric- and adult-onset patients: ≈55% in FPAH and 10% in IPAH patients in both age groups. However, there was significant enrichment of TBX4 mutations in pediatric- compared with adult-onset patients (IPAH: 10/130 pediatric versus 0/178 adult-onset), and TBX4 carriers had younger mean age-of-onset compared with BMPR2 carriers. Mutations in other known PAH risk genes were infrequent in both age groups. Notably, among pediatric IPAH patients without mutations in known risk genes, exome sequencing revealed a 2-fold enrichment of de novo likely gene-damaging and predicted deleterious missense variants. CONCLUSIONS Mutations in known PAH risk genes accounted for ≈70% to 80% of FPAH in both age groups, 21% of pediatric-onset IPAH, and 11% of adult-onset IPAH. Rare, predicted deleterious variants in TBX4 are enriched in pediatric patients and de novo variants in novel genes may explain ≈19% of pediatric-onset IPAH cases.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Medical Center, New York
- Department of Systems Biology, Columbia University, New York, NY
| | | | - Carrie Welch
- Department of Pediatrics, Columbia University Medical Center, New York
| | - Lijiang Ma
- Department of Pediatrics, Columbia University Medical Center, New York
| | - Hongjian Qi
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, NY
- Department of Systems Biology, Columbia University, New York, NY
| | | | - Usha Krishnan
- Department of Pediatrics, Columbia University Medical Center, New York
| | - Erika B. Rosenzweig
- Department of Pediatrics, Columbia University Medical Center, New York
- Department of Medicine, Columbia University Medical Center, New York
| | - D. Dunbar Ivy
- Children’s Hospital Colorado, Department of Pediatric Cardiology, Denver, CO
| | - Eric D. Austin
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN
| | - Rizwan Hamid
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center & Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Michael W. Pauciulo
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center & Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Katie A. Lutz
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center & Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Ashley Sawle
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York
| | - Jeffrey G. Reid
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown
| | - John D. Overton
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown
| | - Aris Baras
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown
| | - Frederick Dewey
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY
- Department of Biomedical Informatics, Columbia University, New York, NY
| | - Wendy K. Chung
- Department of Pediatrics, Columbia University Medical Center, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York
- Department of Medicine, Columbia University Medical Center, New York
| |
Collapse
|
25
|
Frank BS, Ivy DD. Diagnosis, Evaluation and Treatment of Pulmonary Arterial Hypertension in Children. CHILDREN (BASEL, SWITZERLAND) 2018; 5:E44. [PMID: 29570688 PMCID: PMC5920390 DOI: 10.3390/children5040044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/28/2018] [Accepted: 03/16/2018] [Indexed: 12/23/2022]
Abstract
Pulmonary Hypertension (PH), the syndrome of elevated pressure in the pulmonary arteries, is associated with significant morbidity and mortality for affected children. PH is associated with a wide variety of potential underlying causes, including cardiac, pulmonary, hematologic and rheumatologic abnormalities. Regardless of the cause, for many patients the natural history of PH involves progressive elevation in pulmonary arterial resistance and pressure, right ventricular dysfunction, and eventually heart failure. In recent years, a number of pulmonary arterial hypertension (PAH)-targeted therapies have become available to reduce pulmonary artery pressure and improve outcome. A growing body of evidence in both the adult and pediatric literature demonstrates enhanced quality of life, functional status, and survival among treated patients. This review provides a description of select etiologies of PH seen in pediatrics and an update on the most recent data pertaining to evaluation and management of children with PH/PAH. The available evidence for specific classes of PAH-targeted therapies in pediatrics is additionally discussed.
Collapse
Affiliation(s)
- Benjamin S Frank
- Section of Cardiology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA.
| | - D Dunbar Ivy
- Section of Cardiology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
26
|
Zeng X, Lian T, Lin J, Li S, Zheng H, Cheng C, Ye J, Jing Z, Wang X, Huang W. Whole-exome sequencing improves genetic testing accuracy in pulmonary artery hypertension. Pulm Circ 2018; 8:2045894018763682. [PMID: 29480072 PMCID: PMC5858635 DOI: 10.1177/2045894018763682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Sanger sequencing, the traditional “gold standard” for mutation detection, has been wildly used in genetic testing of pulmonary artery hypertension (PAH). However, with the advent of whole-exome sequencing (WES), few studies have compared the accuracy of WES and Sanger sequencing in routine genetic testing of PAH. PAH individuals were enrolled from Fu Wai Hospital and Shanghai Pulmonary Hospital. WES was used to analyze DNA samples from 120 PAH patients whose bone morphogenetic protein receptor type 2 (BMPR2) mutation statuses had been previously studied using Sanger sequencing. The Sanger sequencing and WES agreement was 98.3% (118/120) with near-perfect agreement (κ coefficient = 0.848). There was no significant difference between the two methods on the McNemar–Bowker test (P > 0.05). Twenty-one BMPR2 mutation carriers and 99 non-carriers were detected by Sanger sequencing. Among the 21 BMPR2 carriers detected by Sanger sequencing, one variant (c.1040 T > A) was not found by WES. Among the 99 BMPR2 non-carriers, WES detected an extra mutation carrier (c.76 + 1 G > C) missed by Sanger sequencing. Both false-positive and false-negative results were highly conserved and were re-analyzed by Sanger sequencing. WES improved the accuracy of Sanger sequencing and detected 1% (1/99) false-positive and 4.8% (1/21) false-negative results of Sanger sequencing. No false-positive and false-negative results of WES were identified in our analysis. WES is non-inferior to Sanger sequencing and may play a more important role in genetic testing of PAH patients in the future.
Collapse
Affiliation(s)
- Xiaofang Zeng
- 1 Department of Cardiology, the First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Tianyu Lian
- 2 State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Jianhui Lin
- 3 Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, UK
| | - Suqi Li
- 2 State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Haikuo Zheng
- 4 Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chunyan Cheng
- 2 State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Jue Ye
- 2 State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Zhicheng Jing
- 2 State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Xiaojian Wang
- 2 State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Wei Huang
- 1 Department of Cardiology, the First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Pulmonary Hypertension and Thrombembolism—Long-Term Management and Chronic Oral Anticoagulation. PHYSICIAN ASSISTANT CLINICS 2017. [DOI: 10.1016/j.cpha.2017.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Abstract
Tremendous progress has been made in understanding the genetics of pulmonary arterial hypertension (PAH) since its description in the 1950s as a primary disorder of the pulmonary vasculature. Heterozygous germline mutations in the gene coding bone morphogenetic receptor type 2 (BMPR2) are detectable in the majority of cases of heritable PAH, and in approximately 20% of cases of idiopathic pulmonary arterial hypertension (IPAH). However, recent advances in gene discovery methods have facilitated the discovery of additional genes with mutations among those with and without familial PAH. Heritable PAH is an autosomal dominant disease characterized by reduced penetrance, variable expressivity, and female predominance. Biallelic germline mutations in the gene EIF2AK4 are now associated with pulmonary veno-occlusive disease and pulmonary capillary hemangiomatosis. Growing genetic knowledge enhances our capacity to pursue and provide genetic counseling, although the issue remains complex given that the majority of carriers of PAH-related mutations will never be diagnosed with the disease.
Collapse
Affiliation(s)
- Joshua D. Chew
- Division of Cardiology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James E. Loyd
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eric D. Austin
- Division of Pulmonary, Allergy, and Immunology Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
29
|
Girerd B, Weatherald J, Montani D, Humbert M. Heritable pulmonary hypertension: from bench to bedside. Eur Respir Rev 2017; 26:26/145/170037. [DOI: 10.1183/16000617.0037-2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/02/2017] [Indexed: 02/06/2023] Open
Abstract
Mutations in the BMPR2 gene, and more rarely in ACVRL1, endoglin, caveolin-1, KCNK3 and TBX4 genes predispose to heritable pulmonary arterial hypertension, an autosomal dominant disease with incomplete penetrance. Bi-allelic mutations in the EIF2AK4 gene predispose to heritable pulmonary veno-occlusive disease/pulmonary capillary haemangiomatosis, an autosomal recessive disease with an unknown penetrance.In France, the national pulmonary hypertension referral centre offers genetic counselling and testing to adults and children. Predictive testing is also proposed to adult relatives at risk of carrying a predisposing mutation. In that context, we offer all asymptomatic BMPR2 mutation carriers a programme to detect pulmonary arterial hypertension at an early phase, as recommended by the 2015 European Society Society of Cardiology/European Respiratory Society pulmonary hypertension guidelines. Finally, pre-implantation genetic diagnosis has been conducted on five embryos from two couples in which the fathers were carriers of a pathogenic BMPR2 mutation.
Collapse
|
30
|
|
31
|
van der Bruggen CE, Tedford RJ, Handoko ML, van der Velden J, de Man FS. RV pressure overload: from hypertrophy to failure. Cardiovasc Res 2017; 113:1423-1432. [DOI: 10.1093/cvr/cvx145] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/31/2017] [Indexed: 01/31/2023] Open
Affiliation(s)
- Cathelijne E.E. van der Bruggen
- Department of Pulmonology, Amsterdam Cardiovascular Sciences, VU University Medical Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ryan J. Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Frances S. de Man
- Department of Pulmonology, Amsterdam Cardiovascular Sciences, VU University Medical Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
32
|
Talati MH, Brittain EL, Fessel JP, Penner N, Atkinson J, Funke M, Grueter C, Jerome WG, Freeman M, Newman JH, West J, Hemnes AR. Mechanisms of Lipid Accumulation in the Bone Morphogenetic Protein Receptor Type 2 Mutant Right Ventricle. Am J Respir Crit Care Med 2017; 194:719-28. [PMID: 27077479 DOI: 10.1164/rccm.201507-1444oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
RATIONALE In heritable pulmonary arterial hypertension with germline mutation in the bone morphogenetic protein receptor type 2 (BMPR2) gene, right ventricle (RV) dysfunction is associated with RV lipotoxicity; however, the underlying mechanism for lipid accumulation is not known. OBJECTIVES We hypothesized that lipid accumulation in cardiomyocytes with BMPR2 mutation occurs owing to alterations in lipid transport and impaired fatty acid oxidation (FAO), which is exacerbated by a high-lipid (Western) diet (WD). METHODS We used a transgenic mouse model of pulmonary arterial hypertension with mutant BMPR2 and generated a cardiomyocyte cell line with BMPR2 mutation. Electron microscopy and metabolomic analysis were performed on mouse RVs. MEASUREMENTS AND MAIN RESULTS By metabolomics analysis, we found an increase in long-chain fatty acids in BMPR2 mutant mouse RVs compared with controls, which correlated with cardiac index. BMPR2-mutant cardiomyocytes had increased lipid compared with controls. Direct measurement of FAO in the WD-fed BMPR2-mutant RV showed impaired palmitate-linked oxygen consumption, and metabolomics analysis showed reduced indices of FAO. Using both mutant BMPR2 mouse RVs and cardiomyocytes, we found an increase in the uptake of (14)C-palmitate and fatty acid transporter CD36 that was further exacerbated by WD. CONCLUSIONS Taken together, our data suggest that impaired FAO and increased expression of the lipid transporter CD36 are key mechanisms underlying lipid deposition in the BMPR2-mutant RV, which are exacerbated in the presence of dietary lipids. These findings suggest important features leading to RV lipotoxicity in pulmonary arterial hypertension and may point to novel areas of therapeutic intervention.
Collapse
Affiliation(s)
- Megha H Talati
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Joshua P Fessel
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.,3 Department of Pharmacology
| | - Niki Penner
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Mitch Funke
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - W Gray Jerome
- 4 Department of Pathology, Microbiology, and Immunology.,6 Department of Cancer Biology, and
| | - Michael Freeman
- 7 Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John H Newman
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James West
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Anna R Hemnes
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
33
|
Idiopathic Pulmonary Arterial Hypertension in Children: A Review. Pulm Ther 2017. [DOI: 10.1007/s41030-017-0035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
34
|
Which prognostic factors should be used in pulmonary arterial hypertension in elderly patients? JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2017; 14:28-34. [PMID: 28270839 PMCID: PMC5329730 DOI: 10.11909/j.issn.1671-5411.2017.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In recent times, the prevalence of pulmonary arterial hypertension (PAH) is more commonly seen among elderly populations. The increased prevalence of hypertension, diabetes, obesity, arterial stiffness, as well as diastolic dysfunction, may cause endothelial dysfunction and affect pulmonary vasculature. Furthermore, older patients have certain differences in clinical characteristics and outcomes. In this article, the special characteristics of aging in PAH patients have been reviewed, while the risk predictors of elderly patients are also discussed.
Collapse
|
35
|
Guignabert C, Bailly S, Humbert M. Restoring BMPRII functions in pulmonary arterial hypertension: opportunities, challenges and limitations. Expert Opin Ther Targets 2016; 21:181-190. [DOI: 10.1080/14728222.2017.1275567] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Univ. Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Sabine Bailly
- INSERM U1036, Grenoble, France
- Laboratoire Biologie du Cancer et de l’Infection, Commissariat à l’Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
- Université Grenoble-Alpes, Grenoble, France
| | - Marc Humbert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Univ. Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France
| |
Collapse
|
36
|
Ma L, Chung WK. The role of genetics in pulmonary arterial hypertension. J Pathol 2016; 241:273-280. [PMID: 27770446 DOI: 10.1002/path.4833] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 12/14/2022]
Abstract
Group 1 pulmonary hypertension or pulmonary arterial hypertension (PAH) is a rare disease characterized by proliferation and occlusion of small pulmonary arterioles, leading to progressive elevation of pulmonary artery pressure and pulmonary vascular resistance, and right ventricular failure. Historically, it has been associated with a high mortality rate, although, over the last decade, treatment has improved survival. PAH includes idiopathic PAH (IPAH), heritable PAH (HPAH), and PAH associated with certain medical conditions. The aetiology of PAH is heterogeneous, and genetics play an important role in some cases. Mutations in BMPR2, encoding bone morphogenetic protein receptor 2, a member of the transforming growth factor-β superfamily of receptors, have been identified in 70% of cases of HPAH, and in 10-40% of cases of IPAH. Other genetic causes of PAH include mutations in the genes encoding activin receptor-like type 1, endoglin, SMAD9, caveolin 1, and potassium two-pore-domain channel subfamily K member 3. Mutations in the gene encoding T-box 4 have been identified in 10-30% of paediatric PAH patients, but rarely in adults with PAH. PAH in children is much more heterogeneous than in adults, and can be associated with several genetic syndromes, congenital heart disease, pulmonary disease, and vascular disease. In addition to rare mutations as a monogenic cause of HPAH, common variants in the gene encoding cerebellin 2 increase the risk of PAH by approximately two-fold. A PAH panel of genes is available for clinical testing, and should be considered for use in clinical management, especially for patients with a family history of PAH. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lijiang Ma
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| |
Collapse
|
37
|
Best DH, Sumner KL, Smith BP, Damjanovich-Colmenares K, Nakayama I, Brown LM, Ha Y, Paul E, Morris A, Jama MA, Dodson MW, Bayrak-Toydemir P, Elliott CG. EIF2AK4 Mutations in Patients Diagnosed With Pulmonary Arterial Hypertension. Chest 2016; 151:821-828. [PMID: 27884767 DOI: 10.1016/j.chest.2016.11.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/18/2016] [Accepted: 11/04/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Differentiating pulmonary venoocclusive disease (PVOD) and pulmonary capillary hemangiomatosis (PCH) from idiopathic pulmonary arterial hypertension (IPAH) or heritable pulmonary arterial hypertension (HPAH) is important clinically. Mutations in eukaryotic translation initiation factor 2 alpha kinase 4 (EIF2AK4) cause heritable PVOD and PCH, whereas mutations in other genes cause HPAH. The aim of this study was to describe the frequency of pathogenic EIF2AK4 mutations in patients diagnosed clinically with IPAH or HPAH. METHODS Sanger sequencing and deletion/duplication analysis were performed to detect mutations in the bone morphogenetic protein receptor type II (BMPR2) gene in 81 patients diagnosed at 30 North American medical centers with IPAH (n = 72) or HPAH (n = 9). BMPR2 mutation-negative patients (n = 67) were sequenced for mutations in four other genes (ACVRL1, ENG, CAV1, and KCNK3) known to cause HPAH. Patients negative for mutations in all known PAH genes (n = 66) were then sequenced for mutations in EIF2AK4. We assessed the pathogenicity of EIF2AK4 mutations and reviewed clinical characteristics of patients with pathogenic EIF2AK4 mutations. RESULTS Pathogenic BMPR2 mutations were identified in 8 of 72 (11.1%) patients with IPAH and 6 of 9 (66.7%) patients with HPAH. A novel homozygous EIF2AK4 mutation (c.257+4A>C) was identified in 1 of 9 (11.1%) patients diagnosed with HPAH. The novel EIF2AK4 mutation (c.257+4A>C) was homozygous in two sisters with severe pulmonary hypertension. None of the 72 patients with IPAH had biallelic EIF2AK4 mutations. CONCLUSIONS Pathogenic biallelic EIF2AK4 mutations are rarely identified in patients diagnosed with HPAH. Identification of pathogenic biallelic EIF2AK4 mutations can aid clinicians in differentiating HPAH from heritable PVOD or PCH.
Collapse
Affiliation(s)
- D Hunter Best
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT; Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT; Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT
| | - Kelli L Sumner
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT
| | - Benjamin P Smith
- Department of Medicine, Intermountain Medical Center, Murray, UT; Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | | | - Ikue Nakayama
- Department of Medicine, Intermountain Medical Center, Murray, UT
| | - Lynette M Brown
- Department of Medicine, Intermountain Medical Center, Murray, UT; Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Youna Ha
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT
| | - Eleri Paul
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT
| | - Ashley Morris
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT
| | - Mohamed A Jama
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT
| | - Mark W Dodson
- Department of Medicine, Intermountain Medical Center, Murray, UT; Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Pinar Bayrak-Toydemir
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, UT; Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - C Gregory Elliott
- Department of Medicine, Intermountain Medical Center, Murray, UT; Department of Internal Medicine, University of Utah, Salt Lake City, UT.
| |
Collapse
|
38
|
BMPRII influences the response of pulmonary microvascular endothelial cells to inflammatory mediators. Pflugers Arch 2016; 468:1969-1983. [PMID: 27816994 DOI: 10.1007/s00424-016-1899-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/13/2016] [Accepted: 10/24/2016] [Indexed: 10/20/2022]
Abstract
Mutations in the bone morphogenetic protein receptor (BMPR2) gene have been observed in 70 % of patients with heritable pulmonary arterial hypertension (HPAH) and in 11-40 % with idiopathic PAH (IPAH). However, carriers of a BMPR2 mutation have only 20 % risk of developing PAH. Since inflammatory mediators are increased and predict survival in PAH, they could act as a second hit inducing the development of pulmonary hypertension in BMPR2 mutation carriers. Our specific aim was to determine whether inflammatory mediators could contribute to pulmonary vascular cell dysfunction in PAH patients with and without a BMPR2 mutation. Pulmonary microvascular endothelial cells (PMEC) and arterial smooth muscle cells (PASMC) were isolated from lung parenchyma of transplanted PAH patients, carriers of a BMPR2 mutation or not, and from lobectomy patients or lung donors. The effects of CRP and TNFα on mitogenic activity, adhesiveness capacity, and expression of adhesion molecules were investigated in PMECs and PASMCs. PMECs from BMPR2 mutation carriers induced an increase in PASMC mitogenic activity; moreover, endothelin-1 secretion by PMECs from carriers was higher than by PMECs from non-carriers. Recruitment of monocytes by PMECs isolated from carriers was higher compared to PMECs from non-carriers and from controls, with an elevated ICAM-1 expression. CRP increased adhesion of monocytes to PMECs in carriers and non-carriers, and TNFα only in carriers. PMEC from BMPR2 mutation carriers have enhanced adhesiveness for monocytes in response to inflammatory mediators, suggesting that BMPR2 mutation could generate susceptibility to an inflammatory insult in PAH.
Collapse
|
39
|
Levy M, Eyries M, Szezepanski I, Ladouceur M, Nadaud S, Bonnet D, Soubrier F. Genetic analyses in a cohort of children with pulmonary hypertension. Eur Respir J 2016; 48:1118-1126. [PMID: 27587546 DOI: 10.1183/13993003.00211-2016] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/07/2016] [Indexed: 11/05/2022]
Abstract
The prevalence of germline mutations in paediatric pulmonary hypertension (PH) is poorly documented. The objective of this study was to determine the mutation frequency in PH genes in a paediatric cohort and describe the clinical characteristics of mutation carriers.The study involved 66 index cases with PH: 35 children with idiopathic pulmonary arterial hypertension (IPAH); five children with familial PAH (FPAH); three children with pulmonary veno-occlusive disease (PVOD); and 23 children with PAH associated with congenital heart disease (APAH-CHD).No mutations were found in the 23 children with APAH-CHD. In the 40 children with IPAH or FPAH, 12 mutations were found: five on BMPR2; four on ACVRL1; and three on TBX4. In the three PVOD cases, two carried the EIF2AK4 mutation. Mutation carriers had a more severe disease at diagnosis and more aggressive first-line therapy was required. The three patients with PVOD had a very severe disease at diagnosis and required a lung transplantation.The genetic architecture of paediatric PAH is enriched in ACVRL1 and TBX4 mutations compared to adult PAH, but further studies are required to confirm these results. Childhood-onset PAH in children carrying a mutation in one of the genes tested has a more severe presentation at diagnosis but a similar outcome to that observed in non-carriers.
Collapse
Affiliation(s)
- Marilyne Levy
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France M3C-Unité Médico-Chirugicale de Cardiologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Mélanie Eyries
- Genetics Dept, GH Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR_S 1166-ICAN, Paris, France ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Isabelle Szezepanski
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France M3C-Unité Médico-Chirugicale de Cardiologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Magalie Ladouceur
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France M3C-Unité Médico-Chirugicale de Cardiologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Sophie Nadaud
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR_S 1166-ICAN, Paris, France ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Damien Bonnet
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France M3C-Unité Médico-Chirugicale de Cardiologie Pédiatrique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Florent Soubrier
- Genetics Dept, GH Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR_S 1166-ICAN, Paris, France ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| |
Collapse
|
40
|
Abstract
The prevalence of PH is increasing in the pediatric population, because of improved recognition and increased survival of patients, and remains a significant cause of morbidity and mortality. Recent studies have improved the understanding of pediatric PH, but management remains challenging because of a lack of evidence-based clinical trials. The growing contribution of developmental lung disease requires dedicated research to explore the use of existing therapies as well as the creation of novel therapies. Adequate study of pediatric PH will require multicenter collaboration due to the small numbers of patients, multifactorial disease causes, and practice variability.
Collapse
Affiliation(s)
- Dunbar Ivy
- Section of Pediatric Cardiology, Children's Hospital Colorado, University of Colorado School of Medicine, 13123 East 16th Avenue, B100, Aurora, CO 80045, USA.
| |
Collapse
|
41
|
van der Bruggen CE, Happé CM, Dorfmüller P, Trip P, Spruijt OA, Rol N, Hoevenaars FP, Houweling AC, Girerd B, Marcus JT, Mercier O, Humbert M, Handoko ML, van der Velden J, Vonk Noordegraaf A, Bogaard HJ, Goumans MJ, de Man FS. Bone Morphogenetic Protein Receptor Type 2 Mutation in Pulmonary Arterial Hypertension: A View on the Right Ventricle. Circulation 2016; 133:1747-60. [PMID: 26984938 DOI: 10.1161/circulationaha.115.020696] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 03/11/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND The effect of a mutation in the bone morphogenetic protein receptor 2 (BMPR2) gene on right ventricular (RV) pressure overload in patients with pulmonary arterial hypertension is unknown. Therefore, we investigated RV function in patients who have pulmonary arterial hypertension with and without the BMPR2 mutation by combining in vivo measurements with molecular and histological analysis of human RV and left ventricular tissue. METHODS AND RESULTS In total, 95 patients with idiopathic or familial pulmonary arterial hypertension were genetically screened for the presence of a BMPR2 mutation: 28 patients had a BMPR2 mutation, and 67 patients did not have a BMPR2 mutation. In vivo measurements were assessed using right heart catheterization and cardiac MRI. Despite a similar mean pulmonary artery pressure (noncarriers 54±15 versus mutation carriers 55±9 mm Hg) and pulmonary vascular resistance (755 [483-1043] versus 931 [624-1311] dynes·s(-1)·cm(-5)), mutation carriers presented with a more severely compromised RV function (RV ejection fraction: 37.6±12.8% versus 29.0±9%: P<0.05; cardiac index 2.7±0.9 versus 2.2±0.4 L·min(-1)·m(-2)). Differences continued to exist after treatment. To investigate the role of transforming growth factor β and bone morphogenetic protein receptor II signaling, human RV and left ventricular tissue were studied in controls (n=6), mutation carriers (n=5), and noncarriers (n=11). However, transforming growth factor β and bone morphogenetic protein receptor II signaling, and hypertrophy, apoptosis, fibrosis, capillary density, inflammation, and cardiac metabolism, as well, were similar between mutation carriers and noncarriers. CONCLUSIONS Despite a similar afterload, RV function is more severely affected in mutation carriers than in noncarriers. However, these differences cannot be explained by a differential transforming growth factor β, bone morphogenetic protein receptor II signaling, or cardiac adaptation.
Collapse
Affiliation(s)
- Cathelijne E van der Bruggen
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Chris M Happé
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Peter Dorfmüller
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Pia Trip
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Onno A Spruijt
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Nina Rol
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Femke P Hoevenaars
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Arjan C Houweling
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Barbara Girerd
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Johannes T Marcus
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Olaf Mercier
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Marc Humbert
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - M Louis Handoko
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Jolanda van der Velden
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Anton Vonk Noordegraaf
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Harm Jan Bogaard
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Marie-José Goumans
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.)
| | - Frances S de Man
- From Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.E.E.V.D.B., C.M.H., P.T., O.A.S., N.R., A.V.N., H.J.B., F.S.d.M.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.M.H., N.R., F.P.H., J.V.D.V., F.S.d.M.); Univ. Paris-Sud, Le Kremlin-Bicêtre, France (P.D., B.G., .M.H.); AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (P.D., B.G., M.H.); INSERM999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France (P.D., B.G., M.H.); Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands (A.C.H.); Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands (J.T.M.); Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Hôpital Marie-Lannelongue, Le Plessis Robinson, Paris-Sud University, France (O.M.); Department of Cardiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (M.L.H.); and Department of Molecular Cell Biology, Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, The Netherlands (M.-J.G.).
| |
Collapse
|
42
|
Wang J, Zhang C, Zhang Z, Zheng Z, Sun D, Yang Q, Hadadi C, Li D, Xu X, Xiong M, Zhou Q, Guo M, Wang Y, Tang C, Xu G, Yang K, Zhong N, Lu W. A Functional Variant rs6435156C > T in BMPR2 is Associated With Increased Risk of Chronic Obstructive Pulmonary Disease (COPD) in Southern Chinese Population. EBioMedicine 2016; 5:167-74. [PMID: 27077124 PMCID: PMC4816816 DOI: 10.1016/j.ebiom.2016.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUNDS Bone morphogenetic protein receptor type 2 (BMPR2) signaling is anti-inflammatory. Decreased BMPR2 expression was seen in lung tissue from chronic obstructive pulmonary disease (COPD) patients. METHODS The selected single nucleotide polymorphisms (SNPs) in BMPR2 were genotyped with polymerase chain reaction (PCR) ligase detection reaction. The effects of SNPs on gene expression were analyzed with luciferase assays. The mRNA and protein expression levels of BMPR2 in peripheral blood mononuclear cells (PBMCs) from COPD patients were determined by quantitative PCR and western blotting, respectively. FINDINGS Two SNPs, rs6435156C > T and rs1048829G > T in the 3'-untranslated region (3'UTR) of BMPR2 were selected and genotyped in COPD case and healthy control subjects from southern Chinese population. Both of them were found associated with significantly increased COPD risk (adjusted odds ratio [OR] = 1.58 with 95% confidence interval [CI] = 1.14-2.15, P = 0.0056 for rs6435156C > T; adjusted OR = 1.47 and 95% CI = 1.10-1.97, P = 0.0092 for rs1048829G > T). Older age, cigarette smoking, family history of cancer and COPD were all factors that interacted with rs6435156C > T and rs1048829G > T causing increased COPD risk. Cigarette smokers with rs6435156 (CT + TT) or rs1048829 (GT + TT) were more susceptible to COPD than that with the rs6435156CC or rs1048829GG genotypes. In A549 human alveolar epithelial cells, luciferase reporter assays revealed that introduction of 3'UTR of BMPR2 plasmids carrying rs6435156T allele but not rs1048829T led to lower luciferase activity than the wild-type C or G alleles. Comparing to rs6435156CC, treatment with hsa-miR-20a mimics deceased whereas hsa-miR-20a inhibitor restored the luciferase reporter activity in cells transfected with constructs carrying rs6435156TT. BMPR2 mRNA and protein expressions were significantly lower in PBMCs from COPD smokers than that in non-smokers. COPD patients carrying rs6435156T allele had less BMPR2 expression in PBMCs. INTERPRETATION This study demonstrated that both rs6435156C > T and rs1048829G > T variants in BMPR2 contributed to increased susceptibility to COPD. The T variants of rs6435156 increased COPD risk likely by binding with hsa-miR-20a, thus leading to downregulated BMPR2 expression in lung epithelial and immune cells.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Department of Respiration, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010017, Inner Mongolia, China; Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Chenting Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zili Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zeguang Zheng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dejun Sun
- Department of Respiration, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010017, Inner Mongolia, China
| | - Quan Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cyrus Hadadi
- Geisinger Medical Center, 100 North Academy Avenue, Danville, PA 17822, USA
| | - Defu Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoming Xu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingmei Xiong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qipeng Zhou
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meihua Guo
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingfeng Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chun Tang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guihua Xu
- Department of Respiration, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010017, Inner Mongolia, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
43
|
Evans JDW, Girerd B, Montani D, Wang XJ, Galiè N, Austin ED, Elliott G, Asano K, Grünig E, Yan Y, Jing ZC, Manes A, Palazzini M, Wheeler LA, Nakayama I, Satoh T, Eichstaedt C, Hinderhofer K, Wolf M, Rosenzweig EB, Chung WK, Soubrier F, Simonneau G, Sitbon O, Gräf S, Kaptoge S, Di Angelantonio E, Humbert M, Morrell NW. BMPR2 mutations and survival in pulmonary arterial hypertension: an individual participant data meta-analysis. THE LANCET. RESPIRATORY MEDICINE 2016; 4:129-37. [PMID: 26795434 PMCID: PMC4737700 DOI: 10.1016/s2213-2600(15)00544-5] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/15/2015] [Accepted: 12/17/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Mutations in the gene encoding the bone morphogenetic protein receptor type II (BMPR2) are the commonest genetic cause of pulmonary arterial hypertension (PAH). However, the effect of BMPR2 mutations on clinical phenotype and outcomes remains uncertain. METHODS We analysed individual participant data of 1550 patients with idiopathic, heritable, and anorexigen-associated PAH from eight cohorts that had been systematically tested for BMPR2 mutations. The primary outcome was the composite of death or lung transplantation. All-cause mortality was the secondary outcome. Hazard ratios (HRs) for death or transplantation and all-cause mortality associated with the presence of BMPR2 mutation were calculated using Cox proportional hazards models stratified by cohort. FINDINGS Overall, 448 (29%) of 1550 patients had a BMPR2 mutation. Mutation carriers were younger at diagnosis (mean age 35·4 [SD 14·8] vs 42·0 [17·8] years), had a higher mean pulmonary artery pressure (60·5 [13·8] vs 56·4 [15·3] mm Hg) and pulmonary vascular resistance (16·6 [8·3] vs 12·9 [8·3] Wood units), and lower cardiac index (2·11 [0·69] vs 2·51 [0·92] L/min per m(2); all p<0·0001). Patients with BMPR2 mutations were less likely to respond to acute vasodilator testing (3% [10 of 380] vs 16% [147 of 907]; p<0·0001). Among the 1164 individuals with available survival data, age-adjusted and sex-adjusted HRs comparing BMPR2 mutation carriers with non-carriers were 1·42 (95% CI 1·15-1·75; p=0·0011) for the composite of death or lung transplantation and 1·27 (1·00-1·60; p=0·046) for all-cause mortality. These HRs were attenuated after adjustment for potential mediators including pulmonary vascular resistance, cardiac index, and vasoreactivity. HRs for death or transplantation and all-cause mortality associated with BMPR2 mutation were similar in men and women, but higher in patients with a younger age at diagnosis (p=0·0030 for death or transplantation, p=0·011 for all-cause mortality). INTERPRETATION Patients with PAH and BMPR2 mutations present at a younger age with more severe disease, and are at increased risk of death, and death or transplantation, compared with those without BMPR2 mutations. FUNDING Cambridge NIHR Biomedical Research Centre, Medical Research Council, British Heart Foundation, Assistance Publique-Hôpitaux de Paris, INSERM, Université Paris-Sud, Intermountain Research and Medical Foundation, Vanderbilt University, National Center for Advancing Translational Sciences, National Institutes of Health, National Natural Science Foundation of China, and Beijing Natural Science Foundation.
Collapse
Affiliation(s)
- Jonathan D W Evans
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK; Department of Cardiology, Papworth Hospital, Cambridge, UK
| | - Barbara Girerd
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France; APHP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire Thorax Innovation, Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France; INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France; APHP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire Thorax Innovation, Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France; INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Xiao-Jian Wang
- Thrombosis and Vascular Medicine Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nazzareno Galiè
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Eric D Austin
- Department of Paediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Greg Elliott
- Department of Medicine, Intermountain Medical Center and the University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Koichiro Asano
- Division of Pulmonary Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Ekkehard Grünig
- Centre for Pulmonary Hypertension, Thorax Clinic, University Hospital Heidelberg, Heidelberg, Germany
| | - Yi Yan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhi-Cheng Jing
- Thrombosis and Vascular Medicine Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | | | - Massimiliano Palazzini
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Lisa A Wheeler
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ikue Nakayama
- Department of Medicine, Intermountain Medical Center and the University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Toru Satoh
- Division of Cardiology, Department of Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Christina Eichstaedt
- Centre for Pulmonary Hypertension, Thorax Clinic, University Hospital Heidelberg, Heidelberg, Germany; Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Katrin Hinderhofer
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Wolf
- Centre for Pulmonary Hypertension, Thorax Clinic, University Hospital Heidelberg, Heidelberg, Germany; Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Erika B Rosenzweig
- Department of Pediatric Cardiology, Columbia University Medical Center, New York, NY USA
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, NY USA
| | - Florent Soubrier
- Université Pierre et Marie Curie-Paris 6, Laboratoire d'Oncogénétique et Angiogénétique Moléculaire, Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Gérald Simonneau
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France; APHP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire Thorax Innovation, Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France; INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Olivier Sitbon
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France; APHP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire Thorax Innovation, Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France; INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Stefan Gräf
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK; Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Stephen Kaptoge
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France; APHP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire Thorax Innovation, Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France; INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
44
|
Girerd B, Montani D, Jaïs X, Eyries M, Yaici A, Sztrymf B, Savale L, Parent F, Coulet F, Godinas L, Lau EM, Tamura Y, Sitbon O, Soubrier F, Simonneau G, Humbert M. Genetic counselling in a national referral centre for pulmonary hypertension. Eur Respir J 2015; 47:541-52. [PMID: 26699722 DOI: 10.1183/13993003.00717-2015] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 10/14/2015] [Indexed: 01/22/2023]
Abstract
Genetic causes of pulmonary arterial hypertension (PAH) and pulmonary veno-occlusive disease (PVOD) have been identified, leading to a growing need for genetic counselling.Between 2003 and 2014, genetic counselling was offered to 529 PAH and 100 PVOD patients at the French Referral Centre for Pulmonary Hypertension.Mutations in PAH-predisposing genes were identified in 72 patients presenting as sporadic PAH (17% of cases; 62 mutations in BMPR2, nine in ACVRL1 (ALK1) and one in ENG) and in 94 patients with a PAH family history (89% of cases; 89 mutations in BMPR2, three in ACVRL1 (ALK1) and two in KCNK3). Bi-allelic mutations in EIF2AK4 were identified in all patients with a family history of PVOD (n=19) and in seven patients (8.6%) presenting as sporadic PVOD. Pre-symptomatic genetic diagnosis was offered to 272 relatives of heritable PAH patients, identifying mutations in 36.4% of them. A screening programme is now offered to asymptomatic mutation carriers to detect PAH in an early phase and to identify predictors of outcomes in asymptomatic BMPR2 mutation carriers. BMPR2 screening allowed us to offer pre-implantation diagnosis to two couples with a BMPR2 mutation.Genetic counselling can be implemented in pulmonary hypertension centres.
Collapse
Affiliation(s)
- Barbara Girerd
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France Both authors contributed equally to this work
| | - David Montani
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France Both authors contributed equally to this work
| | - Xavier Jaïs
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Mélanie Eyries
- UMR_S956, Université Pierre et Marie Curie-Paris 6, INSERM, Laboratoire d'Oncogénétique et Angiogénétique Moléculaire, Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Azzedine Yaici
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Benjamin Sztrymf
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Laurent Savale
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Florence Parent
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Florence Coulet
- UMR_S956, Université Pierre et Marie Curie-Paris 6, INSERM, Laboratoire d'Oncogénétique et Angiogénétique Moléculaire, Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Laurent Godinas
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Edmund M Lau
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Yuichi Tamura
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Olivier Sitbon
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Florent Soubrier
- UMR_S956, Université Pierre et Marie Curie-Paris 6, INSERM, Laboratoire d'Oncogénétique et Angiogénétique Moléculaire, Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Gérald Simonneau
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- Université Paris Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital de Bicêtre, Le Kremlin Bicêtre, France UMR_S999, Université Paris-Sud, INSERM, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
45
|
Abman SH, Hansmann G, Archer SL, Ivy DD, Adatia I, Chung WK, Hanna BD, Rosenzweig EB, Raj JU, Cornfield D, Stenmark KR, Steinhorn R, Thébaud B, Fineman JR, Kuehne T, Feinstein JA, Friedberg MK, Earing M, Barst RJ, Keller RL, Kinsella JP, Mullen M, Deterding R, Kulik T, Mallory G, Humpl T, Wessel DL. Pediatric Pulmonary Hypertension: Guidelines From the American Heart Association and American Thoracic Society. Circulation 2015; 132:2037-99. [PMID: 26534956 DOI: 10.1161/cir.0000000000000329] [Citation(s) in RCA: 717] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension is associated with diverse cardiac, pulmonary, and systemic diseases in neonates, infants, and older children and contributes to significant morbidity and mortality. However, current approaches to caring for pediatric patients with pulmonary hypertension have been limited by the lack of consensus guidelines from experts in the field. In a joint effort from the American Heart Association and American Thoracic Society, a panel of experienced clinicians and clinician-scientists was assembled to review the current literature and to make recommendations on the diagnosis, evaluation, and treatment of pediatric pulmonary hypertension. This publication presents the results of extensive literature reviews, discussions, and formal scoring of recommendations for the care of children with pulmonary hypertension.
Collapse
MESH Headings
- Cardiovascular Agents/therapeutic use
- Child
- Child, Preschool
- Combined Modality Therapy
- Diagnostic Imaging/methods
- Disease Management
- Extracorporeal Membrane Oxygenation
- Genetic Counseling
- Heart Defects, Congenital/complications
- Heart Defects, Congenital/therapy
- Hernias, Diaphragmatic, Congenital/complications
- Hernias, Diaphragmatic, Congenital/therapy
- Humans
- Hypertension, Pulmonary/diagnosis
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/therapy
- Infant
- Infant, Newborn
- Lung/embryology
- Lung Transplantation
- Nitric Oxide/administration & dosage
- Nitric Oxide/therapeutic use
- Oxygen Inhalation Therapy
- Persistent Fetal Circulation Syndrome/diagnosis
- Persistent Fetal Circulation Syndrome/therapy
- Postoperative Complications/therapy
- Respiration, Artificial/adverse effects
- Respiration, Artificial/methods
- Ventilator-Induced Lung Injury/prevention & control
Collapse
|
46
|
Machado RD, Southgate L, Eichstaedt CA, Aldred MA, Austin ED, Best DH, Chung WK, Benjamin N, Elliott CG, Eyries M, Fischer C, Gräf S, Hinderhofer K, Humbert M, Keiles SB, Loyd JE, Morrell NW, Newman JH, Soubrier F, Trembath RC, Viales RR, Grünig E. Pulmonary Arterial Hypertension: A Current Perspective on Established and Emerging Molecular Genetic Defects. Hum Mutat 2015; 36:1113-27. [PMID: 26387786 DOI: 10.1002/humu.22904] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/04/2015] [Indexed: 12/20/2022]
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.
Collapse
Affiliation(s)
- Rajiv D Machado
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Laura Southgate
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Division of Genetics & Molecular Medicine, King's College London, London, United Kingdom
| | - Christina A Eichstaedt
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany.,Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | | | - Eric D Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - D Hunter Best
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York
| | - Nicola Benjamin
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany
| | - C Gregory Elliott
- Departments of Medicine, Intermountain Medical Center and the University of Utah School of Medicine, Salt Lake City, Utah
| | - Mélanie Eyries
- Unité Mixte de Recherche en Santé (UMR_S 1166), Université Pierre and Marie Curie Université Paris 06 (UPMC) and Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Genetics Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Institute for Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Christine Fischer
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | | | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Paris, France.,Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital Bicêtre, AP-HP, Paris, France.,INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Paris, France
| | - Steven B Keiles
- Quest Diagnostics, Action from Insight, San Juan Capistrano, California
| | - James E Loyd
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Addenbrooke's & Papworth Hospitals, Cambridge, United Kingdom
| | - John H Newman
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Florent Soubrier
- Unité Mixte de Recherche en Santé (UMR_S 1166), Université Pierre and Marie Curie Université Paris 06 (UPMC) and Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Genetics Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Institute for Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Richard C Trembath
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rebecca Rodríguez Viales
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany.,Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ekkehard Grünig
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
47
|
Girerd B, Coulet F, Jaïs X, Eyries M, Van Der Bruggen C, De Man F, Houweling A, Dorfmüller P, Savale L, Sitbon O, Vonk-Noordegraaf A, Soubrier F, Simonneau G, Humbert M, Montani D. Characteristics of pulmonary arterial hypertension in affected carriers of a mutation located in the cytoplasmic tail of bone morphogenetic protein receptor type 2. Chest 2015; 147:1385-1394. [PMID: 25429696 DOI: 10.1378/chest.14-0880] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Mutations in BMPR2 encoding bone morphogenetic protein receptor type 2 (BMPRII) is the main genetic risk factor for heritable pulmonary arterial hypertension (PAH). The suspected mechanism is considered to be a defect of BMP signaling. The BMPRII receptor exists in a short isoform without a cytoplasmic tail, which has preserved BMP signaling. METHODS This cohort study compared age at PAH diagnosis and severity between patients carrying a BMPR2 mutation affecting the cytoplasmic tail of BMPRII and affected carriers of a mutation upstream of this domain. RESULTS We identified 171 carriers affected with PAH with a mutated BMPR2. Twenty-three were carriers of a point mutation located on the cytoplasmic tail of BMPRII. This population was characterized by having an older age at diagnosis compared with other BMPR2 mutation carriers (43.2 ± 12.1 years and 35.7 ± 14.6 years, P = .040), a lower pulmonary vascular resistance (13.3 ± 3.5 and 17.4 ± 6.7, P = .023), and a higher proportion of acute vasodilator responders with a long-term response to calcium channel blockers (8.7% and 0%, P = .02). No statistically significant differences were observed in survival. An in vitro assay showed that mutations located in the cytoplasmic tail led to normal activation of the Smad pathway, whereas activation was abolished in the presence of mutations located in the kinase domain. CONCLUSIONS Patients carrying a mutation affecting the cytoplasmic tail of BMPRII were characterized by an older age at diagnosis compared with other BMPR2 mutation carriers, less severe hemodynamic characteristics, and a greater chance of being a long-term responder to calcium channel blockers. Further investigations are needed to better understand the consequences of these BMPR2 mutations in BMPRII signaling pathways and their possible role in pulmonary arterial remodeling.
Collapse
Affiliation(s)
- Barbara Girerd
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Florence Coulet
- Genetics Department, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Xavier Jaïs
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Mélanie Eyries
- Genetics Department, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France; ICAN Institute for Cardiometabolism and Nutrition, Paris, France; Unité Mixte de Recherche en Santé (UMR_S 1166), UPMC - Université Paris-Sorbonne, and INSERM, Paris, France
| | - Cathelijne Van Der Bruggen
- Departments of Pulmonary Medicine, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Frances De Man
- Departments of Pulmonary Medicine, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjan Houweling
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter Dorfmüller
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Laurent Savale
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Olivier Sitbon
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Anton Vonk-Noordegraaf
- Departments of Pulmonary Medicine, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Florent Soubrier
- Genetics Department, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France; ICAN Institute for Cardiometabolism and Nutrition, Paris, France; Unité Mixte de Recherche en Santé (UMR_S 1166), UPMC - Université Paris-Sorbonne, and INSERM, Paris, France
| | - Gérald Simonneau
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France.
| |
Collapse
|
48
|
Viales RR, Eichstaedt CA, Ehlken N, Fischer C, Lichtblau M, Grünig E, Hinderhofer K. Mutation in BMPR2 Promoter: A 'Second Hit' for Manifestation of Pulmonary Arterial Hypertension? PLoS One 2015; 10:e0133042. [PMID: 26167679 PMCID: PMC4500409 DOI: 10.1371/journal.pone.0133042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/22/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Hereditary pulmonary arterial hypertension (HPAH) can be caused by autosomal dominant inherited mutations of TGF-β genes, such as the bone morphogenetic protein receptor 2 (BMPR2) and Endoglin (ENG) gene. Additional modifier genes may play a role in disease manifestation and severity. In this study we prospectively assessed two families with known BMPR2 or ENG mutations clinically and genetically and screened for a second mutation in the BMPR2 promoter region. METHODS We investigated the BMPR2 promoter region by direct sequencing in two index-patients with invasively confirmed diagnosis of HPAH, carrying a mutation in the BMPR2 and ENG gene, respectively. Sixteen family members have been assessed clinically by non-invasive methods and genetically by direct sequencing. RESULTS In both index patients with a primary BMPR2 deletion (exon 2 and 3) and Endoglin missense variant (c.1633G>A, p.(G545S)), respectively, we detected a second mutation (c.-669G>A) in the promoter region of the BMPR2 gene. The index patients with 2 mutations/variants were clinically severely affected at early age, whereas further family members with only one mutation had no manifest HPAH. CONCLUSION The finding of this study supports the hypothesis that additional mutations may lead to an early and severe manifestation of HPAH. This study shows for the first time that in the regulatory region of the BMPR2 gene the promoter may be important for disease penetrance. Further studies are needed to assess the incidence and clinical relevance of mutations of the BMPR2 promoter region in a larger patient cohort.
Collapse
Affiliation(s)
- Rebecca Rodríguez Viales
- University Hospital Heidelberg, Centre for pulmonary hypertension of the Thoraxclinic Heidelberg, Heidelberg, Germany; Heidelberg University, Institute of Human Genetics, Heidelberg, Germany
| | - Christina A Eichstaedt
- University Hospital Heidelberg, Centre for pulmonary hypertension of the Thoraxclinic Heidelberg, Heidelberg, Germany
| | - Nicola Ehlken
- University Hospital Heidelberg, Centre for pulmonary hypertension of the Thoraxclinic Heidelberg, Heidelberg, Germany
| | - Christine Fischer
- Heidelberg University, Institute of Human Genetics, Heidelberg, Germany
| | - Mona Lichtblau
- University Hospital Heidelberg, Centre for pulmonary hypertension of the Thoraxclinic Heidelberg, Heidelberg, Germany
| | - Ekkehard Grünig
- University Hospital Heidelberg, Centre for pulmonary hypertension of the Thoraxclinic Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
49
|
Abstract
A biomarker is a characteristic that can be used as an indicator of a biological state. A biomarker can be a clinical observation, laboratory test or an imaging parameter. In this review, we discuss the use of biomarkers in differentiating cardiac from noncardiac disease; predicting the prognosis of patients with heart failure, pulmonary hypertension and dilated cardiomyopathy; diagnosing subclinical cardiac involvement in muscular dystrophy and postchemotherapy cancer patients; detecting acute rejection following heart transplantation; diagnosing Kawasaki disease; aiding the management of postoperative cardiac patients; and managing both common (tetralogy of Fallot) and complex (single-ventricle physiology) congenital heart diseases.
Collapse
Affiliation(s)
- Hythem Nawaytou
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | | |
Collapse
|
50
|
Wang H, Ji R, Meng J, Cui Q, Zou W, Li L, Wang G, Sun L, Li Z, Huo L, Fan Y, Penny DJ. Functional changes in pulmonary arterial endothelial cells associated with BMPR2 mutations. PLoS One 2014; 9:e106703. [PMID: 25187962 PMCID: PMC4154762 DOI: 10.1371/journal.pone.0106703] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/01/2014] [Indexed: 12/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease characterized by abnormal remodeling of small, peripheral pulmonary arteries. Germline mutations in the bone morphogenetic protein receptor type 2 (BMPR2) gene are a major risk factor for developing PAH. At present, the correlation between the BMPR2 mutation and the patient's prognosis remains controversial despite several investigations. In this study, we explored the functional effects of four BMPR2 mutations to dissect the functional significance of the BMPR2 gene defect. Cellular immunofluorescence assay of four mutants (Tyr67Cys, Thr268fs, Ser863Asn, and Gln433X) revealed that the BMPR2 protein containing Thr268fs, Ser863Asn, or Gln433X exhibited abnormal subcellular localization. The BrdU incorporation and TUNEL assay suggested that any of the BMPR2 mutations Thr268fs, Ser863Asn, or Gln433X could improve endothelial cell apoptosis and decrease cell proliferation. All of the four mutants could inhibit nitric oxide (NO) synthesis in HLMVE cells, and ET-1 levels increased in the cells transfected with mutant Ser863Asn. Our results will improve the understanding of the genotype-phenotype correlations and mechanisms associated with BMPR2 mutations.
Collapse
Affiliation(s)
- Hu Wang
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ruirui Ji
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jie Meng
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Qiqiong Cui
- Cardiovascular Clinical Research Core, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wenxin Zou
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lei Li
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Guoliang Wang
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Li Sun
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Zhaohui Li
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lei Huo
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yuxin Fan
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daniel J. Penny
- Section of Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|