1
|
Abstract
Pulmonary arterial hypertension forms the first and most severe of the 5 categories of pulmonary hypertension. Disease pathogenesis is driven by progressive remodeling of peripheral pulmonary arteries, caused by the excessive proliferation of vascular wall cells, including endothelial cells, smooth muscle cells and fibroblasts, and perivascular inflammation. Compelling evidence from animal models suggests endothelial cell dysfunction is a key initial trigger of pulmonary vascular remodeling, which is characterised by hyperproliferation and early apoptosis followed by enrichment of apoptosis-resistant populations. Dysfunctional pulmonary arterial endothelial cells lose their ability to produce vasodilatory mediators, together leading to augmented pulmonary arterial smooth muscle cell responses, increased pulmonary vascular pressures and right ventricular afterload, and progressive right ventricular hypertrophy and heart failure. It is recognized that a range of abnormal cellular molecular signatures underpin the pathophysiology of pulmonary arterial hypertension and are enhanced by loss-of-function mutations in the BMPR2 gene, the most common genetic cause of pulmonary arterial hypertension and associated with worse disease prognosis. Widespread metabolic abnormalities are observed in the heart, pulmonary vasculature, and systemic tissues, and may underpin heterogeneity in responsivity to treatment. Metabolic abnormalities include hyperglycolytic reprogramming, mitochondrial dysfunction, aberrant polyamine and sphingosine metabolism, reduced insulin sensitivity, and defective iron handling. This review critically discusses published mechanisms linking metabolic abnormalities with dysfunctional BMPR2 (bone morphogenetic protein receptor 2) signaling; hypothesized mechanistic links requiring further validation; and their relevance to pulmonary arterial hypertension pathogenesis and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Iona Cuthbertson
- Department of Medicine, University of Cambridge School of Clinical Medicine, Heart and Lung Research Institute, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Heart and Lung Research Institute, United Kingdom
| | - Paola Caruso
- Department of Medicine, University of Cambridge School of Clinical Medicine, Heart and Lung Research Institute, United Kingdom
| |
Collapse
|
2
|
Wang G, Tao X, Peng L. miR-155-5p regulates hypoxia-induced pulmonary artery smooth muscle cell function by targeting PYGL. Bioengineered 2022; 13:12985-12997. [PMID: 35611851 PMCID: PMC9275946 DOI: 10.1080/21655979.2022.2079304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a cardiovascular disease that has high incidence and causes massive deaths. miR-155-5p/PYGL pathway was revealed to play a crucial role in PAH by weighted gene co-expression network analysis (WGCNA). The potential mechanism of miR-155-5p in regulating hypoxia-induced pulmonary artery smooth muscle cell (PASMC) function was analyzed through in vitro experiments. Hypoxia treatment stimulated the proliferation of PASMCs and increased the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α). At the same time, revealed by qRT-PCR and western blot, the level of miR-155-5p was raised, and the level of PYGL was decreased in hypoxia-induced PASMCs. Through CCK-8 assay, transwell assay and flow cytometry, it was revealed that miR-155-5p inhibitor remarkably inhibited the cell proliferation and migration and decreased the proportion of hypoxia-stimulated PASMCs in S and G2/M phases. Dual-luciferase reporter system was subsequently applied to validate the straight regulation of miR-155-5p on PYGL based on the analysis of online database. Furthermore, siPYGL was revealed to reverse the influence of miR-155-5p inhibitor on hypoxia-induced PASMCs. These outcomes indicate that the increased level of miR-155-5p in hypoxia-stimulated PASMCs could enhance the cell proliferation, cell migration, and cell cycle progression by targeting PYGL directly. This study may supply novel treatment strategies for PAH.Abbreviations: PH, pulmonary hypertension; PAH, pulmonary arterial hypertension; WGCNA, weighted gene co-expression network analysis; PASMCs, pulmonary artery smooth muscle cells; VEGF, vascular endothelial growth factor; HIF-1α, hypoxia-inducible factor-1α; SMCs, smooth muscle cells; DEGs, differentially expressed genes; GEO, Gene Expression Omnibus; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; FBS, fetal bovine serum; OD, optical density; BCA, bicinchoninic acid; PVDF, polyvinylidene fluoride; PBS, phosphate-buffered saline; BP, biological process; MF, molecular function; CC, cell component.
Collapse
Affiliation(s)
- Guowen Wang
- Department of Respiratory Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Xuefang Tao
- Department of Respiratory Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Linlin Peng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Liu J, Ke X, Wang L, Zhang Y, Yang J. Deficiency of cold-inducible RNA-binding protein exacerbated monocrotaline-induced pulmonary artery hypertension through Caveolin1 and CAVIN1. J Cell Mol Med 2021; 25:4732-4743. [PMID: 33755319 PMCID: PMC8107102 DOI: 10.1111/jcmm.16437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022] Open
Abstract
Cold‐inducible RNA‐binding protein (CIRP) was a crucial regulator in multiple diseases. However, its role in pulmonary artery hypertension (PAH) is still unknown. Here, we first established monocrotaline (MCT)‐induced rat PAH model and discovered that CIRP was down‐regulated predominantly in the endothelium of pulmonary artery after MCT injection. We then generated Cirp‐knockout (Cirp‐KO) rats, which manifested severer PAH with exacerbated endothelium damage in response to MCT. Subsequently, Caveolin1 (Cav1) and Cavin1 were identified as downstream targets of CIRP in MCT‐induced PAH, and the decreased expression of these two genes aggravated the injury and apoptosis of pulmonary artery endothelium. Moreover, CIRP deficiency intensified monocrotaline pyrrole (MCTP)‐induced rat pulmonary artery endothelial cells (rPAECs) injuries both in vivo and in vitro, which was counteracted by Cav1 or Cavin1 overexpression. In addition, CIRP regulated the proliferative effect of conditioned media from MCTP‐treated rPAECs on rat pulmonary artery smooth muscle cells, which partially explained the exceedingly thickened pulmonary artery intimal media in Cirp‐KO rats after MCT treatment. These results demonstrated that CIRP acts as a critical protective factor in MCT‐induced rat PAH by directly regulating CAV1 and CAVIN1 expression, which may facilitate the development of new therapeutic targets for the intervention of PAH.
Collapse
Affiliation(s)
- Jingjing Liu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Xianting Ke
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Luxin Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Yang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Ryanto GRT, Ikeda K, Miyagawa K, Tu L, Guignabert C, Humbert M, Fujiyama T, Yanagisawa M, Hirata KI, Emoto N. An endothelial activin A-bone morphogenetic protein receptor type 2 link is overdriven in pulmonary hypertension. Nat Commun 2021; 12:1720. [PMID: 33741934 PMCID: PMC7979873 DOI: 10.1038/s41467-021-21961-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension is a progressive fatal disease that is characterized by pathological pulmonary artery remodeling, in which endothelial cell dysfunction is critically involved. We herein describe a previously unknown role of endothelial angiocrine in pulmonary hypertension. By searching for genes highly expressed in lung microvascular endothelial cells, we identify inhibin-β-A as an angiocrine factor produced by pulmonary capillaries. We find that excess production of inhibin-β-A by endothelial cells impairs the endothelial function in an autocrine manner by functioning as activin-A. Mechanistically, activin-A induces bone morphogenetic protein receptor type 2 internalization and targeting to lysosomes for degradation, resulting in the signal deficiency in endothelial cells. Of note, endothelial cells isolated from the lung of patients with idiopathic pulmonary arterial hypertension show higher inhibin-β-A expression and produce more activin-A compared to endothelial cells isolated from the lung of normal control subjects. When endothelial activin-A-bone morphogenetic protein receptor type 2 link is overdriven in mice, hypoxia-induced pulmonary hypertension was exacerbated, whereas conditional knockout of inhibin-β-A in endothelial cells prevents the progression of pulmonary hypertension. These data collectively indicate a critical role for the dysregulated endothelial activin-A-bone morphogenetic protein receptor type 2 link in the progression of pulmonary hypertension, and thus endothelial inhibin-β-A/activin-A might be a potential pharmacotherapeutic target for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Gusty R T Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Higashinada, Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, Kobe, Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Higashinada, Kobe, Japan.
- Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, Kamigyou, Kyoto, Japan.
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kamigyou, Kyoto, Japan.
| | - Kazuya Miyagawa
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Higashinada, Kobe, Japan
| | - Ly Tu
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Université Paris-Saclay, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Université Paris-Saclay, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Université Paris-Saclay, Université Paris-Sud, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Tomoyuki Fujiyama
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, Kobe, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Higashinada, Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, Kobe, Japan
| |
Collapse
|
5
|
Fazal S, Bisserier M, Hadri L. Molecular and Genetic Profiling for Precision Medicines in Pulmonary Arterial Hypertension. Cells 2021; 10:638. [PMID: 33805595 PMCID: PMC7999465 DOI: 10.3390/cells10030638] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare and chronic lung disease characterized by progressive occlusion of the small pulmonary arteries, which is associated with structural and functional alteration of the smooth muscle cells and endothelial cells within the pulmonary vasculature. Excessive vascular remodeling is, in part, responsible for high pulmonary vascular resistance and the mean pulmonary arterial pressure, increasing the transpulmonary gradient and the right ventricular "pressure overload", which may result in right ventricular (RV) dysfunction and failure. Current technological advances in multi-omics approaches, high-throughput sequencing, and computational methods have provided valuable tools in molecular profiling and led to the identification of numerous genetic variants in PAH patients. In this review, we summarized the pathogenesis, classification, and current treatments of the PAH disease. Additionally, we outlined the latest next-generation sequencing technologies and the consequences of common genetic variants underlying PAH susceptibility and disease progression. Finally, we discuss the importance of molecular genetic testing for precision medicine in PAH and the future of genomic medicines, including gene-editing technologies and gene therapies, as emerging alternative approaches to overcome genetic disorders in PAH.
Collapse
Affiliation(s)
| | | | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (S.F.); (M.B.)
| |
Collapse
|
6
|
Tielemans B, Stoian L, Gijsbers R, Michiels A, Wagenaar A, Farre Marti R, Belge C, Delcroix M, Quarck R. Cytokines trigger disruption of endothelium barrier function and p38 MAP kinase activation in BMPR2-silenced human lung microvascular endothelial cells. Pulm Circ 2019; 9:2045894019883607. [PMID: 31692724 PMCID: PMC6811766 DOI: 10.1177/2045894019883607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022] Open
Abstract
The bone morphogenetic protein receptor II (BMPRII) signaling pathway is impaired
in pulmonary arterial hypertension and mutations in the BMPR2
gene have been observed in both heritable and idiopathic pulmonary arterial
hypertension. However, all BMPR2 mutation carriers do not
develop pulmonary arterial hypertension, and inflammation could trigger the
development of the disease in BMPR2 mutation carriers.
Circulating levels and/or lung tissue expression of cytokines such as tumor
necrosis factor-α or interleukin-18 are elevated in patients with pulmonary
arterial hypertension and could be involved in the pathogenesis of pulmonary
arterial hypertension. We consequently hypothesized that cytokines could trigger
endothelial dysfunction in addition to impaired BMPRII signaling. Our aim was to
determine whether impairment of BMPRII signaling might affect endothelium
barrier function and adhesiveness to monocytes, in response to cytokines.
BMPR2 was silenced in human lung microvascular endothelial
cells (HLMVECs) using lentiviral vectors encoding microRNA-based hairpins.
Effects of tumor necrosis factor-α and interleukin-18 on HLMVEC adhesiveness to
the human monocyte cell line THP-1, adhesion molecule expression, endothelial
barrier function and activation of P38MAPK were investigated in vitro. Stable
BMPR2 silencing in HLMVECs resulted in impaired endothelial
barrier function and constitutive activation of P38MAPK. Adhesiveness of
BMPR2-silenced HLMVECs to THP-1 cells was enhanced by tumor
necrosis factor-α and interleukin-18 through ICAM-1 adhesion molecule.
Interestingly, tumor necrosis factor-α induced activation of P38MAPK and
disrupted endothelial barrier function in BMPR2-silenced
HLMVECs. Altogether, our findings showed that stable BMPR2
silencing resulted in impaired endothelial barrier function and activation of
P38MAPK in HLMVECs. In BMPR2-silenced HLMVECs, cytokines
enhanced adhesiveness capacities, activation of P38MAPK and impaired endothelial
barrier function suggesting that cytokines could trigger the development of
pulmonary arterial hypertension in a context of impaired BMPRII signaling
pathway.
Collapse
Affiliation(s)
- Birger Tielemans
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Leanda Stoian
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rik Gijsbers
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium.,Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Annelies Michiels
- Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium.,Leuven Viral Vector Core, KU Leuven - University of Leuven, Leuven, Belgium
| | - Allard Wagenaar
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Ricard Farre Marti
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Catharina Belge
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Marion Delcroix
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rozenn Quarck
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Tielemans B, Delcroix M, Belge C, Quarck R. TGFβ and BMPRII signalling pathways in the pathogenesis of pulmonary arterial hypertension. Drug Discov Today 2019; 24:703-716. [DOI: 10.1016/j.drudis.2018.12.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 12/04/2018] [Indexed: 01/23/2023]
|
8
|
Barman SA, Li X, Haigh S, Kondrikov D, Mahboubi K, Bordan Z, Stepp DW, Zhou J, Wang Y, Weintraub DS, Traber P, Snider W, Jonigk D, Sullivan J, Crislip GR, Butcher JT, Thompson J, Su Y, Chen F, Fulton DJR. Galectin-3 is expressed in vascular smooth muscle cells and promotes pulmonary hypertension through changes in proliferation, apoptosis, and fibrosis. Am J Physiol Lung Cell Mol Physiol 2019; 316:L784-L797. [PMID: 30724100 DOI: 10.1152/ajplung.00186.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A defining characteristic of pulmonary hypertension (PH) is the extensive remodeling of pulmonary arteries (PAs), which results in progressive increases in vascular resistance and stiffness and eventual failure of the right ventricle. There is no cure for PH and identification of novel molecular mechanisms that underlie increased proliferation, reduced apoptosis, and excessive extracellular matrix production in pulmonary artery smooth muscle cells (PASMCs) is a vital objective. Galectin-3 (Gal-3) is a chimeric lectin and potent driver of many aspects of fibrosis, but its role in regulating PASMC behavior in PH remains poorly understood. Herein, we evaluated the importance of increased Gal-3 expression and signaling on PA vascular remodeling and cardiopulmonary function in experimental models of PH. Gal-3 expression was quantified by qRT-PCR, immunoblotting, and immunofluorescence imaging, and its functional role was assessed by specific Gal-3 inhibitors and CRISPR/Cas9-mediated knockout of Gal-3 in the rat. In rat models of PH, we observed increased Gal-3 expression in PASMCs, which stimulated migration and resistance to apoptosis, whereas silencing or genetic deletion reduced cellular migration and PA fibrosis and increased apoptosis. Gal-3 inhibitors attenuated and reversed PA remodeling and fibrosis, as well as hemodynamic indices in monocrotaline (MCT)-treated rats in vivo. These results were supported by genetic deletion of Gal-3 in both MCT and Sugen Hypoxia rat models. In conclusion, our results suggest that elevated Gal-3 levels contribute to inappropriate PA remodeling in PH by enhancing multiple profibrotic mechanisms. Therapeutic strategies targeting Gal-3 may be of benefit in the treatment of PH.
Collapse
Affiliation(s)
- Scott A Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Xueyi Li
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Stephen Haigh
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Dmitry Kondrikov
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Keyvan Mahboubi
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - David W Stepp
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Yusi Wang
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Daniel S Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | | | - William Snider
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Danny Jonigk
- Department of Pathology, Hannover Medical School , Hannover , Germany
| | - Jennifer Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - G Ryan Crislip
- Department of Physiology, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Joshua T Butcher
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Jennifer Thompson
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Feng Chen
- Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia.,Department of Forensic Medicine, Nanjing Medical University , Nanjing, Jiangsu , China
| | - David J R Fulton
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University , Augusta, Georgia.,Vascular Biology Center, Medical College of Georgia at Augusta University , Augusta, Georgia
| |
Collapse
|
9
|
Morrell NW, Aldred MA, Chung WK, Elliott CG, Nichols WC, Soubrier F, Trembath RC, Loyd JE. Genetics and genomics of pulmonary arterial hypertension. Eur Respir J 2019; 53:13993003.01899-2018. [PMID: 30545973 PMCID: PMC6351337 DOI: 10.1183/13993003.01899-2018] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 12/17/2022]
Abstract
Since 2000 there have been major advances in our understanding of the genetic and genomics of pulmonary arterial hypertension (PAH), although there remains much to discover. Based on existing knowledge, around 25-30% of patients diagnosed with idiopathic PAH have an underlying Mendelian genetic cause for their condition and should be classified as heritable PAH (HPAH). Here, we summarise the known genetic and genomic drivers of PAH, the insights these provide into pathobiology, and the opportunities afforded for development of novel therapeutic approaches. In addition, factors determining the incomplete penetrance observed in HPAH are discussed. The currently available approaches to genetic testing and counselling, and the impact of a genetic diagnosis on clinical management of the patient with PAH, are presented. Advances in DNA sequencing technology are rapidly expanding our ability to undertake genomic studies at scale in large cohorts. In the future, such studies will provide a more complete picture of the genetic contribution to PAH and, potentially, a molecular classification of this disease.
Collapse
Affiliation(s)
- Nicholas W Morrell
- University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, UK
| | | | - Wendy K Chung
- Columbia University Medical Center, New York, NY, USA
| | - C Gregory Elliott
- Intermountain Medical Center and University of Utah, Salt Lake City, UT, USA
| | | | | | - Richard C Trembath
- Division of Genetics and Molecular Medicine, School of Basic and Medical Biosciences, King's College London, London, UK
| | - James E Loyd
- Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
10
|
Predescu D, Qin S, Patel M, Bardita C, Bhalli R, Predescu S. Epsin15 Homology Domains: Role in the Pathogenesis of Pulmonary Arterial Hypertension. Front Physiol 2018; 9:1393. [PMID: 30333761 PMCID: PMC6176378 DOI: 10.3389/fphys.2018.01393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/13/2018] [Indexed: 12/24/2022] Open
Abstract
Intersectin-1s (ITSN) deficiency and expression of a biologically active ITSN fragment, result of granzyme B cleavage under inflammatory conditions associated with pulmonary arterial hypertension (PAH), are characteristics of lung tissue of human and animal models of PAH. Recently, we have shown that this ITSN fragment comprising two Epsin15 homology domains (EHITSN) triggers endothelial cell (EC) proliferation and the plexiform arteriopathy in PAH. Limited evidence also indicates that the EH domains of endocytic proteins such as ITSN, upregulate compensatory endocytic pathways in cells with impaired vesicular trafficking. Thus, we sought to investigate whether the EHITSN may be involved in this compensatory mechanism for improving the EC endocytic dysfunction induced by ITSN deficiency and possibly contribute to PAH pathogenesis. We used stably-transfected human pulmonary artery ECs expressing the Myc-EHITSN (ECEH-ITSN) and ITSN knockout heterozygous mice (K0ITSN+/-) transduced with the Myc-EHITSN, in conjunction with functional assays: the biotin assay for caveolae internalization and 8 nm gold (Au)- and dinitrophenylated (DNP)-albumin perfusion of murine lung microvasculature. Pulmonary artery ECs of PAH patients (ECPAH), ITSN knockdown ECs (ECKD-ITSN), the monocrotaline (MCT)-induced mouse and rat models of PAH, as well as untreated animals, served as controls. ELISA via streptavidin-HRP or anti-DNP antibody (Ab), applied on ECs and lung lysates indicated greater than 30% increase in biotin internalization in ECEH-ITSN compared to ECCtrl. Despite their endocytic deficiency, ECPAH internalized biotin similar to ECCtrl which is twofold higher compared to ECKD-ITSN. Moreover, the lung microvascular bed of Myc-EHITSN-transduced mice and MCT-treated animals showed greater than twofold increase in DNP-BSA transendothelial transport, all compared to untreated controls. Electron microscopy (EM) revealed the increased occurrence of non-conventional endocytic/transcytotic structures (i.e., caveolae clusters, tubulo-vesicular and enlarged endocytic structures, membranous rings), usually underrepresented. Most of these structures were labeled by Au-BSA, consistent with their involvement in the transendothelial transport. Furthermore, ITSN deficiency and EHITSN expression alter the subcellular localization of the EH-binding protein 1 (EHBP1) and cortical actin organization, altogether supporting the increase occurrence/trafficking of the alternative endocytic structures. Thus, the EHITSN by shifting the physiological vesicular (caveolae) transport toward the alternative endocytic pathways is a significant contributor to the dysfunctional molecular phenotype of ECPAH.
Collapse
Affiliation(s)
- Dan Predescu
- Division of Pulmonary Medicine, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| | - Shanshan Qin
- Division of Pulmonary Medicine, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| | - Monal Patel
- Division of Pulmonary Medicine, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| | - Cristina Bardita
- Division of Pulmonary Medicine, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| | - Rabia Bhalli
- Division of Pulmonary Medicine, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| | - Sanda Predescu
- Division of Pulmonary Medicine, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| |
Collapse
|
11
|
Rätsep MT, Moore SD, Jafri S, Mitchell M, Brady HJM, Mandelboim O, Southwood M, Morrell NW, Colucci F, Ormiston ML. Spontaneous pulmonary hypertension in genetic mouse models of natural killer cell deficiency. Am J Physiol Lung Cell Mol Physiol 2018; 315:L977-L990. [PMID: 30234375 PMCID: PMC6337009 DOI: 10.1152/ajplung.00477.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are cytotoxic innate lymphoid cells with an established role in the regulation of vascular structure in pregnancy and cancer. Impaired NK cell function has been identified in patients with pulmonary arterial hypertension (PAH), a disease of obstructive vascular remodeling in the lungs, as well as in multiple rodent models of disease. However, the precise contribution of NK cell impairment to the initiation and progression of PAH remains unknown. Here, we report the development of spontaneous pulmonary hypertension in two independent genetic models of NK cell dysfunction, including Nfil3−/− mice, which are deficient in NK cells due to the absence of the NFIL3 transcription factor, and Ncr1-Gfp mice, which lack the NK activating receptor NKp46. Mouse models of NK insufficiency exhibited increased right ventricular systolic pressure and muscularization of the pulmonary arteries in the absence of elevated left ventricular end-diastolic pressure, indicating that the development of pulmonary hypertension was not secondary to left heart dysfunction. In cases of severe NK cell impairment or loss, a subset of mice failed to develop pulmonary hypertension and instead exhibited reduced systemic blood pressure, demonstrating an extension of vascular abnormalities beyond the pulmonary circulation into the systemic vasculature. In both mouse models, the development of PAH was linked to elevated interleukin-23 production, whereas systemic hypotension in Ncr1-Gfp mice was accompanied by a loss of angiopoietin-2. Together, these results support an important role for NK cells in the regulation of pulmonary and systemic vascular function and the pathogenesis of PAH.
Collapse
Affiliation(s)
- Matthew T Rätsep
- Departments of Biomedical and Molecular Sciences, Medicine, and Surgery, Queen's University Kingston , Ontario , Canada
| | - Stephen D Moore
- Department of Medicine, University of Cambridge, Cambridge , United Kingdom
| | - Salema Jafri
- Department of Medicine, University of Cambridge, Cambridge , United Kingdom
| | - Melissa Mitchell
- Departments of Biomedical and Molecular Sciences, Medicine, and Surgery, Queen's University Kingston , Ontario , Canada
| | | | | | - Mark Southwood
- Department of Medicine, University of Cambridge, Cambridge , United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge , United Kingdom
| | - Francesco Colucci
- Department of Obstetrics and Gynecology, University of Cambridge, Cambridge , United Kingdom
| | - Mark L Ormiston
- Departments of Biomedical and Molecular Sciences, Medicine, and Surgery, Queen's University Kingston , Ontario , Canada
| |
Collapse
|
12
|
Zhou C, Francis CM, Xu N, Stevens T. The role of endothelial leak in pulmonary hypertension (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018798569. [PMID: 30124139 PMCID: PMC6134503 DOI: 10.1177/2045894018798569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The canonical transient receptor potential 4 (TRPC4) protein contributes to the molecular make-up of endothelial store-operated calcium entry channels. Store-operated calcium entry is a prominent mode of calcium influx in endothelium. Store-operated calcium entry channels are activated by inflammatory mediators and growth factors, and in endothelium, this process induces inter-endothelial cell gaps that increase permeability. Pulmonary endothelium within extra-alveolar segments, including pulmonary arteries, is especially sensitive to the activation of store-operated calcium entry. Pulmonary arterial hypertension (PAH) is characterized by endothelial cell dysfunction in arteries. As one of the topics for the 2017 Grover Conference Series, we examined whether an endothelial cell permeability defect accompanies PAH and, if so, whether TRPC4 contributes to this defect. Through a series of studies conducted over the past five years, we find endothelial cell barrier dysfunction occurs early in the progression of experimental PAH. Endothelium within the arterial segment, and perhaps in other vascular segments, is highly susceptible to disruption secondary to both activation of store-operated calcium entry channels and high flow. This phenomenon partly depends upon TRPC4 channels. We discuss whether endothelial cell hyperpermeability is relevant to human disease, and more specifically, whether it is relevant to all groups of pulmonary hypertension.
Collapse
Affiliation(s)
- Chun Zhou
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - C Michael Francis
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Ningyong Xu
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Troy Stevens
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,3 Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
13
|
Abdellatif KR, Bakr RB. New advances in synthesis and clinical aspects of pyrazolo[3,4-d]pyrimidine scaffolds. Bioorg Chem 2018; 78:341-357. [DOI: 10.1016/j.bioorg.2018.03.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/17/2018] [Accepted: 03/31/2018] [Indexed: 01/20/2023]
|
14
|
Wauchope OR, Mitchener MM, Beavers WN, Galligan JJ, Camarillo JM, Sanders WD, Kingsley PJ, Shim HN, Blackwell T, Luong T, deCaestecker M, Fessel JP, Marnett LJ. Oxidative stress increases M1dG, a major peroxidation-derived DNA adduct, in mitochondrial DNA. Nucleic Acids Res 2018; 46:3458-3467. [PMID: 29438559 PMCID: PMC5909422 DOI: 10.1093/nar/gky089] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/29/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are formed in mitochondria during electron transport and energy generation. Elevated levels of ROS lead to increased amounts of mitochondrial DNA (mtDNA) damage. We report that levels of M1dG, a major endogenous peroxidation-derived DNA adduct, are 50-100-fold higher in mtDNA than in nuclear DNA in several different human cell lines. Treatment of cells with agents that either increase or decrease mitochondrial superoxide levels leads to increased or decreased levels of M1dG in mtDNA, respectively. Sequence analysis of adducted mtDNA suggests that M1dG residues are randomly distributed throughout the mitochondrial genome. Basal levels of M1dG in mtDNA from pulmonary microvascular endothelial cells (PMVECs) from transgenic bone morphogenetic protein receptor 2 mutant mice (BMPR2R899X) (four adducts per 106 dG) are twice as high as adduct levels in wild-type cells. A similar increase was observed in mtDNA from heterozygous null (BMPR2+/-) compared to wild-type PMVECs. Pulmonary arterial hypertension is observed in the presence of BMPR2 signaling disruptions, which are also associated with mitochondrial dysfunction and oxidant injury to endothelial tissue. Persistence of M1dG adducts in mtDNA could have implications for mutagenesis and mitochondrial gene expression, thereby contributing to the role of mitochondrial dysfunction in diseases.
Collapse
Affiliation(s)
- Orrette R Wauchope
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Michelle M Mitchener
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - William N Beavers
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James J Galligan
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jeannie M Camarillo
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - William D Sanders
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Philip J Kingsley
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ha-Na Shim
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Thomas Blackwell
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Thong Luong
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mark deCaestecker
- Departments of Cell and Developmental Biology, Surgery and Medicine, USA
| | - Joshua P Fessel
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lawrence J Marnett
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
15
|
Frump A, Prewitt A, de Caestecker MP. BMPR2 mutations and endothelial dysfunction in pulmonary arterial hypertension (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018765840. [PMID: 29521190 PMCID: PMC5912278 DOI: 10.1177/2045894018765840] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/26/2018] [Indexed: 12/22/2022] Open
Abstract
Despite the discovery more than 15 years ago that patients with hereditary pulmonary arterial hypertension (HPAH) inherit BMP type 2 receptor ( BMPR2) mutations, it is still unclear how these mutations cause disease. In part, this is attributable to the rarity of HPAH and difficulty obtaining tissue samples from patients with early disease. However, in addition, limitations to the approaches used to study the effects of BMPR2 mutations on the pulmonary vasculature have restricted our ability to determine how individual mutations give rise to progressive pulmonary vascular pathology in HPAH. The importance of understanding the mechanisms by which BMPR2 mutations cause disease in patients with HPAH is underscored by evidence that there is reduced BMPR2 expression in patients with other, more common, non-hereditary form of PAH, and that restoration of BMPR2 expression reverses established disease in experimental models of pulmonary hypertension. In this paper, we focus on the effects on endothelial function. We discuss some of the controversies and challenges that have faced investigators exploring the role of BMPR2 mutations in HPAH, focusing specifically on the effects different BMPR2 mutation have on endothelial function, and whether there are qualitative differences between different BMPR2 mutations. We discuss evidence that BMPR2 signaling regulates a number of responses that may account for endothelial abnormalities in HPAH and summarize limitations of the models that are used to study these effects. Finally, we discuss evidence that BMPR2-dependent effects on endothelial metabolism provides a unifying explanation for the many of the BMPR2 mutation-dependent effects that have been described in patients with HPAH.
Collapse
Affiliation(s)
- Andrea Frump
- Division
of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University
School of Medicine, Indianapolis, IN,
USA
| | | | - Mark P. de Caestecker
- Division
of Nephrology and Hypertension, Department of Medicine, Vanderbilt University
Medical center, Nashville, TN, USA
| |
Collapse
|
16
|
Gamou S, Kataoka M, Aimi Y, Chiba T, Momose Y, Isobe S, Hirayama T, Yoshino H, Fukuda K, Satoh T. Genetics in pulmonary arterial hypertension in a large homogeneous Japanese population. Clin Genet 2018; 94:70-80. [DOI: 10.1111/cge.13154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Affiliation(s)
- S. Gamou
- Kyorin University Center for Comprehensive Regional Collaboration; Tokyo Japan
| | - M. Kataoka
- Department of Cardiology; Keio University School of Medicine; Tokyo Japan
| | - Y. Aimi
- Division of Cardiology, Second Department of Internal Medicine; Kyorin University School of Medicine; Tokyo Japan
| | - T. Chiba
- Department of Pathology; Kyorin University School of Medicine; Tokyo Japan
| | - Y. Momose
- Division of Cardiology, Second Department of Internal Medicine; Kyorin University School of Medicine; Tokyo Japan
| | - S. Isobe
- Department of Cardiology; Keio University School of Medicine; Tokyo Japan
| | - T. Hirayama
- Department of Cardiology; Keio University School of Medicine; Tokyo Japan
- Division of Cardiology, Second Department of Internal Medicine; Kyorin University School of Medicine; Tokyo Japan
| | - H. Yoshino
- Division of Cardiology, Second Department of Internal Medicine; Kyorin University School of Medicine; Tokyo Japan
| | - K. Fukuda
- Department of Cardiology; Keio University School of Medicine; Tokyo Japan
| | - T. Satoh
- Division of Cardiology, Second Department of Internal Medicine; Kyorin University School of Medicine; Tokyo Japan
| |
Collapse
|
17
|
Krispin S, Stratman AN, Melick CH, Stan RV, Malinverno M, Gleklen J, Castranova D, Dejana E, Weinstein BM. Growth Differentiation Factor 6 Promotes Vascular Stability by Restraining Vascular Endothelial Growth Factor Signaling. Arterioscler Thromb Vasc Biol 2017; 38:353-362. [PMID: 29284606 DOI: 10.1161/atvbaha.117.309571] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/05/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The assembly of a functional vascular system requires a coordinated and dynamic transition from activation to maturation. High vascular endothelial growth factor activity promotes activation, including junction destabilization and cell motility. Maturation involves junctional stabilization and formation of a functional endothelial barrier. The identity and mechanism of action of prostabilization signals are still mostly unknown. Bone morphogenetic protein receptors and their ligands have important functions during embryonic vessel assembly and maturation. Previous work has suggested a role for growth differentiation factor 6 (GDF6; bone morphogenetic protein 13) in vascular integrity although GDF6's mechanism of action was not clear. Therefore, we sought to further explore the requirement for GDF6 in vascular stabilization. APPROACH AND RESULTS We investigated the role of GDF6 in promoting endothelial vascular integrity in vivo in zebrafish and in cultured human umbilical vein endothelial cells in vitro. We report that GDF6 promotes vascular integrity by counteracting vascular endothelial growth factor activity. GDF6-deficient endothelium has increased vascular endothelial growth factor signaling, increased vascular endothelial-cadherin Y658 phosphorylation, vascular endothelial-cadherin delocalization from cell-cell interfaces, and weakened endothelial cell adherence junctions that become prone to vascular leak. CONCLUSIONS Our results suggest that GDF6 promotes vascular stabilization by restraining vascular endothelial growth factor signaling. Understanding how GDF6 affects vascular integrity may help to provide insights into hemorrhage and associated vascular pathologies in humans.
Collapse
Affiliation(s)
- Shlomo Krispin
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Amber N Stratman
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Chase H Melick
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Radu V Stan
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Matteo Malinverno
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Jamie Gleklen
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Daniel Castranova
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Elisabetta Dejana
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Brant M Weinstein
- From the Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD (S.K., A.N.S., C.H.M., J.G., D.C., B.M.W.); Departments of Biochemistry and Cell Biology and of Pathology, Geisel School of Medicine at Dartmouth College, Lebanon, NH (R.V.S.); Vascular Biology Program, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy (M.M., E.D.); and Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.).
| |
Collapse
|
18
|
Copeland CA, Han B, Tiwari A, Austin ED, Loyd JE, West JD, Kenworthy AK. A disease-associated frameshift mutation in caveolin-1 disrupts caveolae formation and function through introduction of a de novo ER retention signal. Mol Biol Cell 2017; 28:3095-3111. [PMID: 28904206 PMCID: PMC5662265 DOI: 10.1091/mbc.e17-06-0421] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
Heterozygous mutations in caveolin-1 (CAV1) have been linked to pulmonary arterial hypertension (PAH), but their impact on caveolae is unclear. We show that a PAH-associated frameshift mutation introduces an endoplasmic reticulum retention signal in CAV1 that partially disrupts caveolae assembly and interferes with their ability to serve as membrane buffers. Caveolin-1 (CAV1) is an essential component of caveolae and is implicated in numerous physiological processes. Recent studies have identified heterozygous mutations in the CAV1 gene in patients with pulmonary arterial hypertension (PAH), but the mechanisms by which these mutations impact caveolae assembly and contribute to disease remain unclear. To address this question, we examined the consequences of a familial PAH-associated frameshift mutation in CAV1, P158PfsX22, on caveolae assembly and function. We show that C-terminus of the CAV1 P158 protein contains a functional ER-retention signal that inhibits ER exit and caveolae formation and accelerates CAV1 turnover in Cav1–/– MEFs. Moreover, when coexpressed with wild-type (WT) CAV1 in Cav1–/– MEFs, CAV1-P158 functions as a dominant negative by partially disrupting WT CAV1 trafficking. In patient skin fibroblasts, CAV1 and caveolar accessory protein levels are reduced, fewer caveolae are observed, and CAV1 complexes exhibit biochemical abnormalities. Patient fibroblasts also exhibit decreased resistance to a hypo-osmotic challenge, suggesting the function of caveolae as membrane reservoir is compromised. We conclude that the P158PfsX22 frameshift introduces a gain of function that gives rise to a dominant negative form of CAV1, defining a new mechanism by which disease-associated mutations in CAV1 impair caveolae assembly.
Collapse
Affiliation(s)
- Courtney A. Copeland
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Bing Han
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Ajit Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Eric D. Austin
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232
| | - James E. Loyd
- Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - James D. West
- Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Anne K. Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
- Epithelial Biology Program, Vanderbilt University School of Medicine, Nashville, TN 37232
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
19
|
Yang K, Wang J, Lu W. Bone morphogenetic protein signalling in pulmonary hypertension: advances and therapeutic implications. Exp Physiol 2017; 102:1083-1089. [PMID: 28449240 DOI: 10.1113/ep086041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/24/2017] [Indexed: 01/07/2023]
Abstract
NEW FINDINGS What is the topic of this review? This review covers recent evidence highlighting the crucial pathophysiological roles and molecular mechanisms of the bone morphogenetic protein (BMP) signalling pathway during the progression of pulmonary hypertension (PH) and discusses targeting of BMP signalling as a new treatment option against PH. What advances does it highlight? A series of breakthrough findings have greatly enriched our understanding about the mechanism of action of BMP signalling in PH and proved the feasibility of BMP targeting strategies in experimental PH models. This review collects these ideas and discusses the frontiers of BMP signalling-targeted PH therapy at different steps of the signal transduction. The bone morphogenetic protein (BMP)-mediated signalling pathway plays crucial roles in the development and progression of pulmonary hypertension (PH). Typical BMP signalling involves BMP ligands, specific transmembrane serine/threonine kinase receptors, cellular responsive kinases and secreted antagonists. As more and more studies have been conducted, the specific protective or pathogenic roles of these molecules within all these subgroups of BMP signalling have been continuously uncovered. Based on this evidence, specific strategies have been designed by targeting these factors as a new treatment approach to PH. In this review, we have collected recent advances in the exciting findings that link BMP signalling with the pathogenesis of PH and we discuss the potential future frontiers in therapeutic design.
Collapse
Affiliation(s)
- Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Pousada G, Lupo V, Cástro-Sánchez S, Álvarez-Satta M, Sánchez-Monteagudo A, Baloira A, Espinós C, Valverde D. Molecular and functional characterization of the BMPR2 gene in Pulmonary Arterial Hypertension. Sci Rep 2017; 7:1923. [PMID: 28507310 PMCID: PMC5432510 DOI: 10.1038/s41598-017-02074-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/05/2017] [Indexed: 11/09/2022] Open
Abstract
Pulmonary arterial hypertension is a progressive disease that causes the obstruction of precapillary pulmonary arteries and a sustained increase in pulmonary vascular resistance. The aim was to analyze functionally the variants found in the BMPR2 gene and to establish a genotype-phenotype correlation. mRNA expression studies were performed using pSPL3 vector, studies of subcellular localization were performed using pEGFP-N1 vector and luciferase assays were performed using pGL3-Basic vector. We have identified 30 variants in the BMPR2 gene in 27 of 55 patients. In 16 patients we detected pathogenic mutations. Minigene assays revealed that 6 variants (synonymous, missense) result in splicing defect. By immunofluorescence assay, we observed that 4 mutations affect the protein localization. Finally, 4 mutations located in the 5'UTR region showed a decreased transcriptional activity in luciferase assays. Genotype-phenotype correlation, revealed that patients with pathogenic mutations have a more severe phenotype (sPaP p = 0.042, 6MWT p = 0.041), a lower age at diagnosis (p = 0.040) and seemed to have worse response to phosphodiesterase-5-inhibitors (p = 0.010). Our study confirms that in vitro expression analysis is a suitable approach in order to investigate the phenotypic consequences of the nucleotide variants, especially in cases where the involved genes have a pattern of expression in tissues of difficult access.
Collapse
Affiliation(s)
- Guillermo Pousada
- Dep. Biochemistry, Genetics and Immunology. Faculty of Biology, University of Vigo, As Lagoas Marcosende S/N, 36310, Vigo, Spain.,Grupo de Investigación Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Vincenzo Lupo
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012, Valencia, Spain
| | - Sheila Cástro-Sánchez
- Dep. Biochemistry, Genetics and Immunology. Faculty of Biology, University of Vigo, As Lagoas Marcosende S/N, 36310, Vigo, Spain.,Grupo de Investigación Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - María Álvarez-Satta
- Dep. Biochemistry, Genetics and Immunology. Faculty of Biology, University of Vigo, As Lagoas Marcosende S/N, 36310, Vigo, Spain.,Grupo de Investigación Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Ana Sánchez-Monteagudo
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012, Valencia, Spain
| | - Adolfo Baloira
- Neumology Service, Complexo Hospitalario Universitario de Pontevedra, 36071, Pontevedra, Spain
| | - Carmen Espinós
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012, Valencia, Spain
| | - Diana Valverde
- Dep. Biochemistry, Genetics and Immunology. Faculty of Biology, University of Vigo, As Lagoas Marcosende S/N, 36310, Vigo, Spain. .,Grupo de Investigación Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
21
|
Frump AL, Datta A, Ghose S, West J, de Caestecker MP. Genotype-phenotype effects of Bmpr2 mutations on disease severity in mouse models of pulmonary hypertension. Pulm Circ 2017; 6:597-607. [PMID: 28090303 DOI: 10.1086/688930] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
More than 350 mutations in the type-2 BMP (bone morphogenetic protein) receptor, BMPR2, have been identified in patients with heritable pulmonary arterial hypertension (HPAH). However, only 30% of BMPR2 mutation carriers develop PAH, and we cannot predict which of these carriers will develop clinical disease. One possibility is that the nature of the BMPR2 mutation affects disease severity. This hypothesis has been difficult to test clinically, given the rarity of HPAH and the complexity of the confounding genetic and environmental risk factors. To test this hypothesis, therefore, we evaluated the susceptibility to experimental pulmonary hypertension (PH) of mice carrying different HPAH-associated Bmpr2 mutations on otherwise identical genetic backgrounds. Mice with Bmpr2ΔEx4-5 mutations (Bmpr2+/-), in which the mutant protein is not expressed, develop less severe PH in response to hypoxia or hypoxia with vascular endothelial growth factor receptor inhibition than mice with an extracellular-domain Bmpr2ΔEx2 mutation (Bmpr2ΔEx2/+), in which the mutant protein is expressed. This was associated with a marked decrease in stabilizing phosphorylation of threonine 495 endothelial nitric oxide synthase (pThr495 eNOS) in Bmpr2ΔEx2/+ compared to wild-type and Bmpr2+/- mouse lungs. These findings provide the first experimental evidence that BMPR2 mutation types influence the severity of HPAH and suggest that patients with BMPR2 mutations who express mutant BMPR2 proteins by escaping non-sense-mediated messenger RNA decay (NMD- mutations) will develop more severe disease than HPAH patients with NMD+ mutations who do not express BMPR2 mutant proteins. Since decreased levels of pThr495 eNOS are associated with increased eNOS uncoupling, our data also suggest that this effect may result from defects in eNOS function.
Collapse
Affiliation(s)
- Andrea L Frump
- Department of Cell and Developmental Biology, Vanderbilt University, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Arunima Datta
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sampa Ghose
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James West
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark P de Caestecker
- Department of Cell and Developmental Biology, Vanderbilt University, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
22
|
Targeting Vascular Remodeling to Treat Pulmonary Arterial Hypertension. Trends Mol Med 2017; 23:31-45. [DOI: 10.1016/j.molmed.2016.11.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022]
|
23
|
Barman SA, Fulton D. Adventitial Fibroblast Nox4 Expression and ROS Signaling in Pulmonary Arterial Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:1-11. [PMID: 29047077 DOI: 10.1007/978-3-319-63245-2_1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease arising from remodeling and narrowing of pulmonary arteries (PA) resulting in high pulmonary arterial blood pressure and ultimately right ventricular failure. Elevated production of reactive oxygen species (ROS) by NADPH oxidase 4 (Nox4), a constitutively active enzyme, has been associated with oxygen sensing, vasomotor control, cellular proliferation, differentiation, migration, apoptosis, senescence, fibrosis, and angiogenesis. Further, elevated expression of Nox4 has been reported in a number of cardiovascular diseases, including atherosclerosis, hypertension, cardiac failure, ischemic stroke, and PAH. However, the cellular location of Nox4 and its contribution to aberrant vascular remodeling in PAH remains poorly understood. The goal of this review is to summarize the recent literature on the enzymatic regulation of Nox4 in the production of ROS in PAH. In the vascular wall, Nox4 is present in fibroblasts, a primary cell of the adventitia, and matches the adventitial location of ROS production in PAH. Further, in adventitial fibroblasts, Nox4 overexpression stimulates migration and proliferation as well as matrix gene expression. Collectively, reports indicate that Nox4 contributes to altered fibroblast behavior, ROS production leading to hypertensive vascular remodeling and the development of PAH. Finally, we address the functional significance of Nox4 in fibroblasts, and also suggest an "outside in" (adventitial) process of vascular remodeling that is mediated by Nox4, which although has physiological roles in the intimal layer (i.e., endothelium), may also have pathologic importance in the adventitial layer of the vascular wall through signaling in fibroblasts.
Collapse
Affiliation(s)
- Scott A Barman
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, 30912, USA.
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA
| |
Collapse
|
24
|
Ma L, Chung WK. The role of genetics in pulmonary arterial hypertension. J Pathol 2016; 241:273-280. [PMID: 27770446 DOI: 10.1002/path.4833] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 12/14/2022]
Abstract
Group 1 pulmonary hypertension or pulmonary arterial hypertension (PAH) is a rare disease characterized by proliferation and occlusion of small pulmonary arterioles, leading to progressive elevation of pulmonary artery pressure and pulmonary vascular resistance, and right ventricular failure. Historically, it has been associated with a high mortality rate, although, over the last decade, treatment has improved survival. PAH includes idiopathic PAH (IPAH), heritable PAH (HPAH), and PAH associated with certain medical conditions. The aetiology of PAH is heterogeneous, and genetics play an important role in some cases. Mutations in BMPR2, encoding bone morphogenetic protein receptor 2, a member of the transforming growth factor-β superfamily of receptors, have been identified in 70% of cases of HPAH, and in 10-40% of cases of IPAH. Other genetic causes of PAH include mutations in the genes encoding activin receptor-like type 1, endoglin, SMAD9, caveolin 1, and potassium two-pore-domain channel subfamily K member 3. Mutations in the gene encoding T-box 4 have been identified in 10-30% of paediatric PAH patients, but rarely in adults with PAH. PAH in children is much more heterogeneous than in adults, and can be associated with several genetic syndromes, congenital heart disease, pulmonary disease, and vascular disease. In addition to rare mutations as a monogenic cause of HPAH, common variants in the gene encoding cerebellin 2 increase the risk of PAH by approximately two-fold. A PAH panel of genes is available for clinical testing, and should be considered for use in clinical management, especially for patients with a family history of PAH. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lijiang Ma
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| |
Collapse
|
25
|
BMPRII influences the response of pulmonary microvascular endothelial cells to inflammatory mediators. Pflugers Arch 2016; 468:1969-1983. [PMID: 27816994 DOI: 10.1007/s00424-016-1899-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/13/2016] [Accepted: 10/24/2016] [Indexed: 10/20/2022]
Abstract
Mutations in the bone morphogenetic protein receptor (BMPR2) gene have been observed in 70 % of patients with heritable pulmonary arterial hypertension (HPAH) and in 11-40 % with idiopathic PAH (IPAH). However, carriers of a BMPR2 mutation have only 20 % risk of developing PAH. Since inflammatory mediators are increased and predict survival in PAH, they could act as a second hit inducing the development of pulmonary hypertension in BMPR2 mutation carriers. Our specific aim was to determine whether inflammatory mediators could contribute to pulmonary vascular cell dysfunction in PAH patients with and without a BMPR2 mutation. Pulmonary microvascular endothelial cells (PMEC) and arterial smooth muscle cells (PASMC) were isolated from lung parenchyma of transplanted PAH patients, carriers of a BMPR2 mutation or not, and from lobectomy patients or lung donors. The effects of CRP and TNFα on mitogenic activity, adhesiveness capacity, and expression of adhesion molecules were investigated in PMECs and PASMCs. PMECs from BMPR2 mutation carriers induced an increase in PASMC mitogenic activity; moreover, endothelin-1 secretion by PMECs from carriers was higher than by PMECs from non-carriers. Recruitment of monocytes by PMECs isolated from carriers was higher compared to PMECs from non-carriers and from controls, with an elevated ICAM-1 expression. CRP increased adhesion of monocytes to PMECs in carriers and non-carriers, and TNFα only in carriers. PMEC from BMPR2 mutation carriers have enhanced adhesiveness for monocytes in response to inflammatory mediators, suggesting that BMPR2 mutation could generate susceptibility to an inflammatory insult in PAH.
Collapse
|
26
|
Hickey PM, Thompson AA, Charalampopoulos A, Elliot CA, Hamilton N, Kiely DG, Lawrie A, Sabroe I, Condliffe R. Bosutinib therapy resulting in severe deterioration of pre-existing pulmonary arterial hypertension. Eur Respir J 2016; 48:1514-1516. [DOI: 10.1183/13993003.01004-2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/12/2016] [Indexed: 11/05/2022]
|
27
|
Zhou C, Townsley MI, Alexeyev M, Voelkel NF, Stevens T. Endothelial hyperpermeability in severe pulmonary arterial hypertension: role of store-operated calcium entry. Am J Physiol Lung Cell Mol Physiol 2016; 311:L560-9. [PMID: 27422996 DOI: 10.1152/ajplung.00057.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/07/2016] [Indexed: 11/22/2022] Open
Abstract
Here, we tested the hypothesis that animals with severe pulmonary arterial hypertension (PAH) display increased sensitivity to vascular permeability induced by activation of store-operated calcium entry. To test this hypothesis, wild-type and transient receptor potential channel 4 (TRPC4) knockout Fischer 344 rats were given a single injection of Semaxanib (SU5416; 20 mg/kg) followed by 3 wk of exposure to hypoxia (10% oxygen) and a return to normoxia (21% oxygen) for an additional 2-3 wk. This Semaxanib/hypoxia/normoxia (i.e., SU5416/hypoxia/normoxia) treatment caused PAH, as evidenced by development of right ventricular hypertrophy, pulmonary artery medial hypertrophy, and occlusive lesions within precapillary arterioles. Pulmonary artery pressure was increased fivefold in Semaxanib/hypoxia/normoxia-treated animals compared with untreated, Semaxanib-treated, and hypoxia-treated controls, determined by isolated perfused lung studies. Thapsigargin induced a dose-dependent increase in permeability that was dependent on TRPC4 in the normotensive perfused lung. This increase in permeability was accentuated in PAH lungs but not in Semaxanib- or hypoxia-treated lungs. Fluid accumulated in large perivascular cuffs, and although alveolar fluid accumulation was not seen in histological sections, Evans blue dye conjugated to albumin was present in bronchoalveolar lavage fluid of hypertensive but not normotensive lungs. Thus PAH is accompanied by a TRPC4-dependent increase in the sensitivity to edemagenic agents that activate store-operated calcium entry.
Collapse
Affiliation(s)
- Chun Zhou
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama; and
| | - Mary I Townsley
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Department of Internal Medicine, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama; and
| | - Mikhail Alexeyev
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama; and
| | - Norbert F Voelkel
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Department of Internal Medicine, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama; and
| |
Collapse
|
28
|
Affiliation(s)
- Jianhua Xiong
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
29
|
West JD, Carrier EJ, Bloodworth NC, Schroer AK, Chen P, Ryzhova LM, Gladson S, Shay S, Hutcheson JD, Merryman WD. Serotonin 2B Receptor Antagonism Prevents Heritable Pulmonary Arterial Hypertension. PLoS One 2016; 11:e0148657. [PMID: 26863209 PMCID: PMC4749293 DOI: 10.1371/journal.pone.0148657] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/21/2016] [Indexed: 12/21/2022] Open
Abstract
Serotonergic anorexigens are the primary pharmacologic risk factor associated with pulmonary arterial hypertension (PAH), and the resulting PAH is clinically indistinguishable from the heritable form of disease, associated with BMPR2 mutations. Both BMPR2 mutation and agonists to the serotonin receptor HTR2B have been shown to cause activation of SRC tyrosine kinase; conversely, antagonists to HTR2B inhibit SRC trafficking and downstream function. To test the hypothesis that a HTR2B antagonist can prevent BMRP2 mutation induced PAH by restricting aberrant SRC trafficking and downstream activity, we exposed BMPR2 mutant mice, which spontaneously develop PAH, to a HTR2B antagonist, SB204741, to block the SRC activation caused by BMPR2 mutation. SB204741 prevented the development of PAH in BMPR2 mutant mice, reduced recruitment of inflammatory cells to their lungs, and reduced muscularization of their blood vessels. By atomic force microscopy, we determined that BMPR2 mutant mice normally had a doubling of vessel stiffness, which was substantially normalized by HTR2B inhibition. SB204741 reduced SRC phosphorylation and downstream activity in BMPR2 mutant mice. Gene expression arrays indicate that the primary changes were in cytoskeletal and muscle contractility genes. These results were confirmed by gel contraction assays showing that HTR2B inhibition nearly normalizes the 400% increase in gel contraction normally seen in BMPR2 mutant smooth muscle cells. Heritable PAH results from increased SRC activation, cellular contraction, and vascular resistance, but antagonism of HTR2B prevents SRC phosphorylation, downstream activity, and PAH in BMPR2 mutant mice.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein Receptors, Type II/deficiency
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Cell Movement/drug effects
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Gene Expression Profiling
- Gene Expression Regulation
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/prevention & control
- Indoles/pharmacology
- Lung/drug effects
- Lung/metabolism
- Lung/pathology
- Mice
- Mice, Transgenic
- Muscle Contraction/drug effects
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Mutation
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oligonucleotide Array Sequence Analysis
- Phosphorylation
- Protein Transport
- Receptor, Serotonin, 5-HT2B/genetics
- Receptor, Serotonin, 5-HT2B/metabolism
- Serotonin Antagonists/pharmacology
- Signal Transduction
- Urea/analogs & derivatives
- Urea/pharmacology
- Vascular Stiffness/drug effects
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- James D. West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
- * E-mail: (JDW); (WDM)
| | - Erica J. Carrier
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Nathaniel C. Bloodworth
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - Alison K. Schroer
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - Peter Chen
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Larisa M. Ryzhova
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - Santhi Gladson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Sheila Shay
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - W. David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
- * E-mail: (JDW); (WDM)
| |
Collapse
|
30
|
García de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev 2015; 27:65-79. [PMID: 26823333 DOI: 10.1016/j.cytogfr.2015.12.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular system is critical for developmental growth, tissue homeostasis and repair but also for tumor development. Bone morphogenetic protein (BMP) signaling has recently emerged as a fundamental pathway of the endothelium by regulating cardiovascular and lymphatic development and by being causative for several vascular dysfunctions. Two vascular disorders have been directly linked to impaired BMP signaling: pulmonary arterial hypertension and hereditary hemorrhagic telangiectasia. Endothelial BMP signaling critically depends on the cellular context, which includes among others vascular heterogeneity, exposure to flow, and the intertwining with other signaling cascades (Notch, WNT, Hippo and hypoxia). The purpose of this review is to highlight the most recent findings illustrating the clear need for reconsidering the role of BMPs in vascular biology.
Collapse
Affiliation(s)
- Amaya García de Vinuesa
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease, Leuven, Belgium; KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Médicale (INSERM, U1036), Grenoble F-38000, France; Commissariat à l'Énergie Atomique et aux Energies Alternatives, Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire Biologie du Cancer et de l'Infection, Grenoble F-38000, France; Université Grenoble-Alpes, Grenoble F-38000, France.
| |
Collapse
|
31
|
Benn A, Bredow C, Casanova I, Vukičević S, Knaus P. VE-cadherin facilitates BMP-induced endothelial cell permeability and signaling. J Cell Sci 2015; 129:206-18. [PMID: 26598555 PMCID: PMC4732303 DOI: 10.1242/jcs.179960] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/16/2015] [Indexed: 12/22/2022] Open
Abstract
Several vascular disorders, such as aberrant angiogenesis, atherosclerosis and pulmonary hypertension, have been linked to dysfunctional BMP signaling. Vascular hyperpermeability via distortion of endothelial cell adherens junctions is a common feature of these diseases, but the role of BMPs in this process has not been investigated. BMP signaling is initiated by binding of ligand to, and activation of, BMP type I (BMPRI) and type II (BMPRII) receptors. Internalization of VE-cadherin as well as c-Src kinase-dependent phosphorylation have been implicated in the loosening of cell–cell contacts, thereby modulating vascular permeability. Here we demonstrate that BMP6 induces hyperpermeabilization of human endothelial cells by inducing internalization and c-Src-dependent phosphorylation of VE-cadherin. Furthermore, we show BMP-dependent physical interaction of VE-cadherin with the BMP receptor ALK2 (BMPRI) and BMPRII, resulting in stabilization of the BMP receptor complex and, thereby, the support of BMP6-Smad signaling. Our results provide first insights into the molecular mechanism of BMP-induced vascular permeability, a hallmark of various vascular diseases, and provide the basis for further investigations of BMPs as regulators of vascular integrity, both under physiological and pathophysiological conditions. Summary: We reveal the molecular mechanism by which BMP6 induces hyperpermeabilization of the endothelium. This provides first insights into the mechanism of BMP-dependent vascular integrity in normal physiology and disease.
Collapse
Affiliation(s)
- Andreas Benn
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany DFG Graduate School 1093 Berlin School of Integrative Oncology, Berlin 13353, Germany DFG Graduate School 203 Berlin-Brandenburg School for Regenerative Therapies, Berlin 13353, Germany
| | - Clara Bredow
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Isabel Casanova
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Slobodan Vukičević
- Center for Translational and Clinical Research, University of Zagreb, Zagreb 10000, Croatia
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany DFG Graduate School 1093 Berlin School of Integrative Oncology, Berlin 13353, Germany DFG Graduate School 203 Berlin-Brandenburg School for Regenerative Therapies, Berlin 13353, Germany
| |
Collapse
|
32
|
Selective enhancement of endothelial BMPR-II with BMP9 reverses pulmonary arterial hypertension. Nat Med 2015. [PMID: 26076038 PMCID: PMC4496295 DOI: 10.1038/nm.3877,] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Genetic evidence implicates the loss of bone morphogenetic protein type II receptor (BMPR-II) signaling in the endothelium as an initiating factor in pulmonary arterial hypertension (PAH). However, selective targeting of this signaling pathway using BMP ligands has not yet been explored as a therapeutic strategy. Here, we identify BMP9 as the preferred ligand for preventing apoptosis and enhancing monolayer integrity in both pulmonary arterial endothelial cells and blood outgrowth endothelial cells from subjects with PAH who bear mutations in the gene encoding BMPR-II, BMPR2. Mice bearing a heterozygous knock-in allele of a human BMPR2 mutation, R899X, which we generated as an animal model of PAH caused by BMPR-II deficiency, spontaneously developed PAH. Administration of BMP9 reversed established PAH in these mice, as well as in two other experimental PAH models, in which PAH develops in response to either monocrotaline or VEGF receptor inhibition combined with chronic hypoxia. These results demonstrate the promise of direct enhancement of endothelial BMP signaling as a new therapeutic strategy for PAH.
Collapse
|
33
|
Long L, Ormiston ML, Yang X, Southwood M, Gräf S, Machado RD, Mueller M, Kinzel B, Yung LM, Wilkinson JM, Moore SD, Drake KM, Aldred MA, Yu P, Upton PD, Morrell NW. Selective enhancement of endothelial BMPR-II with BMP9 reverses pulmonary arterial hypertension. Nat Med 2015; 21:777-85. [PMID: 26076038 PMCID: PMC4496295 DOI: 10.1038/nm.3877] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/12/2015] [Indexed: 12/13/2022]
Abstract
Genetic evidence implicates the loss of bone morphogenetic protein type II receptor (BMPR-II) signaling in the endothelium as an initiating factor in pulmonary arterial hypertension (PAH). However, selective targeting of this signaling pathway using BMP ligands has not yet been explored as a therapeutic strategy. Here, we identify BMP9 as the preferred ligand for preventing apoptosis and enhancing monolayer integrity in both pulmonary arterial endothelial cells and blood outgrowth endothelial cells from subjects with PAH who bear mutations in the gene encoding BMPR-II, BMPR2. Mice bearing a heterozygous knock-in allele of a human BMPR2 mutation, R899X, which we generated as an animal model of PAH caused by BMPR-II deficiency, spontaneously developed PAH. Administration of BMP9 reversed established PAH in these mice, as well as in two other experimental PAH models, in which PAH develops in response to either monocrotaline or VEGF receptor inhibition combined with chronic hypoxia. These results demonstrate the promise of direct enhancement of endothelial BMP signaling as a new therapeutic strategy for PAH.
Collapse
Affiliation(s)
- Lu Long
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | - Mark L. Ormiston
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | - Xudong Yang
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | - Mark Southwood
- Department of Pathology, Papworth Hospital, Papworth Everard, UK
| | - Stefan Gräf
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | | | | | - Bernd Kinzel
- Novartis Institute for Biomedical Research, Basel, CH
| | - Lai Ming Yung
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Janine M. Wilkinson
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | - Stephen D. Moore
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | - Kylie M. Drake
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Cleveland, OH
| | - Micheala A. Aldred
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Cleveland, OH
| | - Paul Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Paul D. Upton
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| | - Nicholas W. Morrell
- University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Cambridge, UK
| |
Collapse
|