1
|
Guarino VA, Wertheim BM, Xiao W, Loscalzo J, Zhang Y. Nanoparticle delivery of VEGF and SDF-1α as an approach for treatment of pulmonary arterial hypertension. Pulm Circ 2024; 14:e12412. [PMID: 39380978 PMCID: PMC11459680 DOI: 10.1002/pul2.12412] [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: 02/14/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 10/10/2024] Open
Abstract
Endothelial dysfunction is an underlying mechanism for the development of pulmonary arterial hypertension (PAH). Vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1α (SDF) may help repair the dysfunctional endothelium and provide treatment for PAH. To examine this possibility, nanoparticles carrying human recombinant VEGF and SDF (VEGFNP and SDFNP) were aerosolized into the lungs of nude rats at Day 14 after monocrotaline (MCT) injection and analyses were performed at Day 28. The data show that the VEGFNP/SDFNP delivery led to a lower pulmonary arterial pressure and prevented right ventricular hypertrophy in the MCT rats: the right ventricular systolic pressure of the control, MCT, and MCT + VEGFNP/SDFNP treatment groups were 29±2, 70±9, and 44±5 (mean±SD) mmHg, respectively; the pulmonary vascular resistance indices of the groups were 0.6±0.3, 3.2±0.7, and 1.7±0.5, respectively; and the Fulton indices [-RV/(LV + Septum)] were 0.22±0.01, 0.44±0.07, and 0.23±0.02, respectively. The VEGFNP/SDFNP delivery delayed the thickening of distal pulmonary vessels: the number of nearly occluded vessels in a whole lung section from the MCT and MCT + VEGFNP/SDFNP groups were 46±12 and 2±3, respectively. Gene expression analysis of the endothelial cell markers, VE-cadherin, KDR, BMPR2, and eNOS, and smooth cell markers, SM-MHC and α-SMA, indicated significant loss of distal pulmonary vessels in the MCT- treated rats. VEGFNP/SDFNP delivery did not recover the loss, but significantly increased eNOS and decreased α-SMA expression in the MCT-treated lungs. Thus, the therapeutic effect of VEGFNP/SDFNP may be mediated by improving/repairing endothelial function in the PAH lungs.
Collapse
Affiliation(s)
| | | | - Wusheng Xiao
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Joseph Loscalzo
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Ying‐Yi Zhang
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
2
|
Lawrence A, Myall KJ, Mukherjee B, Marino P. Converging Pathways: A Review of Pulmonary Hypertension in Interstitial Lung Disease. Life (Basel) 2024; 14:1203. [PMID: 39337985 PMCID: PMC11433497 DOI: 10.3390/life14091203] [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: 08/10/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Pulmonary hypertension (PH) in interstitial lung disease (ILD) is relatively common, affecting up to 50% of patients with idiopathic pulmonary fibrosis (IPF). It occurs more frequently in advanced fibrotic ILD, although it may also complicate milder disease and carries significant clinical implications in terms of morbidity and mortality. Key pathological processes driving ILD-PH include hypoxic pulmonary vasoconstriction and pulmonary vascular remodelling. While current understanding of the complex cell signalling pathways and molecular mechanisms underlying ILD-PH remains incomplete, there is evidence for an interplay between the disease pathogenesis of fibrotic ILD and PH, with interest in the role of the pulmonary endothelium in driving pulmonary fibrogenesis more recently. This review examines key clinical trials in ILD-PH therapeutics, including recent research showing promise for the treatment of both ILD-PH and the underlying pulmonary fibrotic process, further supporting the hypothesis of interrelated pathogenesis. Other important management considerations are discussed, including the value of accurate phenotyping in ILD-PH and the success of the "pulmonary vascular" phenotype. This article highlights the close and interconnected nature of fibrotic ILD and PH disease pathogenesis, a perspective likely to improve our understanding and therapeutic approach to this complex condition in the future.
Collapse
Affiliation(s)
| | - Katherine Jane Myall
- Guy's and St Thomas' NHS Foundation Trust, London SE1 9RT, UK
- King's College Hospital, London SE5 9RS, UK
| | - Bhashkar Mukherjee
- Guy's and St Thomas' NHS Foundation Trust, London SE1 9RT, UK
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London SW3 6NP, UK
| | - Philip Marino
- Guy's and St Thomas' NHS Foundation Trust, London SE1 9RT, UK
| |
Collapse
|
3
|
Gomez-Arroyo J, Houweling AC, Bogaard HJ, Aman J, Kitzmiller JA, Porollo A, Dooijes D, Meijboom LJ, Hale P, Pauciulo MW, Hong J, Zhu N, Welch C, Shen Y, Zacharias WJ, McCormack FX, Aldred MA, Weirauch MT, Graf S, Rhodes C, Chung WK, Whitsett JA, Martin LJ, Kalinichenko VV, Nichols WC. Role of Forkhead box F1 in the Pathobiology of Pulmonary Arterial Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.611448. [PMID: 39345371 PMCID: PMC11429893 DOI: 10.1101/2024.09.18.611448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Rationale Approximately 80% of patients with non-familial pulmonary arterial hypertension (PAH) lack identifiable pathogenic genetic variants. While most genetic studies of PAH have focused on predicted loss-of-function variants, recent approaches have identified ultra-rare missense variants associated with the disease. FOXF1 encodes a highly conserved transcription factor, essential for angiogenesis and vasculogenesis in human and mouse lungs. Objectives We identified a rare FOXF1 missense coding variant in two unrelated probands with PAH. FOXF1 is an evolutionarily conserved transcription factor required for lung vascular development and vascular integrity. Our aims were to determine the frequency of FOXF1 variants in larger PAH cohorts compared to the general population, study FOXF1 expression in explanted lung tissue from PAH patients versus control (failed-donor) lungs, and define potential downstream targets linked to PAH development. Methods Three independent, international, multicenter cohorts were analyzed to evaluate the frequency of FOXF1 rare variants. Various composite prediction models assessed the deleteriousness of individual variants. Bulk RNA sequencing datasets from human explanted lung tissues were compared to failed-donor controls to determine FOXF1 expression. Bioinformatic tools identified putative FOXF1 binding targets, which were orthogonally validated using mouse ChIP-seq datasets. Measurements and Main Results Seven novel or ultra-rare missense coding variants were identified across three patient cohorts in different regions of the FOXF1 gene, including the DNA binding domain. FOXF1 expression was dysregulated in PAH lungs, correlating with disease severity. Histological analysis showed heterogeneous FOXF1 expression, with the lowest levels in phenotypically abnormal endothelial cells within complex vascular lesions in PAH samples. A hybrid bioinformatic approach identified FOXF1 downstream targets potentially involved in PAH pathogenesis, including BMPR2 . Conclusions Large genomic and transcriptomic datasets suggest that decreased FOXF1 expression or predicted dysfunction is associated with PAH.
Collapse
|
4
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
5
|
Li W, Quigley K. Bone morphogenetic protein signalling in pulmonary arterial hypertension: revisiting the BMPRII connection. Biochem Soc Trans 2024; 52:1515-1528. [PMID: 38716930 PMCID: PMC11346422 DOI: 10.1042/bst20231547] [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: 03/04/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and life-threatening vascular disorder, characterised by abnormal remodelling of the pulmonary vessels and elevated pulmonary artery pressure, leading to right ventricular hypertrophy and right-sided heart failure. The importance of bone morphogenetic protein (BMP) signalling in the pathogenesis of PAH is demonstrated by human genetic studies. Many PAH risk genes are involved in the BMP signalling pathway and are highly expressed or preferentially act on vascular endothelial cells. Endothelial dysfunction is recognised as an initial trigger for PAH, and endothelial BMP signalling plays a crucial role in the maintenance of endothelial integrity. BMPR2 is the most prevalent PAH gene, found in over 80% of heritable cases. As BMPRII protein is the major type II receptor for a large family of BMP ligands and expressed ubiquitously in many tissues, dysregulated BMP signalling in other cells may also contribute to PAH pathobiology. Sotatercept, which contains the extracellular domain of another transforming growth factor-β family type II receptor ActRIIA fused to immunoglobin Fc domain, was recently approved by the FDA as a treatment for PAH. Neither its target cells nor its mechanism of action is fully understood. This review will revisit BMPRII function and its extracellular regulation, summarise how dysregulated BMP signalling in endothelial cells and smooth muscle cells may contribute to PAH pathogenesis, and discuss how novel therapeutics targeting the extracellular regulation of BMP signalling, such as BMP9 and Sotatercept, can be related to restoring BMPRII function.
Collapse
Affiliation(s)
- Wei Li
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| | - Kate Quigley
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| |
Collapse
|
6
|
Agarwal S, Fineman J, Cornfield DN, Alvira CM, Zamanian RT, Goss K, Yuan K, Bonnet S, Boucherat O, Pullamsetti S, Alcázar MA, Goncharova E, Kudryashova TV, Nicolls MR, de Jesús Pérez V. Seeing pulmonary hypertension through a paediatric lens: a viewpoint. Eur Respir J 2024; 63:2301518. [PMID: 38575157 PMCID: PMC11187317 DOI: 10.1183/13993003.01518-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024]
Abstract
Pulmonary hypertension (PH) is a life-threating condition associated with abnormally elevated pulmonary pressures and right heart failure. Current epidemiological data indicate that PH aetiologies are different between the adult and paediatric population. The most common forms of PH in adults are PH from left heart disease or chronic lung disease, followed by pulmonary arterial hypertension (PAH) [1]; in paediatric patients, PH is most often associated with developmental lung disorders and congenital heart disease (CHD) [2, 3]. In contrast to adults with PH, wherein patients worsen over time despite therapy, PH in children can improve with growth. For example, in infants with bronchopulmonary dysplasia (BPD) and PH morbidity and mortality are high, but with lung growth and ensuring no ongoing lung injury pulmonary vascular disease can improve as evidenced by discontinuation of vasodilator therapy in almost two-thirds of BPD-PH survivors by age 5 years [3, 4]. Paediatric pulmonary hypertension (PH) offers unique genetic and developmental insights that can help in the discovery of novel mechanisms and targets to treat adult PH https://bit.ly/3TMm6bi
Collapse
Affiliation(s)
- Stuti Agarwal
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Jeffrey Fineman
- Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - David N Cornfield
- Division of Pediatric Pulmonary, Asthma, and Sleep Medicine, Stanford University, Palo Alto, CA, USA
| | - Cristina M Alvira
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - Roham T Zamanian
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Kara Goss
- Department of Medicine and Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Ke Yuan
- Boston Children's Hospital, Boston, MA, USA
| | - Sebastien Bonnet
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Olivier Boucherat
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Soni Pullamsetti
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | | - Tatiana V Kudryashova
- University of Pittsburgh Heart, Blood, and Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | |
Collapse
|
7
|
Isobe S, Nair RV, Kang HY, Wang L, Moonen JR, Shinohara T, Cao A, Taylor S, Otsuki S, Marciano DP, Harper RL, Adil MS, Zhang C, Lago-Docampo M, Körbelin J, Engreitz JM, Snyder MP, Rabinovitch M. Reduced FOXF1 links unrepaired DNA damage to pulmonary arterial hypertension. Nat Commun 2023; 14:7578. [PMID: 37989727 PMCID: PMC10663616 DOI: 10.1038/s41467-023-43039-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 10/30/2023] [Indexed: 11/23/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease in which pulmonary arterial (PA) endothelial cell (EC) dysfunction is associated with unrepaired DNA damage. BMPR2 is the most common genetic cause of PAH. We report that human PAEC with reduced BMPR2 have persistent DNA damage in room air after hypoxia (reoxygenation), as do mice with EC-specific deletion of Bmpr2 (EC-Bmpr2-/-) and persistent pulmonary hypertension. Similar findings are observed in PAEC with loss of the DNA damage sensor ATM, and in mice with Atm deleted in EC (EC-Atm-/-). Gene expression analysis of EC-Atm-/- and EC-Bmpr2-/- lung EC reveals reduced Foxf1, a transcription factor with selectivity for lung EC. Reducing FOXF1 in control PAEC induces DNA damage and impaired angiogenesis whereas transfection of FOXF1 in PAH PAEC repairs DNA damage and restores angiogenesis. Lung EC targeted delivery of Foxf1 to reoxygenated EC-Bmpr2-/- mice repairs DNA damage, induces angiogenesis and reverses pulmonary hypertension.
Collapse
Affiliation(s)
- Sarasa Isobe
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramesh V Nair
- Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen Y Kang
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Lingli Wang
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jan-Renier Moonen
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tsutomu Shinohara
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aiqin Cao
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shalina Taylor
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shoichiro Otsuki
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - David P Marciano
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rebecca L Harper
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mir S Adil
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chongyang Zhang
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mauro Lago-Docampo
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jesse M Engreitz
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P Snyder
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Marlene Rabinovitch
- Basic Science and Engineering (BASE) Initiative at the Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA.
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pediatrics - Cardiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
8
|
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
|
9
|
Welch CL, Aldred MA, Balachandar S, Dooijes D, Eichstaedt CA, Gräf S, Houweling AC, Machado RD, Pandya D, Prapa M, Shaukat M, Southgate L, Tenorio-Castano J, Chung WK. Defining the clinical validity of genes reported to cause pulmonary arterial hypertension. Genet Med 2023; 25:100925. [PMID: 37422716 PMCID: PMC10766870 DOI: 10.1016/j.gim.2023.100925] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
PURPOSE Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. Genetic testing is currently recommended for adults diagnosed with heritable, idiopathic, anorexigen-, hereditary hemorrhagic telangiectasia-, and congenital heart disease-associated PAH, PAH with overt features of venous/capillary involvement, and all children diagnosed with PAH. Variants in at least 27 genes have putative evidence for PAH causality. Rigorous assessment of the evidence is needed to inform genetic testing. METHODS An international panel of experts in PAH applied a semi-quantitative scoring system developed by the NIH Clinical Genome Resource to classify the relative strength of evidence supporting PAH gene-disease relationships based on genetic and experimental evidence. RESULTS Twelve genes (BMPR2, ACVRL1, ATP13A3, CAV1, EIF2AK4, ENG, GDF2, KCNK3, KDR, SMAD9, SOX17, and TBX4) were classified as having definitive evidence and 3 genes (ABCC8, GGCX, and TET2) with moderate evidence. Six genes (AQP1, BMP10, FBLN2, KLF2, KLK1, and PDGFD) were classified as having limited evidence for causal effects of variants. TOPBP1 was classified as having no known PAH relationship. Five genes (BMPR1A, BMPR1B, NOTCH3, SMAD1, and SMAD4) were disputed because of a paucity of genetic evidence over time. CONCLUSION We recommend that genetic testing includes all genes with definitive evidence and that caution be taken in the interpretation of variants identified in genes with moderate or limited evidence. Genes with no known evidence for PAH or disputed genes should not be included in genetic testing.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Srimmitha Balachandar
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Dennis Dooijes
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- NIHR BioResource for Translational Research - Rare Diseases, Department of Haemotology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Divya Pandya
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Matina Prapa
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Memoona Shaukat
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Jair Tenorio-Castano
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IDiPAZ, Universidad Autonoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain; ITHACA, European Reference Network, Brussels, Belgium
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY; Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
10
|
Zhong (钟颖) Y, Yu (游博群) PB. Angiogenesis Redux: An Overall Protective Role of VEGF/KDR Signaling in the Microvasculature in Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2023; 43:1784-1787. [PMID: 37675636 PMCID: PMC10803133 DOI: 10.1161/atvbaha.123.319839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Affiliation(s)
- Ying Zhong (钟颖)
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul B. Yu (游博群)
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Saint-Martin Willer A, Santos-Gomes J, Adão R, Brás-Silva C, Eyries M, Pérez-Vizcaino F, Capuano V, Montani D, Antigny F. Physiological and pathophysiological roles of the KCNK3 potassium channel in the pulmonary circulation and the heart. J Physiol 2023; 601:3717-3737. [PMID: 37477289 DOI: 10.1113/jp284936] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
Potassium channel subfamily K member 3 (KCNK3), encoded by the KCNK3 gene, is part of the two-pore domain potassium channel family, constitutively active at resting membrane potentials in excitable cells, including smooth muscle and cardiac cells. Several physiological and pharmacological mediators, such as intracellular signalling pathways, extracellular pH, hypoxia and anaesthetics, regulate KCNK3 channel function. Recent studies show that modulation of KCNK3 channel expression and function strongly influences pulmonary vascular cell and cardiomyocyte function. The altered activity of KCNK3 in pathological situations such as atrial fibrillation, pulmonary arterial hypertension and right ventricular dysfunction demonstrates the crucial role of KCNK3 in cardiovascular homeostasis. Furthermore, loss of function variants of KCNK3 have been identified in patients suffering from pulmonary arterial hypertension and atrial fibrillation. This review focuses on current knowledge of the role of the KCNK3 channel in pulmonary circulation and the heart, in healthy and pathological conditions.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Joana Santos-Gomes
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Rui Adão
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Mélanie Eyries
- Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
12
|
Akiyama T, Sadahiro T, Yamada Y, Fujita R, Abe Y, Nakano K, Honda S, Ema M, Kubota Y, Sakai S, Hizawa N, Ieda M. Flk1 Deficiency and Hypoxia Synergistically Promote Endothelial Dysfunction, Vascular Remodeling, and Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2023; 43:1668-1683. [PMID: 37534464 DOI: 10.1161/atvbaha.123.319266] [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: 03/08/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND The mechanisms underlying pulmonary hypertension (PH) remain largely unknown; further, why advanced vascular remodeling preferentially occurs in arterioles is yet to be answered. VEGF (vascular endothelial growth factor) regulates angiogenesis through Flk1 (fetal liver kinase 1) and Flt1 (fms-like tyrosine kinase 1) on endothelial cells (ECs), which may be related to PH pathogenesis. However, spatiotemporal expression patterns of Flk1 and Flt1 in the pulmonary vascular system and the role of endothelial Flk1 in PH development remain poorly understood. METHODS We analyzed multiple reporter mice, including Flk1-GFP (green fluorescent protein) bacterial artificial chromosome transgenic (Tg), Flt1-DsRed bacterial artificial chromosome Tg, and Flk1-GFP/Flt1-DsRed double Tg mice, to determine the spatiotemporal expression of Flk1 and Flt1 in hypoxia-induced PH. We also used Cdh5CreERT2/Flk1f/f/Tomato (Flk1-KO [knockout]) mice to induce EC-specific Flk1 deletion and lineage tracing in chronic hypoxia. RESULTS Flk1 was specifically expressed in the ECs of small pulmonary vessels, including arterioles. Conversely, Flt1 was more broadly expressed in the ECs of large- to small-sized vessels in adult mouse lungs. Intriguingly, Flk1+ ECs were transiently increased in hypoxia with proliferation, whereas Flt1 expression was unchanged. Flk1-KO mice did not exhibit pulmonary vascular remodeling nor PH in normoxia; however, the arteriolar ECs changed to a cuboidal shape with protrusion. In hypoxia, Flk1 deletion exacerbated EC dysfunction and reduced their number via apoptosis. Additionally, Flk1 deletion promoted medial thickening and neointimal formation in arterioles and worsened PH. Mechanistically, lineage tracing revealed that neointimal cells were derived from Flk1-KO ECs. Moreover, RNA sequencing in pulmonary ECs demonstrated that Flk1 deletion and hypoxia synergistically activated multiple pathways, including cell cycle, senescence/apoptosis, and cytokine/growth factor, concomitant with suppression of cell adhesion and angiogenesis, to promote vascular remodeling. CONCLUSIONS Flk1 and Flt1 were differentially expressed in pulmonary ECs. Flk1 deficiency and hypoxia jointly dysregulated arteriolar ECs to promote vascular remodeling. Thus, dysfunction of Flk1+ ECs may contribute to the pathogenesis of advanced vascular remodeling in pulmonary arterioles.
Collapse
Affiliation(s)
- Tatsuya Akiyama
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Department of Respiratory Medicine (T.A., N.H.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Taketaro Sadahiro
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yu Yamada
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Ryo Fujita
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Division of Regenerative Medicine, Transborder Medical Research Center (R.F.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuto Abe
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Koji Nakano
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Seiichiro Honda
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masatsugu Ema
- Division of Regenerative Medicine, Transborder Medical Research Center (R.F.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshiaki Kubota
- Departments of Anatomy (Y.K.), Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Sakai
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Faculty of Health Science, Tsukuba University of Technology, Japan (S.S.)
| | - Nobuyuki Hizawa
- Department of Respiratory Medicine (T.A., N.H.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Cardiology (M.I.), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Chakraborty A, Nathan A, Orcholski M, Agarwal S, Shamskhou EA, Auer N, Mitra A, Guardado ES, Swaminathan G, Condon DF, Yu J, McCarra M, Juul NH, Mallory A, Guzman-Hernandez RA, Yuan K, Rojas V, Crossno JT, Yung LM, Yu PB, Spencer T, Winn RA, Frump A, Karoor V, Lahm T, Hedlin H, Fineman JR, Lafyatis R, Knutsen CNF, Alvira CM, Cornfield DN, de Jesus Perez VA. Wnt7a deficit is associated with dysfunctional angiogenesis in pulmonary arterial hypertension. Eur Respir J 2023; 61:2201625. [PMID: 37024132 PMCID: PMC10259331 DOI: 10.1183/13993003.01625-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/21/2023] [Indexed: 04/08/2023]
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) is characterised by loss of microvessels. The Wnt pathways control pulmonary angiogenesis but their role in PAH is incompletely understood. We hypothesised that Wnt activation in pulmonary microvascular endothelial cells (PMVECs) is required for pulmonary angiogenesis, and its loss contributes to PAH. METHODS Lung tissue and PMVECs from healthy and PAH patients were screened for Wnt production. Global and endothelial-specific Wnt7a -/- mice were generated and exposed to chronic hypoxia and Sugen-hypoxia (SuHx). RESULTS Healthy PMVECs demonstrated >6-fold Wnt7a expression during angiogenesis that was absent in PAH PMVECs and lungs. Wnt7a expression correlated with the formation of tip cells, a migratory endothelial phenotype critical for angiogenesis. PAH PMVECs demonstrated reduced vascular endothelial growth factor (VEGF)-induced tip cell formation as evidenced by reduced filopodia formation and motility, which was partially rescued by recombinant Wnt7a. We discovered that Wnt7a promotes VEGF signalling by facilitating Y1175 tyrosine phosphorylation in vascular endothelial growth factor receptor 2 (VEGFR2) through receptor tyrosine kinase-like orphan receptor 2 (ROR2), a Wnt-specific receptor. We found that ROR2 knockdown mimics Wnt7a insufficiency and prevents recovery of tip cell formation with Wnt7a stimulation. While there was no difference between wild-type and endothelial-specific Wnt7a -/- mice under either chronic hypoxia or SuHx, global Wnt7a +/- mice in hypoxia demonstrated higher pulmonary pressures and severe right ventricular and lung vascular remodelling. Similar to PAH, Wnt7a +/- PMVECs exhibited an insufficient angiogenic response to VEGF-A that improved with Wnt7a. CONCLUSIONS Wnt7a promotes VEGF signalling in lung PMVECs and its loss is associated with an insufficient VEGF-A angiogenic response. We propose that Wnt7a deficiency contributes to progressive small vessel loss in PAH.
Collapse
Affiliation(s)
- Ananya Chakraborty
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
- These authors contributed equally
| | - Abinaya Nathan
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
- These authors contributed equally
| | - Mark Orcholski
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | | - Natasha Auer
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Ankita Mitra
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | | - Gowri Swaminathan
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - David F Condon
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Joyce Yu
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Matthew McCarra
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Nicholas H Juul
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | | | | - Ke Yuan
- Boston Children's Hospital, Boston, MA, USA
| | | | - Joseph T Crossno
- Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Paul B Yu
- Brigham and Women's Hospital, Boston, MA, USA
| | | | - Robert A Winn
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Tim Lahm
- National Jewish Center, Denver, CO, USA
| | - Haley Hedlin
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Jeffrey R Fineman
- Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Robert Lafyatis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carsten N F Knutsen
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - Cristina M Alvira
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - David N Cornfield
- Division of Pediatric Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | | |
Collapse
|
14
|
Piccari L, Allwood B, Antoniou K, Chung JH, Hassoun PM, Nikkho SM, Saggar R, Shlobin OA, Vitulo P, Nathan SD, Wort SJ. Pathogenesis, clinical features, and phenotypes of pulmonary hypertension associated with interstitial lung disease: A consensus statement from the Pulmonary Vascular Research Institute's Innovative Drug Development Initiative - Group 3 Pulmonary Hypertension. Pulm Circ 2023; 13:e12213. [PMID: 37025209 PMCID: PMC10071306 DOI: 10.1002/pul2.12213] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a frequent complication of interstitial lung disease (ILD). Although PH has mostly been described in idiopathic pulmonary fibrosis, it can manifest in association with many other forms of ILD. Associated pathogenetic mechanisms are complex and incompletely understood but there is evidence of disruption of molecular and genetic pathways, with panvascular histopathologic changes, multiple pathophysiologic sequelae, and profound clinical ramifications. While there are some recognized clinical phenotypes such as combined pulmonary fibrosis and emphysema and some possible phenotypes such as connective tissue disease associated with ILD and PH, the identification of further phenotypes of PH in ILD has thus far proven elusive. This statement reviews the current evidence on the pathogenesis, recognized patterns, and useful diagnostic tools to detect phenotypes of PH in ILD. Distinct phenotypes warrant recognition if they are characterized through either a distinct presentation, clinical course, or treatment response. Furthermore, we propose a set of recommendations for future studies that might enable the recognition of new phenotypes.
Collapse
Affiliation(s)
- Lucilla Piccari
- Department of Pulmonary Medicine Hospital del Mar Barcelona Spain
| | - Brian Allwood
- Department of Medicine, Division of Pulmonology Stellenbosch University & Tygerberg Hospital Cape Town South Africa
| | - Katerina Antoniou
- Department of Thoracic Medicine University of Crete School of Medicine Heraklion Crete Greece
| | - Jonathan H Chung
- Department of Radiology The University of Chicago Medicine Chicago Illinois USA
| | - Paul M Hassoun
- Department of Medicine, Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore Maryland USA
| | | | - Rajan Saggar
- Lung & Heart-Lung Transplant and Pulmonary Hypertension Programs University of California Los Angeles David Geffen School of Medicine Los Angeles California USA
| | - Oksana A Shlobin
- Advanced Lung Disease and Transplant Program, Inova Health System Falls Church Virginia USA
| | - Patrizio Vitulo
- Department of Pulmonary Medicine IRCCS Mediterranean Institute for Transplantation and Advanced Specialized Therapies Palermo Sicilia Italy
| | - Steven D Nathan
- Advanced Lung Disease and Transplant Program, Inova Health System Falls Church Virginia USA
| | - Stephen John Wort
- National Pulmonary Hypertension Service at the Royal Brompton Hospital London UK
- National Heart and Lung Institute, Imperial College London UK
| |
Collapse
|
15
|
Myronenko O, Foris V, Crnkovic S, Olschewski A, Rocha S, Nicolls MR, Olschewski H. Endotyping COPD: hypoxia-inducible factor-2 as a molecular "switch" between the vascular and airway phenotypes? Eur Respir Rev 2023; 32:220173. [PMID: 36631133 PMCID: PMC9879331 DOI: 10.1183/16000617.0173-2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/08/2022] [Indexed: 01/13/2023] Open
Abstract
COPD is a heterogeneous disease with multiple clinical phenotypes. COPD endotypes can be determined by different expressions of hypoxia-inducible factors (HIFs), which, in combination with individual susceptibility and environmental factors, may cause predominant airway or vascular changes in the lung. The pulmonary vascular phenotype is relatively rare among COPD patients and characterised by out-of-proportion pulmonary hypertension (PH) and low diffusing capacity of the lung for carbon monoxide, but only mild-to-moderate airway obstruction. Its histologic feature, severe remodelling of the small pulmonary arteries, can be mediated by HIF-2 overexpression in experimental PH models. HIF-2 is not only involved in the vascular remodelling but also in the parenchyma destruction. Endothelial cells from human emphysema lungs express reduced HIF-2α levels, and the deletion of pulmonary endothelial Hif-2α leads to emphysema in mice. This means that both upregulation and downregulation of HIF-2 have adverse effects and that HIF-2 may represent a molecular "switch" between the development of the vascular and airway phenotypes in COPD. The mechanisms of HIF-2 dysregulation in the lung are only partly understood. HIF-2 levels may be controlled by NAD(P)H oxidases via iron- and redox-dependent mechanisms. A better understanding of these mechanisms may lead to the development of new therapeutic targets.
Collapse
Affiliation(s)
- Oleh Myronenko
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Sonia Rocha
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Horst Olschewski
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| |
Collapse
|
16
|
Smoking history and pulmonary arterial hypertension: Demographics, onset, and outcomes. J Heart Lung Transplant 2023; 42:377-389. [PMID: 36404264 DOI: 10.1016/j.healun.2022.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Smoking prevalence and its association with pulmonary arterial hypertension (PAH) outcomes have not been described in patients in the United States. METHODS Using the US-based Registry to Evaluate Early and Long-term PAH Disease Management (REVEAL), the prevalence, demographics, and outcomes in ever- versus never-smokers with PAH were determined. RESULTS Ever-smoking status was more prevalent in males (61.7%) than in females (42.9%) enrolled in REVEAL. Ever-smokers were older than never-smokers at the time of PAH diagnosis and REVEAL enrollment. The time to first hospitalization, transplant-free survival, and survival did not differ between ever- and never-smokers overall; however, in newly diagnosed males, ever-smoking was associated with earlier death (hazard ratio [HR] 1.8, 95% confidence interval [CI] 1.1-3.0; p = 0.0199), the composite of transplant or death (HR 2.2, 95% CI 1.4-3.6; p = 0.0008), and first hospitalization (HR 1.8, 95% CI 1.2-2.7; p = 0.0063), though smoking exposure (pack-years) did not differ between newly and previously diagnosed males. CONCLUSIONS REVEAL PAH data demonstrate that smoking prevalence in male PAH patients is disproportionate. The prevalence of cigarette smoking was significantly higher in males than females enrolled in REVEAL. Ever-smoking status was associated with increased age at PAH diagnosis and, in newly diagnosed male PAH patients, earlier time to hospitalization and shorter survival after PAH diagnosis.
Collapse
|
17
|
Cullivan S, Gaine S, Sitbon O. New trends in pulmonary hypertension. Eur Respir Rev 2023; 32:32/167/220211. [PMID: 36813291 PMCID: PMC9949382 DOI: 10.1183/16000617.0211-2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/12/2022] [Indexed: 02/24/2023] Open
Abstract
Pulmonary hypertension (PH) is a prevalent disease of the pulmonary vasculature that is characterised by considerable morbidity and mortality. Substantial efforts have been made in recent years to improve disease recognition, diagnosis and management, and this is reflected in current guidelines. The haemodynamic definition of PH has been revised and a definition for exercise PH has been provided. Risk stratification has been refined and the importance of comorbidities and phenotyping have been highlighted. These changes provide an opportunity to potentially identify pulmonary vascular disease at an earlier stage and to enhance patient-centred, goal-orientated treatment decisions. A promising fourth treatment pathway for pulmonary arterial hypertension and potential targeted therapies for group 3 PH are on the horizon, concepts which seemed inconceivable only a few years ago. Beyond medication, there is a greater appreciation for the importance of supervised training in stable PH and the possible role of interventional therapies in select cases. The landscape of PH is changing and it is characterised by progress, innovation and opportunities. In this article, we highlight some of the new trends in PH, with a specific focus on the revised European Society of Cardiology/European Respiratory Society 2022 guidelines for the diagnosis and management of PH.
Collapse
Affiliation(s)
- Sarah Cullivan
- National Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sean Gaine
- National Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Olivier Sitbon
- Department of Respiratory and Intensive Care Medicine, CHU Bicêtre, Paris-Saclay University, INSERM URM_S999, Le Kremlin-Bicêtre, France
| |
Collapse
|
18
|
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: 8.0] [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
|
19
|
Le Ribeuz H, Masson B, Dutheil M, Boët A, Beauvais A, Sabourin J, De Montpreville VT, Capuano V, Mercier O, Humbert M, Montani D, Antigny F. Involvement of SUR2/Kir6.1 channel in the physiopathology of pulmonary arterial hypertension. Front Cardiovasc Med 2023; 9:1066047. [PMID: 36704469 PMCID: PMC9871631 DOI: 10.3389/fcvm.2022.1066047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Aims We hypothesized that the ATP-sensitive K+ channels (KATP) regulatory subunit (ABCC9) contributes to PAH pathogenesis. ABCC9 gene encodes for two regulatory subunits of KATP channels: the SUR2A and SUR2B proteins. In the KATP channel, the SUR2 subunits are associated with the K+ channel Kir6.1. We investigated how the SUR2/Kir6.1 channel contributes to PAH pathogenesis and its potential as a therapeutic target in PAH. Methods and results Using in vitro, ex vivo, and in vivo approaches, we analyzed the localization and expression of SUR2A, SUR2B, and Kir6.1 in the pulmonary vasculature of controls and patients with PAH as in experimental pulmonary hypertension (PH) rat models and its contribution to PAH physiopathology. Finally, we deciphered the consequences of in vivo activation of SUR2/Kir6.1 in the monocrotaline (MCT)-induced PH model. We found that SUR2A, SUR2B, and Kir6.1 were expressed in the lungs of controls and patients with PAH and MCT-induced PH rat models. Organ bath studies showed that SUR2 activation by pinacidil induced relaxation of pulmonary arterial in rats and humans. In vitro experiments on human pulmonary arterial smooth muscle cells and endothelial cells (hPASMCs and hPAECs) in controls and PAH patients showed decreased cell proliferation and migration after SUR2 activation. We demonstrated that SUR2 activation in rat right ventricular (RV) cardiomyocytes reduced RV action potential duration by patch-clamp. Chronic pinacidil administration in control rats increased heart rate without changes in hemodynamic parameters. Finally, in vivo pharmacological activation of SUR2 on MCT and Chronic-hypoxia (CH)-induced-PH rats showed improved PH. Conclusion We showed that SUR2A, SUR2B, and Kir6.1 are presented in hPASMCs and hPAECs of controls and PAH patients. In vivo SUR2 activation reduced the MCT-induced and CH-induced PH phenotype, suggesting that SUR2 activation should be considered for treating PAH.
Collapse
Affiliation(s)
- Hélène Le Ribeuz
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Bastien Masson
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Mary Dutheil
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France
| | - Angèle Boët
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Antoine Beauvais
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Jessica Sabourin
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Orsay, France
| | | | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France
| | - Olaf Mercier
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Le Plessis Robinson, France
| | - Marc Humbert
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique–Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique–Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 « Hypertension Pulmonaire Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
20
|
Krompa A, Marino P. Diagnosis and management of pulmonary hypertension related to chronic respiratory disease. Breathe (Sheff) 2022; 18:220205. [PMID: 36865930 PMCID: PMC9973528 DOI: 10.1183/20734735.0205-2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 01/12/2023] Open
Abstract
Pulmonary hypertension (PH) is a recognised and significant complication of chronic lung disease (CLD) and hypoxia (referred to as group 3 PH) that is associated with increased morbidity, decreased quality of life and worse survival. The prevalence and severity of group 3 PH varies within the current literature, with the majority of CLD-PH patients tending to have non-severe disease. The aetiology of this condition is multifactorial and complex, while the prevailing pathogenetic mechanisms include hypoxic vasoconstriction, parenchymal lung (and vascular bed) destruction, vascular remodelling and inflammation. Comorbidities such as left heart dysfunction and thromboembolic disease can further confound the clinical picture. Noninvasive assessment is initially undertaken in suspected cases (e.g. cardiac biomarkers, lung function, echocardiogram), while haemodynamic evaluation with right heart catheterisation remains the diagnostic gold standard. For patients with suspected severe PH, those with a pulmonary vascular phenotype or when there is uncertainty regarding further management, referral to specialist PH centres for further investigation and definitive management is mandated. No disease-specific therapy is currently available for group 3 PH and the focus of management remains optimisation of the underlying lung therapy, along with treating hypoventilation syndromes as indicated.
Collapse
Affiliation(s)
- Anastasia Krompa
- Lane Fox Respiratory Service, Guy's and St Thomas’ Hospital NHS Foundation Trust, London, UK
| | - Philip Marino
- Lane Fox Respiratory Service, Guy's and St Thomas’ Hospital NHS Foundation Trust, London, UK,Corresponding author: Philip Marino ()
| |
Collapse
|
21
|
Aman J, Morrell NW, Rhodes CJ, Wilkins MR, Bogaard HJ. The SOX17 phenotype in pulmonary arterial hypertension: lessons for pathobiology and clinical management. Eur Respir J 2022; 60:2201438. [PMID: 37651375 DOI: 10.1183/13993003.01438-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/11/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Jurjan Aman
- Dept of Pulmonology, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Christopher J Rhodes
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Dept of Pulmonology, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Hoeper MM, Dwivedi K, Pausch C, Lewis RA, Olsson KM, Huscher D, Pittrow D, Grünig E, Staehler G, Vizza CD, Gall H, Distler O, Opitz C, Gibbs JSR, Delcroix M, Park DH, Ghofrani HA, Ewert R, Kaemmerer H, Kabitz HJ, Skowasch D, Behr J, Milger K, Lange TJ, Wilkens H, Seyfarth HJ, Held M, Dumitrescu D, Tsangaris I, Vonk-Noordegraaf A, Ulrich S, Klose H, Claussen M, Eisenmann S, Schmidt KH, Swift AJ, Thompson AAR, Elliot CA, Rosenkranz S, Condliffe R, Kiely DG, Halank M. Phenotyping of idiopathic pulmonary arterial hypertension: a registry analysis. THE LANCET. RESPIRATORY MEDICINE 2022; 10:937-948. [PMID: 35777416 PMCID: PMC9514996 DOI: 10.1016/s2213-2600(22)00097-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Among patients meeting diagnostic criteria for idiopathic pulmonary arterial hypertension (IPAH), there is an emerging lung phenotype characterised by a low diffusion capacity for carbon monoxide (DLCO) and a smoking history. The present study aimed at a detailed characterisation of these patients. METHODS We analysed data from two European pulmonary hypertension registries, COMPERA (launched in 2007) and ASPIRE (from 2001 onwards), to identify patients diagnosed with IPAH and a lung phenotype defined by a DLCO of less than 45% predicted and a smoking history. We compared patient characteristics, response to therapy, and survival of these patients to patients with classical IPAH (defined by the absence of cardiopulmonary comorbidities and a DLCO of 45% or more predicted) and patients with pulmonary hypertension due to lung disease (group 3 pulmonary hypertension). FINDINGS The analysis included 128 (COMPERA) and 185 (ASPIRE) patients with classical IPAH, 268 (COMPERA) and 139 (ASPIRE) patients with IPAH and a lung phenotype, and 910 (COMPERA) and 375 (ASPIRE) patients with pulmonary hypertension due to lung disease. Most patients with IPAH and a lung phenotype had normal or near normal spirometry, a severe reduction in DLCO, with the majority having no or a mild degree of parenchymal lung involvement on chest computed tomography. Patients with IPAH and a lung phenotype (median age, 72 years [IQR 65-78] in COMPERA and 71 years [65-76] in ASPIRE) and patients with group 3 pulmonary hypertension (median age 71 years [65-77] in COMPERA and 69 years [63-74] in ASPIRE) were older than those with classical IPAH (median age, 45 years [32-60] in COMPERA and 52 years [38-64] in ASPIRE; p<0·0001 for IPAH with a lung phenotype vs classical IPAH in both registries). While 99 (77%) patients in COMPERA and 133 (72%) patients in ASPIRE with classical IPAH were female, there was a lower proportion of female patients in the IPAH and a lung phenotype cohort (95 [35%] COMPERA; 75 [54%] ASPIRE), which was similar to group 3 pulmonary hypertension (336 [37%] COMPERA; 148 [39%] ASPIRE]). Response to pulmonary arterial hypertension therapies at first follow-up was available from COMPERA. Improvements in WHO functional class were observed in 54% of patients with classical IPAH, 26% of patients with IPAH with a lung phenotype, and 22% of patients with group 3 pulmonary hypertension (p<0·0001 for classical IPAH vs IPAH and a lung phenotype, and p=0·194 for IPAH and a lung phenotype vs group 3 pulmonary hypertension); median improvements in 6 min walking distance were 63 m, 25 m, and 23 m for these cohorts respectively (p=0·0015 for classical IPAH vs IPAH and a lung phenotype, and p=0·64 for IPAH and a lung phenotype vs group 3 pulmonary hypertension), and median reductions in N-terminal-pro-brain-natriuretic-peptide were 58%, 27%, and 16% respectively (p=0·0043 for classical IPAH vs IPAH and a lung phenotype, and p=0·14 for IPAH and a lung phenotype vs group 3 pulmonary hypertension). In both registries, survival of patients with IPAH and a lung phenotype (1 year, 89% in COMPERA and 79% in ASPIRE; 5 years, 31% in COMPERA and 21% in ASPIRE) and group 3 pulmonary hypertension (1 year, 78% in COMPERA and 64% in ASPIRE; 5 years, 26% in COMPERA and 18% in ASPIRE) was worse than survival of patients with classical IPAH (1 year, 95% in COMPERA and 98% in ASPIRE; 5 years, 84% in COMPERA and 80% in ASPIRE; p<0·0001 for IPAH with a lung phenotype vs classical IPAH in both registries). INTERPRETATION A cohort of patients meeting diagnostic criteria for IPAH with a distinct, presumably smoking-related form of pulmonary hypertension accompanied by a low DLCO, resemble patients with pulmonary hypertension due to lung disease rather than classical IPAH. These observations have pathogenetic, diagnostic, and therapeutic implications, which require further exploration. FUNDING COMPERA is funded by unrestricted grants from Acceleron, Bayer, GlaxoSmithKline, Janssen, and OMT. The ASPIRE Registry is supported by Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
Collapse
Affiliation(s)
- Marius M Hoeper
- Clinic of Respiratory Medicine, Hannover Medical School, member of the German Center of Lung Research (DZL), Germany.
| | - Krit Dwivedi
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Christine Pausch
- GWT-TUD, Epidemiological Centre, Technical University Dresden, Dresden, Germany
| | - Robert A Lewis
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Karen M Olsson
- Clinic of Respiratory Medicine, Hannover Medical School, member of the German Center of Lung Research (DZL), Germany
| | - Doerte Huscher
- Institute of Biometry and Clinical Epidemiology, and Berlin Insitute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - David Pittrow
- GWT-TUD, Epidemiological Centre, Technical University Dresden, Dresden, Germany; Institute for Clinical Pharmacology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Ekkehard Grünig
- Center for Pulmonary Hypertension, Thoraxklinik at Heidelberg University Hospital, Translational Lung Research Center Heidelberg, member of the German Center for Lung Research (DZL), Germany
| | | | - Carmine Dario Vizza
- Dipartimento di Scienze Cliniche Internistiche, Anestiologiche e Cardiolohiche, Sapienza, University of Rome, Rome, Italy
| | - Henning Gall
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian Opitz
- Department of Cardiology, DRK Kliniken Berlin Westend, Berlin, Germany
| | - John Simon R Gibbs
- Department of Cardiology, National Heart & Lung Institute, Imperial College London, London, UK
| | - Marion Delcroix
- Clinical Department of Respiratory Diseases, University Hospitals of Leuven and Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, Katholieke Universiteit Leuven University of Leuven, Leuven, Belgium
| | - Da-Hee Park
- Clinic of Respiratory Medicine, Hannover Medical School, member of the German Center of Lung Research (DZL), Germany
| | - Hossein Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, Giessen, Germany; Department of Medicine, Imperial College London, London, UK
| | - Ralf Ewert
- Clinic of Internal Medicine, Department of Respiratory Medicine, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Harald Kaemmerer
- Deutsches Herzzentrum München, Klinik für angeborene Herzfehler und Kinderkardiologie; TU München, Munich, Germany
| | - Hans-Joachim Kabitz
- Gemeinnützige Krankenhausbetriebsgesellschaft Konstanz, Medizinische Klinik II, Konstanz, Germany
| | - Dirk Skowasch
- Universitätsklinikum Bonn, Medizinische Klinik und Poliklinik II, Innere Medizin - Kardiologie/Pneumologie, Bonn, Germany
| | - Juergen Behr
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Germany
| | - Katrin Milger
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Germany
| | - Tobias J Lange
- University Medical Center Regensburg, Department of Internal Medicine II, Regensburg, Germany
| | - Heinrike Wilkens
- Klinik für Innere Medizin V, Pneumologie, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - Hans-Jürgen Seyfarth
- Universitätsklinikum Leipzig, Medizinische Klinik und Poliklinik II, Abteilung für Pneumologie, Leipzig, Germany
| | - Matthias Held
- Department of Internal Medicine, Respiratory Medicine and Ventilatory Support, Medical Mission Hospital, Central Clinic Würzburg, Germany
| | - Daniel Dumitrescu
- Clinic for General and Interventional Cardiology and Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Iraklis Tsangaris
- Attikon University Hospital, 2nd Critical Care Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Anton Vonk-Noordegraaf
- Amsterdam UMC, Vrije Universiteit Amsterdam, dept of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Silvia Ulrich
- Clinic of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hans Klose
- Department of Respiratory Medicine, Eppendorf University Hospital, Hamburg, Germany
| | - Martin Claussen
- LungenClinic Grosshansdorf, Fachabteilung Pneumologie, Großhansdorf, Germany
| | - Stephan Eisenmann
- Universitätsklinikum Halle, Klinik für Innere Medizin I, Department of Respiratory Medicine, Halle, Germany
| | - Kai-Helge Schmidt
- Department of Cardiology and Center of Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Andrew J Swift
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alfred A Roger Thompson
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Charlie A Elliot
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Stephan Rosenkranz
- Clinic III for Internal Medicine (Cardiology) and Center for Molecular Medicine, and the Cologne Cardiovascular Research Center, University of Cologne, Germany
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Michael Halank
- Universitätsklinikum Carl Gustav Carus der Technischen Universität Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| |
Collapse
|
23
|
Taha F, Southgate L. Molecular genetics of pulmonary hypertension in children. Curr Opin Genet Dev 2022; 75:101936. [PMID: 35772304 PMCID: PMC9763127 DOI: 10.1016/j.gde.2022.101936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/20/2022] [Accepted: 05/19/2022] [Indexed: 12/24/2022]
Abstract
Until recently, the molecular aetiology of paediatric pulmonary hypertension (PH) was relatively poorly understood. While the TGF-β/BMP pathway was recognised as central to disease progression, genetic analyses in children were largely confined to targeted screening of risk genes in small cohorts, with clinical management extrapolated from adult data. In recent years, next-generation sequencing has highlighted notable differences in the genetic architecture underlying childhood-onset cases, with a higher genetic burden in children partly explained by comorbidities such as congenital heart disease. Here, we review recent genetic advances in paediatric PH and highlight important risk factors such as dysregulation of the transcription factors SOX17 and TBX4. Given the poorer prognosis in paediatric cases, molecular diagnosis offers a vital tool to enhance clinical care of children with PH.
Collapse
Affiliation(s)
- Fatima Taha
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK.
| |
Collapse
|
24
|
Abstract
Pulmonary hypertension (PH) because of chronic lung disease is categorized as Group 3 PH in the most recent classification system. Prevalence of these diseases is increasing over time, creating a growing need for effective therapeutic options. Recent approval of the first pulmonary arterial hypertension therapy for the treatment of Group 3 PH related to interstitial lung disease represents an encouraging advancement. This review focuses on molecular mechanisms contributing to pulmonary vasculopathy in chronic hypoxia, the pathology and epidemiology of Group 3 PH, the right ventricular dysfunction observed in this population and clinical trial data that inform the use of pulmonary vasodilators in Group 3 PH.
Collapse
Affiliation(s)
- Navneet Singh
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI
| | - Peter Dorfmüller
- Department of Pathology, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Germany (P.D.).,German Center for Lung Research (DZL), Giessen, Germany (P.D.)
| | - Oksana A Shlobin
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA (O.A.S.)
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI.,Department of Health Services, Policy and Practice (C.E.V.), Brown University, Providence, RI
| |
Collapse
|
25
|
Aldred MA, Morrell NW, Guignabert C. New Mutations and Pathogenesis of Pulmonary Hypertension: Progress and Puzzles in Disease Pathogenesis. Circ Res 2022; 130:1365-1381. [PMID: 35482831 PMCID: PMC9897592 DOI: 10.1161/circresaha.122.320084] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex multifactorial disease with poor prognosis characterized by functional and structural alterations of the pulmonary circulation causing marked increase in pulmonary vascular resistance, ultimately leading to right heart failure and death. Mutations in the gene encoding BMPRII-a receptor for the TGF-β (transforming growth factor-beta) superfamily-account for over 70% of families with PAH and ≈20% of sporadic cases. In recent years, however, less common or rare mutations in other genes have been identified. This review will consider how these newly discovered PAH genes could help to provide a better understanding of the molecular and cellular bases of the maintenance of the pulmonary vascular integrity, as well as their role in the PAH pathogenesis underlying occlusion of arterioles in the lung. We will also discuss how insights into the genetic contributions of these new PAH-related genes may open up new therapeutic targets for this, currently incurable, cardiopulmonary disorder.
Collapse
Affiliation(s)
- Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicholas W Morrell
- University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, UK
| | - Christophe Guignabert
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France,Université Paris-Saclay, Faculté de Médecine, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
26
|
Le Ribeuz H, Masson B, Capuano V, Dutheil M, Gooroochurn H, Boët A, Ghigna MR, De Montpreville V, Girerd B, Lambert M, Mercier O, Chung WK, Humbert M, Montani D, Antigny F. SUR1 as a New Therapeutic Target for Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2022; 66:539-554. [PMID: 35175177 DOI: 10.1165/rcmb.2021-0180oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mutations in ABCC8 have been identified in pulmonary arterial hypertension (PAH). ABCC8 encodes SUR1, a regulatory subunit of the ATP-sensitive-potassium channel Kir6.2. However, the pathophysiological role of the SUR1/Kir6.2 channel in PAH is unknown. We hypothesized that activation of SUR1 could be a novel potential target for PAH. We analysed the expression of SUR1/Kir6.2 in the lungs and pulmonary artery (PA) in human PAH or experimental pulmonary hypertension (PH). The contribution of SUR1 in human or rat PA tone was evaluated, and we measured the consequences of in vivo activation of SUR1 in control and PH rats. SUR1 and Kir6.2 protein expression was not reduced in the lungs or human pulmonary arterial endothelial cells and smooth muscle cells (hPAECs and hPASMCs) from PAH or experimentally induced PH. We showed that pharmacological activation of SUR1 by 3 different SUR1 activators (diazoxide, VU0071063, and NN414) leads to PA relaxation. Conversely, the inhibition of SUR1/Kir6.2 channels causes PA constriction. In vivo, long- and short-term activation of SUR1 with diazoxide reversed monocrotaline-induced PH in rats. Additionally, in vivo diazoxide application (short protocol) reduced the severity of PH in chronic-hypoxia rats. Moreover, 3 weeks of diazoxide exposure in control rats had no cardiovascular effects. Finally, in vivo, activation of SUR1 with NN414 reduced monocrotaline-induced PH in rats. In PAH and experimental PH, the expression of SUR1/Kir6.2 was still presented. In vivo pharmacological SUR1 activation by two different molecules alleviated experimental PH, providing proof-of-concept that SUR1 activation should be considered for PAH and evaluated more thoroughly.
Collapse
Affiliation(s)
| | | | | | - Mary Dutheil
- INSERM U999, 130034, Le Plessis Robinson, France
| | | | - Angèle Boët
- INSERM U999, 130034, Le Plessis Robinson, France
| | - Maria-Rosa Ghigna
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,School of Medicine, Université Paris-Sud / Université Paris Saclay, Le Kremlin-Bicêtre, France
| | | | - Barbara Girerd
- INSERM U999, 130034, pneumolgie, Le Plessis Robinson, France
| | - Mélanie Lambert
- INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Olaf Mercier
- INSERM U999, 130034, Thoracic Surgery , Le Plessis Robinson, France
| | - Wendy K Chung
- Departments of Pediatrics and Medicine Columbia University, New York, New York, United States
| | - Marc Humbert
- INSERM U999, 130034, Le Plessis Robinson, France
| | - David Montani
- CHU de Bicetre, Service de Pneumologie, Le Kremlin Bicetre, France.,INSERM UMRS 999, Hôpital Marie Lannelongue, Le plessis robinson, France
| | | |
Collapse
|
27
|
Leopold JA. Personalizing treatments for patients based on cardiovascular phenotyping. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022; 7:4-16. [PMID: 36778892 PMCID: PMC9913616 DOI: 10.1080/23808993.2022.2028548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Introduction Cardiovascular disease persists as the leading cause of death worldwide despite continued advances in diagnostics and therapeutics. Our current approach to patients with cardiovascular disease is rooted in reductionism, which presupposes that all patients share a similar phenotype and will respond the same to therapy; however, this is unlikely as cardiovascular diseases exhibit complex heterogeneous phenotypes. Areas covered With the advent of high-throughput platforms for omics testing, phenotyping cardiovascular diseases has advanced to incorporate large-scale molecular data with classical history, physical examination, and laboratory results. Findings from genomics, proteomics, and metabolomics profiling have been used to define more precise cardiovascular phenotypes and predict adverse outcomes in population-based and disease-specific patient cohorts. These molecular data have also been utilized to inform drug efficacy based on a patient's unique phenotype. Expert opinion Multiscale phenotyping of cardiovascular disease has revealed diversity among patients that can be used to personalize pharmacotherapies and predict outcomes. Nonetheless, precision phenotyping for cardiovascular disease remains a nascent field that has not yet translated into widespread clinical practice despite its many potential advantages for patient care. Future endeavors that demonstrate improved pharmacotherapeutic responses and associated reduction in adverse events will facilitate mainstream adoption of precision cardiovascular phenotyping.
Collapse
Affiliation(s)
- Jane A. Leopold
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, 77 Ave Louis Pasteur, NRB0630K, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Škorić-Milosavljević D, Lahrouchi N, Bosada FM, Dombrowsky G, Williams SG, Lesurf R, Tjong FVY, Walsh R, El Bouchikhi I, Breckpot J, Audain E, Ilgun A, Beekman L, Ratbi I, Strong A, Muenke M, Heide S, Muir AM, Hababa M, Cross L, Zhou D, Pastinen T, Zackai E, Atmani S, Ouldim K, Adadi N, Steindl K, Rauch A, Brook D, Wilsdon A, Kuipers I, Blom NA, Mulder BJ, Mefford HC, Keren B, Joset P, Kruszka P, Thiffault I, Sheppard SE, Roberts A, Lodder EM, Keavney BD, Clur SAB, Mital S, Hitz MP, Christoffels VM, Postma AV, Bezzina CR. Rare variants in KDR, encoding VEGF Receptor 2, are associated with tetralogy of Fallot. Genet Med 2021; 23:1952-1960. [PMID: 34113005 PMCID: PMC8486653 DOI: 10.1038/s41436-021-01212-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Rare genetic variants in KDR, encoding the vascular endothelial growth factor receptor 2 (VEGFR2), have been reported in patients with tetralogy of Fallot (TOF). However, their role in disease causality and pathogenesis remains unclear. METHODS We conducted exome sequencing in a familial case of TOF and large-scale genetic studies, including burden testing, in >1,500 patients with TOF. We studied gene-targeted mice and conducted cell-based assays to explore the role of KDR genetic variation in the etiology of TOF. RESULTS Exome sequencing in a family with two siblings affected by TOF revealed biallelic missense variants in KDR. Studies in knock-in mice and in HEK 293T cells identified embryonic lethality for one variant when occurring in the homozygous state, and a significantly reduced VEGFR2 phosphorylation for both variants. Rare variant burden analysis conducted in a set of 1,569 patients of European descent with TOF identified a 46-fold enrichment of protein-truncating variants (PTVs) in TOF cases compared to controls (P = 7 × 10-11). CONCLUSION Rare KDR variants, in particular PTVs, strongly associate with TOF, likely in the setting of different inheritance patterns. Supported by genetic and in vivo and in vitro functional analysis, we propose loss-of-function of VEGFR2 as one of the mechanisms involved in the pathogenesis of TOF.
Collapse
Affiliation(s)
- Doris Škorić-Milosavljević
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Najim Lahrouchi
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Fernanda M Bosada
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Gregor Dombrowsky
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
| | - Simon G Williams
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Robert Lesurf
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Fleur V Y Tjong
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Ihssane El Bouchikhi
- Laboratory of Medical Genetics and Oncogenetics, HASSAN II University Hospital, Fez, Morocco
| | - Jeroen Breckpot
- Center for Human Genetics Leuven and Catholic University Leuven, Leuven, Belgium
| | - Enrique Audain
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
| | - Aho Ilgun
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Leander Beekman
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Ilham Ratbi
- Centre de Recherche en Génomique des Pathologies Humaines (GENOPATH), Faculté de Médecine et de Pharmacie, Mohammed V University of Rabat, Rabat, Morocco
- Département de génétique médicale, Institut National d'Hygiène, Rabat, Morocco
| | - Alanna Strong
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maximilian Muenke
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Solveig Heide
- Département de génétique, Hôpital Pitié-Salpêtrière, APHP Sorbonne Université, Paris, France
| | - Alison M Muir
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
| | - Mariam Hababa
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Laura Cross
- Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Dihong Zhou
- Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Tomi Pastinen
- Center for Pediatric Genomic Medicine, Children's Mercy Hospital and School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Elaine Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Samir Atmani
- HASSAN II University Hospital, Fez, Morocco
- University of Sidi Mohammed Ben Abdellah, Fez, Morocco
| | - Karim Ouldim
- Faculty of Medicine and Pharmacy, Medical Genetics and Oncogenetics Unit, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Najlae Adadi
- Centre de Recherche en Génomique des Pathologies Humaines (GENOPATH), Faculté de Médecine et de Pharmacie, Mohammed V University of Rabat, Rabat, Morocco
- Département de génétique médicale, Institut National d'Hygiène, Rabat, Morocco
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - David Brook
- University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Anna Wilsdon
- University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Irene Kuipers
- Department of Pediatric Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Nico A Blom
- Department of Pediatric Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Barbara J Mulder
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Heather C Mefford
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
| | - Boris Keren
- Département de génétique, Hôpital Pitié-Salpêtrière, APHP Sorbonne Université, Paris, France
| | - Pascal Joset
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Paul Kruszka
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Thiffault
- Center for Pediatric Genomic Medicine, Children's Mercy Hospital and School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Sarah E Sheppard
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amy Roberts
- Department of Cardiology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Sally-Ann B Clur
- Department of Pediatric Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Seema Mital
- The Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Marc-Philip Hitz
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site, Kiel, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Alex V Postma
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands.
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Reuter MS, Chaturvedi RR, Jobling RK, Pellecchia G, Hamdan O, Sung WW, Nalpathamkalam T, Attaluri P, Silversides CK, Wald RM, Marshall CR, Williams S, Keavney BD, Thiruvahindrapuram B, Scherer SW, Bassett AS. Clinical Genetic Risk Variants Inform a Functional Protein Interaction Network for Tetralogy of Fallot. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003410. [PMID: 34328347 PMCID: PMC8373675 DOI: 10.1161/circgen.121.003410] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Tetralogy of Fallot (TOF)-the most common cyanotic heart defect in newborns-has evidence of multiple genetic contributing factors. Identifying variants that are clinically relevant is essential to understand patient-specific disease susceptibility and outcomes and could contribute to delineating pathomechanisms. METHODS Using a clinically driven strategy, we reanalyzed exome sequencing data from 811 probands with TOF, to identify rare loss-of-function and other likely pathogenic variants in genes associated with congenital heart disease. RESULTS We confirmed a major contribution of likely pathogenic variants in FLT4 (VEGFR3 [vascular endothelial growth factor receptor 3]; n=14) and NOTCH1 (n=10) and identified 1 to 3 variants in each of 21 other genes, including ATRX, DLL4, EP300, GATA6, JAG1, NF1, PIK3CA, RAF1, RASA1, SMAD2, and TBX1. In addition, multiple loss-of-function variants provided support for 3 emerging congenital heart disease/TOF candidate genes: KDR (n=4), IQGAP1 (n=3), and GDF1 (n=8). In total, these variants were identified in 63 probands (7.8%). Using the 26 composite genes in a STRING protein interaction enrichment analysis revealed a biologically relevant network (P=3.3×10-16), with VEGFR2 (vascular endothelial growth factor receptor 2; KDR) and NOTCH1 (neurogenic locus notch homolog protein 1) representing central nodes. Variants associated with arrhythmias/sudden death and heart failure indicated factors that could influence long-term outcomes. CONCLUSIONS The results are relevant to precision medicine for TOF. They suggest considerable clinical yield from genome-wide sequencing, with further evidence for KDR (VEGFR2) as a congenital heart disease/TOF gene and for VEGF (vascular endothelial growth factor) and Notch signaling as mechanisms in human disease. Harnessing the genetic heterogeneity of single gene defects could inform etiopathogenesis and help prioritize novel candidate genes for TOF.
Collapse
Affiliation(s)
- Miriam S. Reuter
- CGEn, Univ Health Network, Toronto, ON, Canada
- The Ctr for Applied Genomics, Univ Health Network, Toronto, ON, Canada
- Program in Genetics & Genome Biology, Univ Health Network, Toronto, ON, Canada
| | - Rajiv R. Chaturvedi
- Labatt Family Heart Ctr, Univ Health Network, Toronto, ON, Canada
- Ontario Fetal Ctr, Mt Sinai Hospital, Univ Health Network, Toronto, ON, Canada
- Ted Rogers Ctr for Heart Rsrch, Cardiac Genome Clinic, Univ Health Network, Toronto, ON, Canada
| | - Rebekah K. Jobling
- Ted Rogers Ctr for Heart Rsrch, Cardiac Genome Clinic, Univ Health Network, Toronto, ON, Canada
- Division of Clinical & Metabolic Genetics, Univ Health Network, Toronto, ON, Canada
- Genome Diagnostics, Dept of Paediatric Laboratory Medicine, The Hospital for Sick Children, Univ Health Network, Toronto, ON, Canada
| | | | - Omar Hamdan
- The Ctr for Applied Genomics, Univ Health Network, Toronto, ON, Canada
| | - Wilson W.L. Sung
- The Ctr for Applied Genomics, Univ Health Network, Toronto, ON, Canada
| | | | - Pratyusha Attaluri
- Medical Genomics Program, Dept of Molecular Genetics, Univ Health Network, Toronto, ON, Canada
| | - Candice K. Silversides
- Division of Cardiology, Toronto Congenital Cardiac Ctr for Adults at the Peter Munk Cardiac Ctr, Dept of Medicine, Univ Health Network, Toronto, ON, Canada
| | - Rachel M. Wald
- Labatt Family Heart Ctr, Univ Health Network, Toronto, ON, Canada
- Division of Cardiology, Toronto Congenital Cardiac Ctr for Adults at the Peter Munk Cardiac Ctr, Dept of Medicine, Univ Health Network, Toronto, ON, Canada
| | - Christian R. Marshall
- The Ctr for Applied Genomics, Univ Health Network, Toronto, ON, Canada
- Genome Diagnostics, Dept of Paediatric Laboratory Medicine, The Hospital for Sick Children, Univ Health Network, Toronto, ON, Canada
- Laboratory Medicine & Pathobiology, Univ Health Network, Toronto, ON, Canada
| | - Simon Williams
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine & Health, The Univ of Manchester, Manchester, UK
- Manchester Univ NHS Foundation Trust, Manchester Academic Health Science Ctr, Manchester, UK
| | - Bernard D. Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine & Health, The Univ of Manchester, Manchester, UK
- Manchester Univ NHS Foundation Trust, Manchester Academic Health Science Ctr, Manchester, UK
| | | | - Stephen W. Scherer
- The Ctr for Applied Genomics, Univ Health Network, Toronto, ON, Canada
- Program in Genetics & Genome Biology, Univ Health Network, Toronto, ON, Canada
- Dept of Molecular Genetics, Univ Health Network, Toronto, ON, Canada
- McLaughlin Ctr, Univ Health Network, Toronto, ON, Canada
| | - Anne S. Bassett
- Division of Cardiology, Toronto Congenital Cardiac Ctr for Adults at the Peter Munk Cardiac Ctr, Dept of Medicine, Univ Health Network, Toronto, ON, Canada
- Clinical Genetics Research Program, Ctr for Addiction & Mental Health, Toronto, ON, Canada
- The Dalglish Family 22q Clinic for Adults with 22q11.2 Deletion Syndrome, Dept of Psychiatry & Toronto General Rsrch Inst, Univ Health Network, Toronto, ON, Canada
- Dept of Psychiatry, Univ of Toronto, Univ Health Network, Toronto, ON, Canada
| |
Collapse
|
30
|
Wilkins MR. Personalized Medicine for Pulmonary Hypertension:: The Future Management of Pulmonary Hypertension Requires a New Taxonomy. Clin Chest Med 2021; 42:207-216. [PMID: 33541614 DOI: 10.1016/j.ccm.2020.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pulmonary hypertension is a convergent phenotype that presents late in the natural history of the condition. The current clinical classification of patients lacks granularity, and this impacts on the development and deployment of treatment. Deep molecular phenotyping using platform 'omic' technologies is beginning to reveal the genetic and molecular architecture that underlies the phenotype, promising better targeting of patients with new treatments. The future treatment of pulmonary hypertension depends on the integration of clinical and molecular information to create a new taxonomy that defines patient groups coupled to druggable targets.
Collapse
Affiliation(s)
- Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
31
|
Zhu N, Swietlik EM, Welch CL, Pauciulo MW, Hagen JJ, Zhou X, Guo Y, Karten J, Pandya D, Tilly T, Lutz KA, Martin JM, Treacy CM, Rosenzweig EB, Krishnan U, Coleman AW, Gonzaga-Jauregui C, Lawrie A, Trembath RC, Wilkins MR, Morrell NW, Shen Y, Gräf S, Nichols WC, Chung WK. Rare variant analysis of 4241 pulmonary arterial hypertension cases from an international consortium implicates FBLN2, PDGFD, and rare de novo variants in PAH. Genome Med 2021; 13:80. [PMID: 33971972 PMCID: PMC8112021 DOI: 10.1186/s13073-021-00891-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a lethal vasculopathy characterized by pathogenic remodeling of pulmonary arterioles leading to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. PAH can be associated with other diseases (APAH: connective tissue diseases, congenital heart disease, and others) but often the etiology is idiopathic (IPAH). Mutations in bone morphogenetic protein receptor 2 (BMPR2) are the cause of most heritable cases but the vast majority of other cases are genetically undefined. METHODS To identify new risk genes, we utilized an international consortium of 4241 PAH cases with exome or genome sequencing data from the National Biological Sample and Data Repository for PAH, Columbia University Irving Medical Center, and the UK NIHR BioResource - Rare Diseases Study. The strength of this combined cohort is a doubling of the number of IPAH cases compared to either national cohort alone. We identified protein-coding variants and performed rare variant association analyses in unrelated participants of European ancestry, including 1647 IPAH cases and 18,819 controls. We also analyzed de novo variants in 124 pediatric trios enriched for IPAH and APAH-CHD. RESULTS Seven genes with rare deleterious variants were associated with IPAH with false discovery rate smaller than 0.1: three known genes (BMPR2, GDF2, and TBX4), two recently identified candidate genes (SOX17, KDR), and two new candidate genes (fibulin 2, FBLN2; platelet-derived growth factor D, PDGFD). The new genes were identified based solely on rare deleterious missense variants, a variant type that could not be adequately assessed in either cohort alone. The candidate genes exhibit expression patterns in lung and heart similar to that of known PAH risk genes, and most variants occur in conserved protein domains. For pediatric PAH, predicted deleterious de novo variants exhibited a significant burden compared to the background mutation rate (2.45×, p = 2.5e-5). At least eight novel pediatric candidate genes carrying de novo variants have plausible roles in lung/heart development. CONCLUSIONS Rare variant analysis of a large international consortium identified two new candidate genes-FBLN2 and PDGFD. The new genes have known functions in vasculogenesis and remodeling. Trio analysis predicted that ~ 15% of pediatric IPAH may be explained by de novo variants.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jacob J Hagen
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Xueya Zhou
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Yicheng Guo
- Department of Systems Biology, Columbia University, New York, NY, USA
| | | | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Tobias Tilly
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jennifer M Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
| | - Carmen M Treacy
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Erika B Rosenzweig
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Usha Krishnan
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Anna W Coleman
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Richard C Trembath
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Martin R Wilkins
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | | | | | | | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
- Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
32
|
Pienkos S, Gallego N, Condon DF, Cruz-Utrilla A, Ochoa N, Nevado J, Arias P, Agarwal S, Patel H, Chakraborty A, Lapunzina P, Escribano P, Tenorio-Castaño J, de Jesús Pérez VA. Novel TNIP2 and TRAF2 Variants Are Implicated in the Pathogenesis of Pulmonary Arterial Hypertension. Front Med (Lausanne) 2021; 8:625763. [PMID: 33996849 PMCID: PMC8119639 DOI: 10.3389/fmed.2021.625763] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is a rare disease characterized by pulmonary vascular remodeling and right heart failure. Specific genetic variants increase the incidence of PAH in carriers with a family history of PAH, those who suffer from certain medical conditions, and even those with no apparent risk factors. Inflammation and immune dysregulation are related to vascular remodeling in PAH, but whether genetic susceptibility modifies the PAH immune response is unclear. TNIP2 and TRAF2 encode for immunomodulatory proteins that regulate NF-κB activation, a transcription factor complex associated with inflammation and vascular remodeling in PAH. Methods: Two unrelated families with PAH cases underwent whole-exome sequencing (WES). A custom pipeline for variant prioritization was carried out to obtain candidate variants. To determine the impact of TNIP2 and TRAF2 in cell proliferation, we performed an MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay on healthy lung pericytes transfected with siRNA specific for each gene. To measure the effect of loss of TNIP2 and TRAF2 on NF-kappa-beta (NF-κB) activity, we measured levels of Phospho-p65-NF-κB in siRNA-transfected pericytes using western immunoblotting. Results: We discovered a novel missense variant in the TNIP2 gene in two affected individuals from the same family. The two patients had a complex form of PAH with interatrial communication and scleroderma. In the second family, WES of the proband with PAH and primary biliary cirrhosis revealed a de novo protein-truncating variant in the TRAF2. The knockdown of TNIP2 and TRAF2 increased NF-κB activity in healthy lung pericytes, which correlated with a significant increase in proliferation over 24 h. Conclusions: We have identified two rare novel variants in TNIP2 and TRAF2 using WES. We speculate that loss of function in these genes promotes pulmonary vascular remodeling by allowing overactivation of the NF-κB signaling activity. Our findings support a role for WES in helping identify novel genetic variants associated with dysfunctional immune response in PAH.
Collapse
Affiliation(s)
- Shaun Pienkos
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Natalia Gallego
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Alejandro Cruz-Utrilla
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Nuria Ochoa
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Julián Nevado
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Pedro Arias
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Pablo Lapunzina
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Pilar Escribano
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Jair Tenorio-Castaño
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Vinicio A. de Jesús Pérez
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
33
|
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: 1] [Impact Index Per Article: 0.3] [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
|
34
|
Wu Y, Wharton J, Walters R, Vasilaki E, Aman J, Zhao L, Wilkins MR, Rhodes CJ. The pathophysiological role of novel pulmonary arterial hypertension gene SOX17. Eur Respir J 2021; 58:13993003.04172-2020. [PMID: 33632800 DOI: 10.1183/13993003.04172-2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/08/2021] [Indexed: 11/05/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease predominantly targeting pre-capillary blood vessels. Adverse structural remodelling and increased pulmonary vascular resistance result in cardiac hypertrophy and ultimately failure of the right ventricle. Recent whole-genome and whole-exome sequencing studies have identified SOX17 as a novel risk gene in PAH, with a dominant mode of inheritance and incomplete penetrance. Rare deleterious variants in the gene and more common variants in upstream enhancer sites have both been associated with the disease, and a deficiency of SOX17 expression may predispose to PAH. This review aims to consolidate the evidence linking genetic variants in SOX17 to PAH, and explores the numerous targets and effects of the transcription factor, focusing on the pulmonary vasculature and the pathobiology of PAH.
Collapse
Affiliation(s)
- Yukyee Wu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rachel Walters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Eleni Vasilaki
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jurjan Aman
- National Heart and Lung Institute, Imperial College London, London, UK.,VUmc, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lan Zhao
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
35
|
Swietlik EM, Greene D, Zhu N, Megy K, Cogliano M, Rajaram S, Pandya D, Tilly T, Lutz KA, Welch CC, Pauciulo MW, Southgate L, Martin JM, Treacy CM, Penkett CJ, Stephens JC, Bogaard HJ, Church C, Coghlan G, Coleman AW, Condliffe R, Eichstaedt CA, Eyries M, Gall H, Ghio S, Girerd B, Grünig E, Holden S, Howard L, Humbert M, Kiely DG, Kovacs G, Lordan J, Machado RD, MacKenzie Ross RV, McCabe C, Moledina S, Montani D, Olschewski H, Pepke-Zaba J, Price L, Rhodes CJ, Seeger W, Soubrier F, Suntharalingam J, Toshner MR, Vonk Noordegraaf A, Wharton J, Wild JM, Wort SJ, Lawrie A, Wilkins MR, Trembath RC, Shen Y, Chung WK, Swift AJ, Nichols WC, Morrell NW, Gräf S. Bayesian Inference Associates Rare KDR Variants with Specific Phenotypes in Pulmonary Arterial Hypertension. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2020; 14. [PMID: 33320693 PMCID: PMC7892262 DOI: 10.1161/circgen.120.003155] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 11/29/2020] [Indexed: 11/26/2022]
Abstract
Background - Approximately 25% of patients with pulmonary arterial hypertension (PAH) have been found to harbor rare mutations in disease-causing genes. To identify missing heritability in PAH we integrated deep phenotyping with whole-genome sequencing data using Bayesian statistics. Methods - We analyzed 13,037 participants enrolled in the NIHR BioResource - Rare Diseases (NBR) study, of which 1,148 were recruited to the PAH domain. To test for genetic associations between genes and selected phenotypes of pulmonary hypertension (PH), we used the Bayesian rare-variant association method BeviMed. Results - Heterozygous, high impact, likely loss-of-function variants in the Kinase Insert Domain Receptor (KDR) gene were strongly associated with significantly reduced transfer coefficient for carbon monoxide (KCO, posterior probability (PP)=0.989) and older age at diagnosis (PP=0.912). We also provide evidence for familial segregation of a rare nonsense KDR variant with these phenotypes. On computed tomographic imaging of the lungs, a range of parenchymal abnormalities were observed in the five patients harboring these predicted deleterious variants in KDR. Four additional PAH cases with rare likely loss-of-function variants in KDR were independently identified in the US PAH Biobank cohort with similar phenotypic characteristics. Conclusions - The Bayesian inference approach allowed us to independently validate KDR, which encodes for the Vascular Endothelial Growth Factor Receptor 2 (VEGFR2), as a novel PAH candidate gene. Furthermore, this approach specifically associated high impact likely loss-of-function variants in the genetically constrained gene with distinct phenotypes. These findings provide evidence for KDR being a clinically actionable PAH gene and further support the central role of the vascular endothelium in the pathobiology of PAH.
Collapse
Affiliation(s)
- Emilia M. Swietlik
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Daniel Greene
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Na Zhu
- Department of Pediatrics (N.Z., C.C.L.W.), Columbia University, NY
- Department of Systems Biology (N.Z., Y.S.), Columbia University, NY
| | - Karyn Megy
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Marcella Cogliano
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - Smitha Rajaram
- Sheffield Teaching Hospitals NHS Foundation Trust, United Kingdom (S.R.)
| | - Divya Pandya
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Tobias Tilly
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Katie A. Lutz
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
| | | | - Michael W. Pauciulo
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (M.W.P., W.C.N.)
| | - Laura Southgate
- Molecular & Clinical Sciences Research Institute, St George’s, University of London, United Kingdom (L.S., R.D.M.)
| | - Jennifer M. Martin
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Carmen M. Treacy
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
| | - Christopher J. Penkett
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Jonathan C. Stephens
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| | - Harm J. Bogaard
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands (H.J.B., A.V.N.)
| | - Colin Church
- Golden Jubilee National Hospital, Glasgow (C.C.)
| | | | - Anna W. Coleman
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (R.C., D.G.K.)
| | - Christina A. Eichstaedt
- Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University (C.A.E.)
- Center for Pulmonary Hypertension, Thoraxklinik gGmbH Heidelberg at Heidelberg University Hospital (C.A.E., E.G.)
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany (C.A.E., E.G.)
| | - Mélanie Eyries
- Département de génétique, hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris & UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France (M.E., F.S.)
| | - Henning Gall
- University of Giessen & Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany (H.G., W.S.)
| | - Stefano Ghio
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy (S. Ghio)
| | - Barbara Girerd
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay (B.G., M.H., D.M.)
- AP-HP, Service de Pneumologie, Centre de référence de l’hypertension pulmonaire (B.G., M.H., D.M.)
- INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France (B.G., M.H., D.M.)
| | - Ekkehard Grünig
- Center for Pulmonary Hypertension, Thoraxklinik gGmbH Heidelberg at Heidelberg University Hospital (C.A.E., E.G.)
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany (C.A.E., E.G.)
| | - Simon Holden
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge (S.H., N.W.M.)
| | - Luke Howard
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay (B.G., M.H., D.M.)
- AP-HP, Service de Pneumologie, Centre de référence de l’hypertension pulmonaire (B.G., M.H., D.M.)
- INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France (B.G., M.H., D.M.)
| | - David G. Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, United Kingdom (R.C., D.G.K.)
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research (G.K., H.O.)
- Medical University of Graz, Austria (G.K., H.O.)
| | - Jim Lordan
- Freeman Hospital, Newcastle upon Tyne (J.L.)
| | - Rajiv D. Machado
- Molecular & Clinical Sciences Research Institute, St George’s, University of London, United Kingdom (L.S., R.D.M.)
| | | | - Colm McCabe
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
- Royal Brompton & Harefield NHS Foundation Trust (C.M., L.P., S.J.W.)
| | | | - David Montani
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay (B.G., M.H., D.M.)
- AP-HP, Service de Pneumologie, Centre de référence de l’hypertension pulmonaire (B.G., M.H., D.M.)
- INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France (B.G., M.H., D.M.)
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research (G.K., H.O.)
- Medical University of Graz, Austria (G.K., H.O.)
| | - Joanna Pepke-Zaba
- Royal Papworth Hospital NHS Foundation Trust (J.P.-Z., M.R.T., N.W.M.)
| | - Laura Price
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
- Royal Brompton & Harefield NHS Foundation Trust (C.M., L.P., S.J.W.)
| | - Christopher J. Rhodes
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - Werner Seeger
- University of Giessen & Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany (H.G., W.S.)
| | - Florent Soubrier
- Département de génétique, hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris & UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, France (M.E., F.S.)
| | | | - Mark R. Toshner
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
- Royal Papworth Hospital NHS Foundation Trust (J.P.-Z., M.R.T., N.W.M.)
| | - Anton Vonk Noordegraaf
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands (H.J.B., A.V.N.)
| | - John Wharton
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - James M. Wild
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - Stephen John Wort
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
- Royal Brompton & Harefield NHS Foundation Trust (C.M., L.P., S.J.W.)
| | | | | | | | - Allan Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - Martin R. Wilkins
- National Heart & Lung Institute, Imperial College London, United Kingdom (L.H., C.M., L.P., C.J.R., J.W., S.J.W., M.R.W.)
| | - Richard C. Trembath
- Department of Medical & Molecular Genetics, King’s College London, United Kingdom (R.C.T.)
| | - Yufeng Shen
- Department of Systems Biology (N.Z., Y.S.), Columbia University, NY
- Department of Biomedical Informatics (Y.S.), Columbia University, NY
| | | | - Andrew J. Swift
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield (M.C., J.M.W., A.L., A.J.S.)
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (K.A.L., M.W.P., A.W.C., W.C.N.)
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (M.W.P., W.C.N.)
| | - Nicholas W. Morrell
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge (S.H., N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust (J.P.-Z., M.R.T., N.W.M.)
| | - Stefan Gräf
- Department of Medicine (E.M.S., D.P., T.T., C.M.T., M.R.T., N.W.M., S. Gräf), University of Cambridge
- Department of Haematology (D.G., K.M., C.J.P., J.C.S., S. Gräf), University of Cambridge
- NIHR BioResource for Translational Research, Cambridge, United Kingdom (D.G., K.M., J.M.M., C.J.P., J.C.S., N.W.M., S. Gräf)
| |
Collapse
|
36
|
Swietlik EM, Prapa M, Martin JM, Pandya D, Auckland K, Morrell NW, Gräf S. 'There and Back Again'-Forward Genetics and Reverse Phenotyping in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:E1408. [PMID: 33256119 PMCID: PMC7760524 DOI: 10.3390/genes11121408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Although the invention of right heart catheterisation in the 1950s enabled accurate clinical diagnosis of pulmonary arterial hypertension (PAH), it was not until 2000 when the landmark discovery of the causative role of bone morphogenetic protein receptor type II (BMPR2) mutations shed new light on the pathogenesis of PAH. Since then several genes have been discovered, which now account for around 25% of cases with the clinical diagnosis of idiopathic PAH. Despite the ongoing efforts, in the majority of patients the cause of the disease remains elusive, a phenomenon often referred to as "missing heritability". In this review, we discuss research approaches to uncover the genetic architecture of PAH starting with forward phenotyping, which in a research setting should focus on stable intermediate phenotypes, forward and reverse genetics, and finally reverse phenotyping. We then discuss potential sources of "missing heritability" and how functional genomics and multi-omics methods are employed to tackle this problem.
Collapse
Affiliation(s)
- Emilia M. Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Matina Prapa
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Jennifer M. Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Kathryn Auckland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| |
Collapse
|
37
|
Pasupneti S, Tian W, Tu AB, Dahms P, Granucci E, Gandjeva A, Xiang M, Butcher EC, Semenza GL, Tuder RM, Jiang X, Nicolls MR. Endothelial HIF-2α as a Key Endogenous Mediator Preventing Emphysema. Am J Respir Crit Care Med 2020; 202:983-995. [PMID: 32515984 DOI: 10.1164/rccm.202001-0078oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Rationale: Endothelial injury may provoke emphysema, but molecular pathways of disease development require further discernment. Emphysematous lungs exhibit decreased expression of HIF-2α (hypoxia-inducible factor-2α)-regulated genes, and tobacco smoke decreases pulmonary HIF-2α concentrations. These findings suggest that decreased HIF-2α expression is important in the development of emphysema.Objectives: The objective of this study was to evaluate the roles of endothelial-cell (EC) HIF-2α in the pathogenesis of emphysema in mice.Methods: Mouse lungs were examined for emphysema after either the loss or the overexpression of EC Hif-2α. In addition, SU5416, a VEGFR2 inhibitor, was used to induce emphysema. Lungs were evaluated for HGF (hepatocyte growth factor), a protein involved in alveolar development and homeostasis. Lungs from patients with emphysema were measured for endothelial HIF-2α expression.Measurements and Main Results: EC Hif-2α deletion resulted in emphysema in association with fewer ECs and pericytes. After SU5416 exposure, EC Hif-2α-knockout mice developed more severe emphysema, whereas EC Hif-2α-overexpressing mice were protected. EC Hif-2α-knockout mice demonstrated lower levels of HGF. Human emphysema lung samples exhibited reduced EC HIF-2α expression.Conclusions: Here, we demonstrate a unique protective role for pulmonary endothelial HIF-2α and how decreased expression of this endogenous factor causes emphysema; its pivotal protective function is suggested by its ability to overcome VEGF antagonism. HIF-2α may maintain alveolar architecture by promoting vascular survival and associated HGF production. In summary, HIF-2α may be a key endogenous factor that prevents the development of emphysema, and its upregulation has the potential to foster lung health in at-risk patients.
Collapse
Affiliation(s)
- Shravani Pasupneti
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Wen Tian
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Allen B Tu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Petra Dahms
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Eric Granucci
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Aneta Gandjeva
- School of Medicine, University of Colorado, Colorado; and
| | - Menglan Xiang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Eugene C Butcher
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Gregg L Semenza
- Vascular Program, Institute for Cell Engineering.,Sidney Kimmel Comprehensive Cancer Center.,Department of Genetic Medicine.,Department of Pediatrics.,Department of Medicine.,Department of Oncology.,Department of Radiation Oncology, and.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rubin M Tuder
- School of Medicine, University of Colorado, Colorado; and
| | - Xinguo Jiang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| | - Mark R Nicolls
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California.,School of Medicine, Stanford University, Stanford, California
| |
Collapse
|
38
|
Le Ribeuz H, Capuano V, Girerd B, Humbert M, Montani D, Antigny F. Implication of Potassium Channels in the Pathophysiology of Pulmonary Arterial Hypertension. Biomolecules 2020; 10:biom10091261. [PMID: 32882918 PMCID: PMC7564204 DOI: 10.3390/biom10091261] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare and severe cardiopulmonary disease without curative treatments. PAH is a multifactorial disease that involves genetic predisposition, epigenetic factors, and environmental factors (drugs, toxins, viruses, hypoxia, and inflammation), which contribute to the initiation or development of irreversible remodeling of the pulmonary vessels. The recent identification of loss-of-function mutations in KCNK3 (KCNK3 or TASK-1) and ABCC8 (SUR1), or gain-of-function mutations in ABCC9 (SUR2), as well as polymorphisms in KCNA5 (Kv1.5), which encode two potassium (K+) channels and two K+ channel regulatory subunits, has revived the interest of ion channels in PAH. This review focuses on KCNK3, SUR1, SUR2, and Kv1.5 channels in pulmonary vasculature and discusses their pathophysiological contribution to and therapeutic potential in PAH.
Collapse
Affiliation(s)
- Hélène Le Ribeuz
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Véronique Capuano
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Barbara Girerd
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - David Montani
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
- Correspondence: or ; Tel.: +33-1-40-94-22-99
| |
Collapse
|
39
|
Gassmann M, Cowburn A, Gu H, Li J, Rodriguez M, Babicheva A, Jain PP, Xiong M, Gassmann NN, Yuan JXJ, Wilkins MR, Zhao L. Hypoxia-induced pulmonary hypertension-Utilizing experiments of nature. Br J Pharmacol 2020; 178:121-131. [PMID: 32464698 DOI: 10.1111/bph.15144] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
An increase in pulmonary artery pressure is a common observation in adult mammals exposed to global alveolar hypoxia. It is considered a maladaptive response that places an increased workload on the right ventricle. The mechanisms initiating and maintaining the elevated pressure are of considerable interest in understanding pulmonary vascular homeostasis. There is an expectation that identifying the key molecules in the integrated vascular response to hypoxia will inform potential drug targets. One strategy is to take advantage of experiments of nature, specifically, to understand the genetic basis for the inter-individual variation in the pulmonary vascular response to acute and chronic hypoxia. To date, detailed phenotyping of highlanders has focused on haematocrit and oxygen saturation rather than cardiovascular phenotypes. This review explores what we can learn from those studies with respect to the pulmonary circulation. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,University Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Andrew Cowburn
- National Heart and Lung Institute (NHLI), Imperial College London, Hammersmith Hospital, London, UK
| | - Hong Gu
- Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jia Li
- Clinical Physiology Laboratory, Institute of Pediatrics, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Mingmei Xiong
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Norina N Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Martin R Wilkins
- National Heart and Lung Institute (NHLI), Imperial College London, Hammersmith Hospital, London, UK
| | - Lan Zhao
- National Heart and Lung Institute (NHLI), Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
40
|
Affiliation(s)
- Florent Soubrier
- Genetics Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, UMR_S 1166-ICAN, Sorbonne-Université, INSERM, Paris, France
| |
Collapse
|
41
|
Cool CD, Kuebler WM, Bogaard HJ, Spiekerkoetter E, Nicolls MR, Voelkel NF. The hallmarks of severe pulmonary arterial hypertension: the cancer hypothesis-ten years later. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1115-L1130. [PMID: 32023082 PMCID: PMC9847334 DOI: 10.1152/ajplung.00476.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 01/25/2023] Open
Abstract
Severe forms of pulmonary arterial hypertension (PAH) are most frequently the consequence of a lumen-obliterating angiopathy. One pathobiological model is that the initial pulmonary vascular endothelial cell injury and apoptosis is followed by the evolution of phenotypically altered, apoptosis-resistant, proliferating cells and an inflammatory vascular immune response. Although there may be a vasoconstrictive disease component, the increased pulmonary vascular shear stress in established PAH is caused largely by the vascular wall pathology. In this review, we revisit the "quasi-malignancy concept" of severe PAH and examine to what extent the hallmarks of PAH can be compared with the hallmarks of cancer. The cancer model of severe PAH, based on the growth of abnormal vascular and bone marrow-derived cells, may enable the emergence of novel cell-based PAH treatment strategies.
Collapse
Affiliation(s)
- Carlyne D Cool
- Department of Pathology, University of Colorado, Anschuetz Campus, Aurora, Colorado
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitaetsmedizin, Berlin, Germany
| | - Harm Jan Bogaard
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - Norbert F Voelkel
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Swietlik EM, Gräf S, Morrell NW. The role of genomics and genetics in pulmonary arterial hypertension. Glob Cardiol Sci Pract 2020; 2020:e202013. [PMID: 33150157 PMCID: PMC7590931 DOI: 10.21542/gcsp.2020.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
43
|
Hernandez-Gonzalez I, Tenorio J, Palomino-Doza J, Martinez Meñaca A, Morales Ruiz R, Lago-Docampo M, Valverde Gomez M, Gomez Roman J, Enguita Valls AB, Perez-Olivares C, Valverde D, Gil Carbonell J, Garrido-Lestache Rodríguez-Monte E, del Cerro MJ, Lapunzina P, Escribano-Subias P. Clinical heterogeneity of Pulmonary Arterial Hypertension associated with variants in TBX4. PLoS One 2020; 15:e0232216. [PMID: 32348326 PMCID: PMC7190146 DOI: 10.1371/journal.pone.0232216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022] Open
Abstract
Background The knowledge of hereditary predisposition has changed our understanding of Pulmonary Arterial Hypertension. Genetic testing has been widely extended and the application of Pulmonary Arterial Hypertension specific gene panels has allowed its inclusion in the diagnostic workup and increase the diagnostic ratio compared to the traditional sequencing techniques. This is particularly important in the differential diagnosis between Pulmonary Arterial Hypertension and Pulmonary Venoocclusive Disease. Methods Since November 2011, genetic testing is offered to all patients with idiopathic, hereditable and associated forms of Pulmonary Arterial Hypertension or Pulmonary Venoocclusive Disease included in the Spanish Registry of Pulmonary Arterial Hypertension. Herein, we present the clinical phenotype and prognosis of all Pulmonary Arterial Hypertension patients with disease-associated variants in TBX4. Results Out of 579 adults and 45 children, we found in eight patients from seven families, disease-causing associated variants in TBX4. All adult patients had a moderate-severe reduction in diffusion capacity. However, we observed a wide spectrum of clinical presentations, including Pulmonary Venoocclusive Disease suspicion, interstitial lung disease, pulmonary vascular abnormalities and congenital heart disease. Conclusions Genetic testing is now essential for a correct diagnosis work-up in Pulmonary Arterial Hypertension. TBX4-associated Pulmonary Arterial Hypertension has marked clinical heterogeneity. In this regard, a genetic study is extremely useful to obtain an accurate diagnosis and provide appropriate management.
Collapse
Affiliation(s)
| | - Jair Tenorio
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institto de Salud Carlos III, Madrid, Spain
| | - Julian Palomino-Doza
- Department of Cardiology, Inherited Cardiac Disease Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
- Department of Cardiology, Pulmonary Hypertension Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Institto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Amaya Martinez Meñaca
- Department of Pneumology, Pulmonary Hypertension Unit, Lung Transplant Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Rafael Morales Ruiz
- Department of Radiology, Pulmonary Hypertension Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Mauro Lago-Docampo
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain
| | - María Valverde Gomez
- Department of Cardiology, Inherited Cardiac Disease Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Javier Gomez Roman
- Department of Pathology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Ana Belén Enguita Valls
- Department of Pathology, Pulmonary Hypertension Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Carmen Perez-Olivares
- Department of Cardiology, Pulmonary Hypertension Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Diana Valverde
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain
| | | | | | - Maria Jesus del Cerro
- Paediatric Cardiology and Grown Up Congenital Heart Disease Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Pablo Lapunzina
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institto de Salud Carlos III, Madrid, Spain
| | - Pilar Escribano-Subias
- Department of Cardiology, Pulmonary Hypertension Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Institto de Salud Carlos III (CIBERCV), Madrid, Spain
- * E-mail: (PES); (IHG)
| |
Collapse
|
44
|
Harbaum L, Rhodes CJ, Otero-Núñez P, Wharton J, Wilkins MR. The application of 'omics' to pulmonary arterial hypertension. Br J Pharmacol 2020; 178:108-120. [PMID: 32201940 DOI: 10.1111/bph.15056] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/03/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
Recent genome-wide analyses of rare and common sequence variations have brought greater clarity to the genetic architecture of pulmonary arterial hypertension and implicated novel genes in disease development. Transcriptional signatures have been reported in whole lung tissue, pulmonary vascular cells and peripheral circulating cells. High-throughput platforms for plasma proteomics and metabolomics have identified novel biomarkers associated with clinical outcomes and provided molecular instruments for risk assessment. There are methodological challenges to integrating these datasets, coupled to statistical power limitations inherent to the study of a rare disease, but the expectation is that this approach will reveal novel druggable targets and biomarkers that will open the way to personalized medicine. Here, we review the current state-of-the-art and future promise of 'omics' in the field of translational medicine in pulmonary arterial hypertension. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Pablo Otero-Núñez
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
45
|
Botros L, Vonk Noordegraaf A, Aman J. Vanishing vessels aboding pulmonary disease: a role for VEGFR2. Eur Respir J 2020; 55:55/4/2000326. [PMID: 32245777 DOI: 10.1183/13993003.00326-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Liza Botros
- Dept of Pulmonary Diseases, Amsterdam University Medical Center, Location VU University Medical Center, Amsterdam, The Netherlands
| | - Anton Vonk Noordegraaf
- Dept of Pulmonary Diseases, Amsterdam University Medical Center, Location VU University Medical Center, Amsterdam, The Netherlands
| | - Jurjan Aman
- Dept of Pulmonary Diseases, Amsterdam University Medical Center, Location VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|