1
|
Deng L, Cao C, Cai Z, Wang Z, Leng B, Chen Z, Kong F, Zhou Z, He J, Nie X, Bian JS. STING Contributes to Pulmonary Hypertension by Targeting IFN and BMPR2 Signaling through Regulating of F2RL3. Am J Respir Cell Mol Biol 2024; 71:356-371. [PMID: 38864771 DOI: 10.1165/rcmb.2023-0308oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 06/12/2024] [Indexed: 06/13/2024] Open
Abstract
Pulmonary hypertension (PH) is an incurable disease characterized by pulmonary vascular remodeling. Endothelial injury and inflammation are the key triggers of disease initiation. Recent findings suggest that STING (stimulator of IFN genes) activation plays a critical role in endothelial dysfunction and IFN signaling. Here, we investigated the involvement of STING in the pathogenesis of PH. Patients with PH and rodent PH model samples, a Sugen 5416/hypoxia PH model, and pulmonary artery endothelial cells (PAECs) were used to evaluate the hypothesis. We found that the cyclic guanosine monophosphate-AMP synthase-STING signaling pathway was activated in lung tissues from rodent PH models and patients with PH and in TNF-α-induced PAECs in vitro. Specifically, STING expression was significantly elevated in the endothelial cells in PH disease settings. In the Sugen 5416/hypoxia mouse model, genetic knockout or pharmacological inhibition of STING prevented the progression of PH. Functionally, knockdown of STING reduced the proliferation and migration of PAECs. Mechanistically, STING transcriptionally regulates its binding partner F2RL3 (F2R-like thrombin or trypsin receptor 3) through the STING-NF-κB axis, which activated IFN signaling and repressed BMPR2 (bone morphogenetic protein receptor 2) signaling both in vitro and in vivo. Further analysis revealed that F2RL3 expression was increased in PH settings and identified negative feedback regulation of F2RL3/BMPR2 signaling. Accordingly, a positive correlation of expression amounts between STING and F2RL3/IFN-stimulated genes was observed in vivo. Our findings suggest that STING activation in PAECs plays a critical role in the pathobiology of PH. Targeting STING may be a promising therapeutic strategy for preventing the development of PH.
Collapse
Affiliation(s)
- Lin Deng
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chengrui Cao
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zongye Cai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziping Wang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Bin Leng
- Department of Food Science and Technology, National University of Singapore, Singapore
- National University of Singapore (Suzhou) Research Institute, Suzhou, China; and
| | - Zhen Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Fanhao Kong
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhiyue Zhou
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jun He
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiaowei Nie
- Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jin-Song Bian
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
2
|
Liu B, Azfar M, Legchenko E, West JA, Martin S, Van den Haute C, Baekelandt V, Wharton J, Howard L, Wilkins MR, Vangheluwe P, Morrell NW, Upton PD. ATP13A3 variants promote pulmonary arterial hypertension by disrupting polyamine transport. Cardiovasc Res 2024; 120:756-768. [PMID: 38626311 PMCID: PMC11135649 DOI: 10.1093/cvr/cvae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/23/2024] [Accepted: 02/25/2024] [Indexed: 04/18/2024] Open
Abstract
AIMS Potential loss-of-function variants of ATP13A3, the gene encoding a P5B-type transport ATPase of undefined function, were recently identified in patients with pulmonary arterial hypertension (PAH). ATP13A3 is implicated in polyamine transport but its function has not been fully elucidated. In this study, we sought to determine the biological function of ATP13A3 in vascular endothelial cells (ECs) and how PAH-associated variants may contribute to disease pathogenesis. METHODS AND RESULTS We studied the impact of ATP13A3 deficiency and overexpression in EC models [human pulmonary ECs, blood outgrowth ECs (BOECs), and human microvascular EC 1], including a PAH patient-derived BOEC line harbouring an ATP13A3 variant (LK726X). We also generated mice harbouring an Atp13a3 variant analogous to a human disease-associated variant to establish whether these mice develop PAH. ATP13A3 localized to the recycling endosomes of human ECs. Knockdown of ATP13A3 in ECs generally reduced the basal polyamine content and altered the expression of enzymes involved in polyamine metabolism. Conversely, overexpression of wild-type ATP13A3 increased polyamine uptake. Functionally, loss of ATP13A3 was associated with reduced EC proliferation, increased apoptosis in serum starvation, and increased monolayer permeability to thrombin. The assessment of five PAH-associated missense ATP13A3 variants (L675V, M850I, V855M, R858H, and L956P) confirmed loss-of-function phenotypes represented by impaired polyamine transport and dysregulated EC function. Furthermore, mice carrying a heterozygous germline Atp13a3 frameshift variant representing a human variant spontaneously developed a PAH phenotype, with increased pulmonary pressures, right ventricular remodelling, and muscularization of pulmonary vessels. CONCLUSION We identify ATP13A3 as a polyamine transporter controlling polyamine homeostasis in ECs, a deficiency of which leads to EC dysfunction and predisposes to PAH. This suggests a need for targeted therapies to alleviate the imbalances in polyamine homeostasis and EC dysfunction in PAH.
Collapse
Affiliation(s)
- Bin Liu
- Section of Cardio and Respiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge CB2 0BB, UK
| | - Mujahid Azfar
- Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, Box 802, 3000 Leuven, Belgium
| | - Ekaterina Legchenko
- Section of Cardio and Respiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge CB2 0BB, UK
| | - James A West
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, Hills Road, Cambridge CB2 0QQ, UK
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Shaun Martin
- Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, Box 802, 3000 Leuven, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Herestraat 49, Box 1023, 3000 Leuven, Belgium
- Leuven Viral Vector Core, KU Leuven, Herestraat 49, Box 1023, 3000 Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Herestraat 49, Box 1023, 3000 Leuven, Belgium
| | - John Wharton
- Faculty of Medicine, National Heart and Lung Institute, ICTEM Building, Imperial College, Du Cane Road, London W12 0NN, UK
| | - Luke Howard
- Faculty of Medicine, National Heart and Lung Institute, ICTEM Building, Imperial College, Du Cane Road, London W12 0NN, UK
| | - Martin R Wilkins
- Faculty of Medicine, National Heart and Lung Institute, ICTEM Building, Imperial College, Du Cane Road, London W12 0NN, UK
| | - Peter Vangheluwe
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Nicholas W Morrell
- Section of Cardio and Respiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge CB2 0BB, UK
| | - Paul D Upton
- Section of Cardio and Respiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge CB2 0BB, UK
| |
Collapse
|
3
|
Murugesan P, Zhang Y, Huang Y, Chenggong Zong N, Youn JY, Chen W, Wang C, Loscalzo J, Cai H. Reversal of Pulmonary Hypertension in a Human-Like Model: Therapeutic Targeting of Endothelial DHFR. Circ Res 2024; 134:351-370. [PMID: 38299369 PMCID: PMC10880947 DOI: 10.1161/circresaha.123.323090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and elevated mean pulmonary arterial pressure, resulting in right heart failure. METHODS Here, we show that direct targeting of the endothelium to uncouple eNOS (endothelial nitric oxide synthase) with DAHP (2,4-diamino 6-hydroxypyrimidine; an inhibitor of GTP cyclohydrolase 1, the rate-limiting synthetic enzyme for the critical eNOS cofactor tetrahydrobiopterin) induces human-like, time-dependent progression of PH phenotypes in mice. RESULTS Critical phenotypic features include progressive elevation in mean pulmonary arterial pressure, right ventricular systolic blood pressure, and right ventricle (RV)/left ventricle plus septum (LV+S) weight ratio; extensive vascular remodeling of pulmonary arterioles with increased medial thickness/perivascular collagen deposition and increased expression of PCNA (proliferative cell nuclear antigen) and alpha-actin; markedly increased total and mitochondrial superoxide production, substantially reduced tetrahydrobiopterin and nitric oxide bioavailabilities; and formation of an array of human-like vascular lesions. Intriguingly, novel in-house generated endothelial-specific dihydrofolate reductase (DHFR) transgenic mice (tg-EC-DHFR) were completely protected from the pathophysiological and molecular features of PH upon DAHP treatment or hypoxia exposure. Furthermore, DHFR overexpression with a pCMV-DHFR plasmid transfection in mice after initiation of DAHP treatment completely reversed PH phenotypes. DHFR knockout mice spontaneously developed PH at baseline and had no additional deterioration in response to hypoxia, indicating an intrinsic role of DHFR deficiency in causing PH. RNA-sequencing experiments indicated great similarity in gene regulation profiles between the DAHP model and human patients with PH. CONCLUSIONS Taken together, these results establish a novel human-like murine model of PH that has long been lacking in the field, which can be broadly used for future mechanistic and translational studies. These data also indicate that targeting endothelial DHFR deficiency represents a novel and robust therapeutic strategy for the treatment of PH.
Collapse
Affiliation(s)
- Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yuanli Huang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Nobel Chenggong Zong
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Wenhui Chen
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Chen Wang
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| |
Collapse
|
4
|
Welch CL, Aldred MA, Balachandar S, Dooijes D, Eichstaedt CA, Gräf S, Houweling AC, Machado RD, Pandya D, Prapa M, Shaukat M, Southgate L, Tenorio-Castano J, Chung WK. Defining the clinical validity of genes reported to cause pulmonary arterial hypertension. Genet Med 2023; 25:100925. [PMID: 37422716 PMCID: PMC10766870 DOI: 10.1016/j.gim.2023.100925] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
PURPOSE Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. Genetic testing is currently recommended for adults diagnosed with heritable, idiopathic, anorexigen-, hereditary hemorrhagic telangiectasia-, and congenital heart disease-associated PAH, PAH with overt features of venous/capillary involvement, and all children diagnosed with PAH. Variants in at least 27 genes have putative evidence for PAH causality. Rigorous assessment of the evidence is needed to inform genetic testing. METHODS An international panel of experts in PAH applied a semi-quantitative scoring system developed by the NIH Clinical Genome Resource to classify the relative strength of evidence supporting PAH gene-disease relationships based on genetic and experimental evidence. RESULTS Twelve genes (BMPR2, ACVRL1, ATP13A3, CAV1, EIF2AK4, ENG, GDF2, KCNK3, KDR, SMAD9, SOX17, and TBX4) were classified as having definitive evidence and 3 genes (ABCC8, GGCX, and TET2) with moderate evidence. Six genes (AQP1, BMP10, FBLN2, KLF2, KLK1, and PDGFD) were classified as having limited evidence for causal effects of variants. TOPBP1 was classified as having no known PAH relationship. Five genes (BMPR1A, BMPR1B, NOTCH3, SMAD1, and SMAD4) were disputed because of a paucity of genetic evidence over time. CONCLUSION We recommend that genetic testing includes all genes with definitive evidence and that caution be taken in the interpretation of variants identified in genes with moderate or limited evidence. Genes with no known evidence for PAH or disputed genes should not be included in genetic testing.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Srimmitha Balachandar
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Dennis Dooijes
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- NIHR BioResource for Translational Research - Rare Diseases, Department of Haemotology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Divya Pandya
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Matina Prapa
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Memoona Shaukat
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Jair Tenorio-Castano
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IDiPAZ, Universidad Autonoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain; ITHACA, European Reference Network, Brussels, Belgium
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY; Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
5
|
Fließer E, Lins T, Berg JL, Kolb M, Kwapiszewska G. The endothelium in lung fibrosis: a core signaling hub in disease pathogenesis? Am J Physiol Cell Physiol 2023; 325:C2-C16. [PMID: 37184232 DOI: 10.1152/ajpcell.00097.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
Pulmonary fibrosis (PF) is a progressive chronic lung disease characterized by excessive deposition of extracellular matrix (ECM) and structural destruction, associated with a severe 5-year mortality rate. The onset of the disease is thought to be triggered by chronic damage to the alveolar epithelium. Since the pulmonary endothelium is an important component of the alveolar-capillary niche, it is also affected by the initial injury. In addition to ensuring proper gas exchange, the endothelium has critical functional properties, including regulation of vascular tone, inflammatory responses, coagulation, and maintenance of vascular homeostasis and integrity. Recent single-cell analyses have shown that shifts in endothelial cell (EC) subtypes occur in PF. Furthermore, the increased vascular remodeling associated with PF leads to deteriorated outcomes for patients, underscoring the importance of the vascular bed in PF. To date, the causes and consequences of endothelial and vascular involvement in lung fibrosis are poorly understood. Therefore, it is of great importance to investigate the involvement of EC and the vascular system in the pathogenesis of the disease. In this review, we will outline the current knowledge on the role of the pulmonary vasculature in PF, in terms of abnormal cellular interactions, hyperinflammation, vascular barrier disorders, and an altered basement membrane composition. Finally, we will summarize recent advances in extensive therapeutic research and discuss the significant value of novel therapies targeting the endothelium.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Thomas Lins
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Johannes Lorenz Berg
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Cardiopulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
6
|
Inactivating the Uninhibited: The Tale of Activins and Inhibins in Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24043332. [PMID: 36834742 PMCID: PMC9963072 DOI: 10.3390/ijms24043332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Advances in technology and biomedical knowledge have led to the effective diagnosis and treatment of an increasing number of rare diseases. Pulmonary arterial hypertension (PAH) is a rare disorder of the pulmonary vasculature that is associated with high mortality and morbidity rates. Although significant progress has been made in understanding PAH and its diagnosis and treatment, numerous unanswered questions remain regarding pulmonary vascular remodeling, a major factor contributing to the increase in pulmonary arterial pressure. Here, we discuss the role of activins and inhibins, both of which belong to the TGF-β superfamily, in PAH development. We examine how these relate to signaling pathways implicated in PAH pathogenesis. Furthermore, we discuss how activin/inhibin-targeting drugs, particularly sotatercep, affect pathophysiology, as these target the afore-mentioned specific pathway. We highlight activin/inhibin signaling as a critical mediator of PAH development that is to be targeted for therapeutic gain, potentially improving patient outcomes in the future.
Collapse
|
7
|
Aman J, Morrell NW, Rhodes CJ, Wilkins MR, Bogaard HJ. The SOX17 phenotype in pulmonary arterial hypertension: lessons for pathobiology and clinical management. Eur Respir J 2022; 60:2201438. [PMID: 37651375 DOI: 10.1183/13993003.01438-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/11/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Jurjan Aman
- Dept of Pulmonology, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Christopher J Rhodes
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Dept of Pulmonology, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Zhang V, Ganz T, Nemeth E, Umar S, Kim A. Bmpr2 mutant mice are an inadequate model for studying iron deficiency in pulmonary hypertension. Pulm Circ 2022; 12:e12151. [PMID: 36568690 PMCID: PMC9768458 DOI: 10.1002/pul2.12151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/06/2022] [Accepted: 10/10/2022] [Indexed: 02/06/2023] Open
Abstract
As bone morphogenetic protein receptor type II (Bmpr2) mutations are the most common genetic cause of pulmonary arterial hypertension (PAH), and iron deficiency (ID) is associated with worse clinical outcomes in PAH patients, we proposed to use Bmpr2 ± mice to induce a model of ID in pulmonary vascular disease. Our study shows that these transgenic mice are not a good model for this clinical phenomenon.
Collapse
Affiliation(s)
- Vida Zhang
- Department of MedicineDavid Geffen School of Medicine, UCLALos AngelesCaliforniaUSA
- Department of Molecular and Medical PharmacologyUCLALos AngelesCaliforniaUSA
| | - Tomas Ganz
- Department of MedicineDavid Geffen School of Medicine, UCLALos AngelesCaliforniaUSA
| | - Elizabeta Nemeth
- Department of MedicineDavid Geffen School of Medicine, UCLALos AngelesCaliforniaUSA
| | - Soban Umar
- Department of AnesthesiologyDavid Geffen School of Medicine, UCLALos AngelesCaliforniaUSA
| | - Airie Kim
- Department of MedicineDavid Geffen School of Medicine, UCLALos AngelesCaliforniaUSA
| |
Collapse
|
9
|
Aldred MA, Morrell NW, Guignabert C. New Mutations and Pathogenesis of Pulmonary Hypertension: Progress and Puzzles in Disease Pathogenesis. Circ Res 2022; 130:1365-1381. [PMID: 35482831 PMCID: PMC9897592 DOI: 10.1161/circresaha.122.320084] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex multifactorial disease with poor prognosis characterized by functional and structural alterations of the pulmonary circulation causing marked increase in pulmonary vascular resistance, ultimately leading to right heart failure and death. Mutations in the gene encoding BMPRII-a receptor for the TGF-β (transforming growth factor-beta) superfamily-account for over 70% of families with PAH and ≈20% of sporadic cases. In recent years, however, less common or rare mutations in other genes have been identified. This review will consider how these newly discovered PAH genes could help to provide a better understanding of the molecular and cellular bases of the maintenance of the pulmonary vascular integrity, as well as their role in the PAH pathogenesis underlying occlusion of arterioles in the lung. We will also discuss how insights into the genetic contributions of these new PAH-related genes may open up new therapeutic targets for this, currently incurable, cardiopulmonary disorder.
Collapse
Affiliation(s)
- Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicholas W Morrell
- University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, UK
| | - Christophe Guignabert
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France,Université Paris-Saclay, Faculté de Médecine, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
10
|
Abstract
Pulmonary hypertension (PH) describes heterogeneous population of patients with a mean pulmonary arterial pressure >20 mm Hg. Rarely, PH presents as a primary disorder but is more commonly part of a complex phenotype associated with comorbidities. Regardless of the cause, PH reduces life expectancy and impacts quality of life. The current clinical classification divides PH into 1 of 5 diagnostic groups to assign treatment. There are currently no pharmacological cures for any form of PH. Animal models are essential to help decipher the molecular mechanisms underlying the disease, to assign genotype-phenotype relationships to help identify new therapeutic targets, and for clinical translation to assess the mechanism of action and putative efficacy of new therapies. However, limitations inherent of all animal models of disease limit the ability of any single model to fully recapitulate complex human disease. Within the PH community, we are often critical of animal models due to the perceived low success upon clinical translation of new drugs. In this review, we describe the characteristics, advantages, and disadvantages of existing animal models developed to gain insight into the molecular and pathological mechanisms and test new therapeutics, focusing on adult forms of PH from groups 1 to 3. We also discuss areas of improvement for animal models with approaches combining several hits to better reflect the clinical situation and elevate their translational value.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allan Lawrie
- Dept of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK & Insigneo institute for in silico medicine, Sheffield, UK
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
11
|
Shi B, Zhou T, Lv S, Wang M, Chen S, Heidari AA, Huang X, Chen H, Wang L, Wu P. An evolutionary machine learning for pulmonary hypertension animal model from arterial blood gas analysis. Comput Biol Med 2022; 146:105529. [PMID: 35594682 DOI: 10.1016/j.compbiomed.2022.105529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/03/2022]
Abstract
Pulmonary hypertension (PH) is a rare and fatal condition that leads to right heart failure and death. The pathophysiology of PH and potential therapeutic approaches are yet unknown. PH animal models' development and proper evaluation are critical to PH research. This work presents an effective analysis technology for PH from arterial blood gas analysis utilizing an evolutionary kernel extreme learning machine with multiple strategies integrated slime mould algorithm (MSSMA). In MSSMA, two efficient bee-foraging learning operators are added to the original slime mould algorithm, ensuring a suitable trade-off between intensity and diversity. The proposed MSSMA is evaluated on thirty IEEE benchmarks and the statistical results show that the search performance of the MSSMA is significantly improved. The MSSMA is utilised to develop a kernel extreme learning machine (MSSMA-KELM) on PH from arterial blood gas analysis. Comprehensively, the proposed MSSMA-KELM can be used as an effective analysis technology for PH from arterial Blood gas analysis with an accuracy of 93.31%, Matthews coefficient of 90.13%, Sensitivity of 91.12%, and Specificity of 90.73%. MSSMA-KELM can be treated as an effective approach for evaluating mouse PH models.
Collapse
Affiliation(s)
- Beibei Shi
- Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu, 212000, China.
| | - Tao Zhou
- The First Clinical College, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Shushu Lv
- The First Clinical College, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Mingjing Wang
- College of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou, 325035, China.
| | - Siyuan Chen
- Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu, 212000, China.
| | - Ali Asghar Heidari
- School of Surveying and Geospatial Engineering, College of Engineering, University of Tehran, Tehran, Iran; Department of Computer Science, School of Computing, National University of Singapore, Singapore, Singapore.
| | - Xiaoying Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Huiling Chen
- College of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou, 325035, China.
| | - Liangxing Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Peiliang Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
12
|
Andre P, Joshi SR, Briscoe SD, Alexander MJ, Li G, Kumar R. Therapeutic Approaches for Treating Pulmonary Arterial Hypertension by Correcting Imbalanced TGF-β Superfamily Signaling. Front Med (Lausanne) 2022; 8:814222. [PMID: 35141256 PMCID: PMC8818880 DOI: 10.3389/fmed.2021.814222] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterized by high blood pressure in the pulmonary circulation driven by pathological remodeling of distal pulmonary arteries, leading typically to death by right ventricular failure. Available treatments improve physical activity and slow disease progression, but they act primarily as vasodilators and have limited effects on the biological cause of the disease—the uncontrolled proliferation of vascular endothelial and smooth muscle cells. Imbalanced signaling by the transforming growth factor-β (TGF-β) superfamily contributes extensively to dysregulated vascular cell proliferation in PAH, with overactive pro-proliferative SMAD2/3 signaling occurring alongside deficient anti-proliferative SMAD1/5/8 signaling. We review the TGF-β superfamily mechanisms underlying PAH pathogenesis, superfamily interactions with inflammation and mechanobiological forces, and therapeutic strategies under development that aim to restore SMAD signaling balance in the diseased pulmonary arterial vessels. These strategies could potentially reverse pulmonary arterial remodeling in PAH by targeting causative mechanisms and therefore hold significant promise for the PAH patient population.
Collapse
|
13
|
Kelly NJ, Chan SY. Pulmonary Arterial Hypertension: Emerging Principles of Precision Medicine across Basic Science to Clinical Practice. Rev Cardiovasc Med 2022; 23:378. [PMID: 36875282 PMCID: PMC9980296 DOI: 10.31083/j.rcm2311378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an enigmatic and deadly vascular disease with no known cure. Recent years have seen rapid advances in our understanding of the molecular underpinnings of PAH, with an expanding knowledge of the molecular, cellular, and systems-level drivers of disease that are being translated into novel therapeutic modalities. Simultaneous advances in clinical technology have led to a growing list of tools with potential application to diagnosis and phenotyping. Guided by fundamental biology, these developments hold the potential to usher in a new era of personalized medicine in PAH with broad implications for patient management and great promise for improved outcomes.
Collapse
Affiliation(s)
- Neil J Kelly
- Center for Pulmonary Vascular Biology and Medicine and Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute; Division of Cardiology; Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine and Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute; Division of Cardiology; Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
14
|
Ruhr I, Bierstedt J, Rhen T, Das D, Singh SK, Miller S, Crossley DA, Galli GLJ. Developmental programming of DNA methylation and gene expression patterns is associated with extreme cardiovascular tolerance to anoxia in the common snapping turtle. Epigenetics Chromatin 2021; 14:42. [PMID: 34488850 PMCID: PMC8420019 DOI: 10.1186/s13072-021-00414-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022] Open
Abstract
Background Environmental fluctuation during embryonic and fetal development can permanently alter an organism’s morphology, physiology, and behaviour. This phenomenon, known as developmental plasticity, is particularly relevant to reptiles that develop in subterranean nests with variable oxygen tensions. Previous work has shown hypoxia permanently alters the cardiovascular system of snapping turtles and may improve cardiac anoxia tolerance later in life. The mechanisms driving this process are unknown but may involve epigenetic regulation of gene expression via DNA methylation. To test this hypothesis, we assessed in situ cardiac performance during 2 h of acute anoxia in juvenile turtles previously exposed to normoxia (21% oxygen) or hypoxia (10% oxygen) during embryogenesis. Next, we analysed DNA methylation and gene expression patterns in turtles from the same cohorts using whole genome bisulfite sequencing, which represents the first high-resolution investigation of DNA methylation patterns in any reptilian species. Results Genome-wide correlations between CpG and CpG island methylation and gene expression patterns in the snapping turtle were consistent with patterns observed in mammals. As hypothesized, developmental hypoxia increased juvenile turtle cardiac anoxia tolerance and programmed DNA methylation and gene expression patterns. Programmed differences in expression of genes such as SCN5A may account for differences in heart rate, while genes such as TNNT2 and TPM3 may underlie differences in calcium sensitivity and contractility of cardiomyocytes and cardiac inotropy. Finally, we identified putative transcription factor-binding sites in promoters and in differentially methylated CpG islands that suggest a model linking programming of DNA methylation during embryogenesis to differential gene expression and cardiovascular physiology later in life. Binding sites for hypoxia inducible factors (HIF1A, ARNT, and EPAS1) and key transcription factors activated by MAPK and BMP signaling (RREB1 and SMAD4) are implicated. Conclusions Our data strongly suggests that DNA methylation plays a conserved role in the regulation of gene expression in reptiles. We also show that embryonic hypoxia programs DNA methylation and gene expression patterns and that these changes are associated with enhanced cardiac anoxia tolerance later in life. Programming of cardiac anoxia tolerance has major ecological implications for snapping turtles, because these animals regularly exploit anoxic environments throughout their lifespan. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00414-7.
Collapse
Affiliation(s)
- Ilan Ruhr
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, M13 9NT, UK
| | - Jacob Bierstedt
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Turk Rhen
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Debojyoti Das
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Sunil Kumar Singh
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Soleille Miller
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Dane A Crossley
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Gina L J Galli
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, M13 9NT, UK
| |
Collapse
|
15
|
Boehm M, Tian X, Ali MK, Mao Y, Ichimura K, Zhao M, Kuramoto K, Dannewitz Prosseda S, Fajardo G, Dufva MJ, Qin X, Kheyfets VO, Bernstein D, Reddy S, Metzger RJ, Zamanian RT, Haddad F, Spiekerkoetter E. Improving Right Ventricular Function by Increasing BMP Signaling with FK506. Am J Respir Cell Mol Biol 2021; 65:272-287. [PMID: 33938785 PMCID: PMC8485990 DOI: 10.1165/rcmb.2020-0528oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/20/2021] [Indexed: 11/24/2022] Open
Abstract
Right ventricular (RV) function is the predominant determinant of survival in patients with pulmonary arterial hypertension (PAH). In preclinical models, pharmacological activation of BMP (bone morphogenetic protein) signaling with FK506 (tacrolimus) improved RV function by decreasing RV afterload. FK506 therapy further stabilized three patients with end-stage PAH. Whether FK506 has direct effects on the pressure-overloaded right ventricle is yet unknown. We hypothesized that increasing cardiac BMP signaling with FK506 improves RV structure and function in a model of fixed RV afterload after pulmonary artery banding (PAB). Direct cardiac effects of FK506 on the microvasculature and RV fibrosis were studied after surgical PAB in wild-type and heterozygous Bmpr2 mutant mice. RV function and strain were assessed longitudinally via cardiac magnetic resonance imaging during continuous FK506 infusion. Genetic lineage tracing of endothelial cells (ECs) was performed to assess the contribution of ECs to fibrosis. Molecular mechanistic studies were performed in human cardiac fibroblasts and ECs. In mice, low BMP signaling in the right ventricle exaggerated PAB-induced RV fibrosis. FK506 therapy restored cardiac BMP signaling, reduced RV fibrosis in a BMP-dependent manner independent from its immunosuppressive effect, preserved RV capillarization, and improved RV function and strain over the time course of disease. Endothelial mesenchymal transition was a rare event and did not significantly contribute to cardiac fibrosis after PAB. Mechanistically, FK506 required ALK1 in human cardiac fibroblasts as a BMPR2 co-receptor to reduce TGFβ1-induced proliferation and collagen production. Our study demonstrates that increasing cardiac BMP signaling with FK506 improves RV structure and function independent from its previously described beneficial effects on pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Mario Boehm
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardio-Pulmonary Institute, Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany
| | - Xuefei Tian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Md Khadem Ali
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Yuqiang Mao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Kenzo Ichimura
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Mingming Zhao
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Kazuya Kuramoto
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Svenja Dannewitz Prosseda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Giovanni Fajardo
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Melanie J. Dufva
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado; and
- Department of Pediatrics, Section of Cardiology, Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Xulei Qin
- Cardiovascular Institute, and
- Department of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Vitaly O. Kheyfets
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado; and
- Department of Pediatrics, Section of Cardiology, Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Daniel Bernstein
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Sushma Reddy
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Ross J. Metzger
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Roham T. Zamanian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
| | - Francois Haddad
- Cardiovascular Institute, and
- Department of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
| |
Collapse
|
16
|
Dignam JP, Scott TE, Kemp-Harper BK, Hobbs AJ. Animal models of pulmonary hypertension: Getting to the heart of the problem. Br J Pharmacol 2021; 179:811-837. [PMID: 33724447 DOI: 10.1111/bph.15444] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/04/2021] [Accepted: 03/06/2021] [Indexed: 12/12/2022] Open
Abstract
Despite recent therapeutic advances, pulmonary hypertension (PH) remains a fatal disease due to the development of right ventricular (RV) failure. At present, no treatments targeted at the right ventricle are available, and RV function is not widely considered in the preclinical assessment of new therapeutics. Several small animal models are used in the study of PH, including the classic models of exposure to either hypoxia or monocrotaline, newer combinational and genetic models, and pulmonary artery banding, a surgical model of pure RV pressure overload. These models reproduce selected features of the structural remodelling and functional decline seen in patients and have provided valuable insight into the pathophysiology of RV failure. However, significant reversal of remodelling and improvement in RV function remains a therapeutic obstacle. Emerging animal models will provide a deeper understanding of the mechanisms governing the transition from adaptive remodelling to a failing right ventricle, aiding the hunt for druggable molecular targets.
Collapse
Affiliation(s)
- Joshua P Dignam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tara E Scott
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University Clayton Campus, Clayton, Victoria, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Parkville, Victoria, Australia
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University Clayton Campus, Clayton, Victoria, Australia
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
17
|
Guignabert C, Humbert M. Targeting transforming growth factor-β receptors in pulmonary hypertension. Eur Respir J 2021; 57:2002341. [PMID: 32817256 DOI: 10.1183/13993003.02341-2020] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
Abstract
The transforming growth factor-β (TGF-β) superfamily includes several groups of multifunctional proteins that form two major branches, namely the TGF-β-activin-nodal branch and the bone morphogenetic protein (BMP)-growth differentiation factor (GDF) branch. The response to the activation of these two branches, acting through canonical (small mothers against decapentaplegic (Smad) 2/3 and Smad 1/5/8, respectively) and noncanonical signalling pathways, are diverse and vary for different environmental conditions and cell types. An extensive body of data gathered in recent years has demonstrated a central role for the cross-talk between these two branches in a number of cellular processes, which include the regulation of cell proliferation and differentiation, as well as the transduction of signalling cascades for the development and maintenance of different tissues and organs. Importantly, alterations in these pathways, which include heterozygous germline mutations and/or alterations in the expression of several constitutive members, have been identified in patients with familial/heritable pulmonary arterial hypertension (PAH) or idiopathic PAH (IPAH). Consequently, loss or dysfunction in the delicate, finely-tuned balance between the TGF-β-activin-nodal branch and the BMP-GDF branch are currently viewed as the major molecular defect playing a critical role in PAH predisposition and disease progression. Here we review the role of the TGF-β-activin-nodal branch in PAH and illustrate how this knowledge has not only provided insight into understanding its pathogenesis, but has also paved the way for possible novel therapeutic approaches.
Collapse
Affiliation(s)
- Christophe Guignabert
- Faculty of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 (Pulmonary Hypertension: Pathophysiology and Novel Therapies), Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Marc Humbert
- Faculty of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 (Pulmonary Hypertension: Pathophysiology and Novel Therapies), Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Dept of Respiratory and Intensive Care Medicine, French Pulmonary Hypertension Reference Center, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| |
Collapse
|
18
|
Dunmore BJ, Jones RJ, Toshner MR, Upton PD, Morrell NW. Approaches to treat pulmonary arterial hypertension by targeting bmpr2 - from cell membrane to nucleus. Cardiovasc Res 2021; 117:2309-2325. [PMID: 33399862 DOI: 10.1093/cvr/cvaa350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/06/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is estimated to affect between 10-50 people per million worldwide. The lack of cure and devastating nature of the disease means that treatment is crucial to arrest rapid clinical worsening. Current therapies are limited by their focus on inhibiting residual vasoconstriction rather than targeting key regulators of the cellular pathology. Potential disease-modifying therapies may come from research directed towards causal pathways involved in the cellular and molecular mechanisms of disease. It is widely acknowledged, that targeting reduced expression of the critical bone morphogenetic protein type-2 receptor (BMPR2) and its associated signalling pathways is a compelling therapeutic avenue to explore. In this review we highlight the advances that have been made in understanding this pathway and the therapeutics that are being tested in clinical trials and the clinic to treat PAH.
Collapse
Affiliation(s)
- Benjamin J Dunmore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Rowena J Jones
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Mark R Toshner
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Paul D Upton
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| |
Collapse
|
19
|
Swietlik EM, Prapa M, Martin JM, Pandya D, Auckland K, Morrell NW, Gräf S. 'There and Back Again'-Forward Genetics and Reverse Phenotyping in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:E1408. [PMID: 33256119 PMCID: PMC7760524 DOI: 10.3390/genes11121408] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Although the invention of right heart catheterisation in the 1950s enabled accurate clinical diagnosis of pulmonary arterial hypertension (PAH), it was not until 2000 when the landmark discovery of the causative role of bone morphogenetic protein receptor type II (BMPR2) mutations shed new light on the pathogenesis of PAH. Since then several genes have been discovered, which now account for around 25% of cases with the clinical diagnosis of idiopathic PAH. Despite the ongoing efforts, in the majority of patients the cause of the disease remains elusive, a phenomenon often referred to as "missing heritability". In this review, we discuss research approaches to uncover the genetic architecture of PAH starting with forward phenotyping, which in a research setting should focus on stable intermediate phenotypes, forward and reverse genetics, and finally reverse phenotyping. We then discuss potential sources of "missing heritability" and how functional genomics and multi-omics methods are employed to tackle this problem.
Collapse
Affiliation(s)
- Emilia M. Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Matina Prapa
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Jennifer M. Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Kathryn Auckland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| |
Collapse
|
20
|
Theilmann AL, Hawke LG, Hilton LR, Whitford MKM, Cole DV, Mackeil JL, Dunham-Snary KJ, Mewburn J, James PD, Maurice DH, Archer SL, Ormiston ML. Endothelial BMPR2 Loss Drives a Proliferative Response to BMP (Bone Morphogenetic Protein) 9 via Prolonged Canonical Signaling. Arterioscler Thromb Vasc Biol 2020; 40:2605-2618. [PMID: 32998516 PMCID: PMC7571847 DOI: 10.1161/atvbaha.119.313357] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Supplemental Digital Content is available in the text. Pulmonary arterial hypertension is a disease of proliferative vascular occlusion that is strongly linked to mutations in BMPR2—the gene encoding the BMPR-II (BMP [bone morphogenetic protein] type II receptor). The endothelial-selective BMPR-II ligand, BMP9, reverses disease in animal models of pulmonary arterial hypertension and suppresses the proliferation of healthy endothelial cells. However, the impact of BMPR2 loss on the antiproliferative actions of BMP9 has yet to be assessed.
Collapse
Affiliation(s)
- Anne L Theilmann
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - Lindsey G Hawke
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - L Rhiannon Hilton
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - Mara K M Whitford
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - Devon V Cole
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - Jodi L Mackeil
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - Kimberly J Dunham-Snary
- Department of Medicine (K.J.D.-S., J.M., P.D.J., S.L.A., M.L.O.), Queen's University, Kingston, Canada
| | - Jeffrey Mewburn
- Department of Medicine (K.J.D.-S., J.M., P.D.J., S.L.A., M.L.O.), Queen's University, Kingston, Canada
| | - Paula D James
- Department of Medicine (K.J.D.-S., J.M., P.D.J., S.L.A., M.L.O.), Queen's University, Kingston, Canada
| | - Donald H Maurice
- Department of Biomedical and Molecular Sciences (A.L.T., L.G.H., L.R.H., M.K.M.W., D.V.C., J.L.M., D.H.M., M.L.O.), Queen's University, Kingston, Canada
| | - Stephen L Archer
- Department of Medicine (K.J.D.-S., J.M., P.D.J., S.L.A., M.L.O.), Queen's University, Kingston, Canada
| | - Mark L Ormiston
- Department of Surgery (M.L.O.), Queen's University, Kingston, Canada
| |
Collapse
|
21
|
Emanuelli G, Nassehzadeh-Tabriz N, Morrell NW, Marciniak SJ. The integrated stress response in pulmonary disease. Eur Respir Rev 2020; 29:29/157/200184. [PMID: 33004527 PMCID: PMC7116220 DOI: 10.1183/16000617.0184-2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
The respiratory tract and its resident immune cells face daily exposure
to stress, both from without and from within. Inhaled pathogens, including
severe acute respiratory syndrome coronavirus 2, and toxins from pollution
trigger a cellular defence system that reduces protein synthesis to minimise
viral replication or the accumulation of misfolded proteins. Simultaneously, a
gene expression programme enhances antioxidant and protein folding machineries
in the lung. Four kinases (PERK, PKR, GCN2 and HRI) sense a diverse range of
stresses to trigger this “integrated stress response”. Here we review recent
advances identifying the integrated stress response as a critical pathway in the
pathogenesis of pulmonary diseases, including pneumonias, thoracic malignancy,
pulmonary fibrosis and pulmonary hypertension. Understanding the integrated
stress response provides novel targets for the development of therapies.
Collapse
Affiliation(s)
- Giulia Emanuelli
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK.,Division of Respiratory Medicine, Dept of Medicine, University of Cambridge, Cambridge, UK.,Equal first authors
| | - Nikou Nassehzadeh-Tabriz
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK.,Equal first authors
| | - Nick W Morrell
- Division of Respiratory Medicine, Dept of Medicine, University of Cambridge, Cambridge, UK
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK .,Division of Respiratory Medicine, Dept of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Liparulo A, Esposito R, Santonocito D, Muñoz-Ramírez A, Spaziano G, Bruno F, Xiao J, Puglia C, Filosa R, Berrino L, D'Agostino B. Formulation and Characterization of Solid Lipid Nanoparticles Loading RF22-c, a Potent and Selective 5-LO Inhibitor, in a Monocrotaline-Induced Model of Pulmonary Hypertension. Front Pharmacol 2020; 11:83. [PMID: 32180715 PMCID: PMC7059131 DOI: 10.3389/fphar.2020.00083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare but fatal disease characterized by persistent elevated blood pressure in the pulmonary circulation, due to increased resistance to blood flow, through the lungs. Advances in the understanding of the pathobiology of PAH clarify the role of leukotrienes (LTs) that appear to be an exciting new target for disease intervention. Over the years, our group has long investigated this field, detecting the 1,2-benzoquinone RF-22c as the most powerful and selective competitive inhibitor of the enzyme 5-lipoxygenase (5-LO). With the aim to improve the bioavailability of RF-22c and to confirm the role of 5-LO as therapeutic strategy for PAH treatment, we developed a solid lipid nanoparticle (SLN) loaded with drug. Therefore, in monocrotaline (MCT) rat model of PAH, the role of 5-LO has been investigated through the formulation of RF-22c-SLN. The rats were randomly grouped into control group, MCT group, and MCT + RF22-c group. After 21 days, all the animals were sacrificed to perform functional and histological evaluations. RF22-c-SLN treatment was able to significantly reduce the mean pulmonary arterial pressure (mPAP) and precapillary resistance (R-pre) compared to the MCT group. The MCT induced rise in medial wall thickness of pulmonary arterioles, and the cardiomyocytes width were significantly attenuated by RF22-c-SLN formulation upon treatment. The results showed that the selective inhibition of 5-LO improved hemodynamic parameters as well as vascular and cardiac remodeling by preventing induced pulmonary hypertension. The improved sustained release properties and targeting abilities achieved with the innovative nanotechnological approach may be therapeutically beneficial for PAH patients as a consequence of the increase of pharmacological effects and of the possible reduction and/or optimization of the drug frequency of administration.
Collapse
Affiliation(s)
- Angela Liparulo
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli,” University of Campania “L. Vanvitelli,”Naples, Italy
| | - Renata Esposito
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli,” University of Campania “L. Vanvitelli,”Naples, Italy
| | | | - Alejandra Muñoz-Ramírez
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago, Casilla, Correo, Chile
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “L. Vanvitelli,”Naples, Italy
| | - Giuseppe Spaziano
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli,” University of Campania “L. Vanvitelli,”Naples, Italy
| | - Ferdinando Bruno
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “L. Vanvitelli,”Naples, Italy
| | - Jianbo Xiao
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| | - Carmelo Puglia
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Rosanna Filosa
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “L. Vanvitelli,”Naples, Italy
- Consorzio Sannio Tech-AMP Biotec, Apollosa, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli,” University of Campania “L. Vanvitelli,”Naples, Italy
| | - Bruno D'Agostino
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli,” University of Campania “L. Vanvitelli,”Naples, Italy
| |
Collapse
|
23
|
Voelkel NF, Peters-Golden M. A new treatment for severe pulmonary arterial hypertension based on an old idea: inhibition of 5-lipoxygenase. Pulm Circ 2020; 10:2045894019882635. [PMID: 32257113 PMCID: PMC7103594 DOI: 10.1177/2045894019882635] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022] Open
Abstract
It has been generally accepted that severe forms of pulmonary arterial hypertension are associated with inflammation. Plasma levels in patients with severe pulmonary arterial hypertension show elevated levels of interleukins and mediators of inflammation and histologically the diseased small pulmonary arterioles show infiltrates of inflammatory and immune cells. Here, we review the literature that connects pulmonary hypertension with the arachidonic acid/5-lipoxygenase-derived leukotriens. This mostly preclinical background data together with the availability of 5-lipoxygenase inhibitors and leukotriene receptor blockers provide the rationale for testing the hypothesis that 5-lipoxygenase products contribute to the pathobiology of severe pulmonary arterial hypertension in a subgroup of patients.
Collapse
Affiliation(s)
- Norbert F. Voelkel
- Department of Pulmonary Medicine,
University of Amsterdam Medical Centers, Amsterdam, the Netherlands
| | - Marc Peters-Golden
- Pulmonary and Critical Care Medicine
Division,
University
of Michigan Medical School, Ann Arbor, MI,
USA
| |
Collapse
|
24
|
Ikeda KT, Hale PT, Pauciulo MW, Dasgupta N, Pastura PA, Le Cras TD, Pandey MK, Nichols WC. Hypoxia-induced Pulmonary Hypertension in Different Mouse Strains: Relation to Transcriptome. Am J Respir Cell Mol Biol 2019; 60:106-116. [PMID: 30134121 DOI: 10.1165/rcmb.2017-0435oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Patients with pulmonary arterial hypertension (PAH) can harbor mutations in several genes, most commonly in BMPR2. However, disease penetrance in patients with BMPR2 mutations is low. In addition, most patients do not carry known PAH gene mutations, suggesting that other factors determine susceptibility to PAH. To begin to identify additional genomic factors contributing to PAH pathogenesis, we exposed 32 mouse strains to chronic hypoxia. We found that the PL/J strain has extremely high right ventricular systolic pressure (RVSP; 86.58 mm Hg) but minimal lung remodeling. To identify potential genomic factors contributing to the high RVSP, RNAseq analysis of PL/J lung mRNAs and microRNAs (miRNAs) after hypoxia was performed, and it demonstrated that 4 of 43 upregulated miRNAs in the Dlk1-Dio3 imprinting region are predicted to target T cell marker mRNAs. These target mRNAs, as well as the numbers of T cells were downregulated. In addition, C5a and its receptor, C5AR1, were increased. Analysis of Rho-associated protein kinase (Rock) 2 mRNA expression, in the RhoA/Rock pathway, demonstrated a significant increase in PL/J. Inhibition of Rock2 ameliorated a portion of the elevated RVSP. In addition, we identified miR-150-5p as a potential regulator of Rock2 expression. In conclusion, we identified two possible pathways contributing to the hypoxia pulmonary hypertension phenotype of extreme RVSP elevation: aberrant T cell expression driven by hypoxia-induced miRNAs and increased expression of C5a and C5AR1. We suggest that the PL/J mouse will be a good model for seeking mechanism(s) of RVSP elevation in hypoxia-induced PAH.
Collapse
Affiliation(s)
| | | | - Michael W Pauciulo
- 1 Division of Human Genetics and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Patricia A Pastura
- 3 Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - Timothy D Le Cras
- 3 Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - William C Nichols
- 1 Division of Human Genetics and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
25
|
Wang S, Cao W, Gao S, Nie X, Zheng X, Xing Y, Chen Y, Bao H, Zhu D. TUG1 Regulates Pulmonary Arterial Smooth Muscle Cell Proliferation in Pulmonary Arterial Hypertension. Can J Cardiol 2019; 35:1534-1545. [DOI: 10.1016/j.cjca.2019.07.630] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/07/2019] [Accepted: 07/16/2019] [Indexed: 01/07/2023] Open
|
26
|
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
|
27
|
Abdul-Salam VB, Russomanno G, Chien-Nien C, Mahomed AS, Yates LA, Wilkins MR, Zhao L, Gierula M, Dubois O, Schaeper U, Endruschat J, Wojciak-Stothard B. CLIC4/Arf6 Pathway. Circ Res 2019; 124:52-65. [PMID: 30582444 PMCID: PMC6325770 DOI: 10.1161/circresaha.118.313705] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RATIONALE Increased expression of CLIC4 (chloride intracellular channel 4) is a feature of endothelial dysfunction in pulmonary arterial hypertension, but its role in disease pathology is not fully understood. OBJECTIVE To identify CLIC4 effectors and evaluate strategies targeting CLIC4 signaling in pulmonary hypertension. METHODS AND RESULTS Proteomic analysis of CLIC4-interacting proteins in human pulmonary artery endothelial cells identified regulators of endosomal trafficking, including Arf6 (ADP ribosylation factor 6) GTPase activating proteins and clathrin, while CLIC4 overexpression affected protein regulators of vesicular trafficking, lysosomal function, and inflammation. CLIC4 reduced BMPRII (bone morphogenetic protein receptor II) expression and signaling as a result of Arf6-mediated reduction in gyrating clathrin and increased lysosomal targeting of the receptor. BMPRII expression was restored by Arf6 siRNA, Arf inhibitor Sec7 inhibitor H3 (SecinH3), and inhibitors of clathrin-mediated endocytosis but was unaffected by chloride channel inhibitor, indanyloxyacetic acid 94 or Arf1 siRNA. The effects of CLIC4 on NF-κB (nuclear factor-kappa B), HIF (hypoxia-inducible factor), and angiogenic response were prevented by Arf6 siRNA and SecinH3. Sugen/hypoxia mice and monocrotaline rats showed elevated expression of CLIC4, activation of Arf6 and NF-κB, and reduced expression of BMPRII in the lung. These changes were established early during disease development. Lung endothelium-targeted delivery of CLIC4 siRNA or treatment with SecinH3 attenuated the disease, reduced CLIC4/Arf activation, and restored BMPRII expression in the lung. Endothelial colony-forming cells from idiopathic pulmonary hypertensive patients showed upregulation of CLIC4 expression and Arf6 activity, suggesting potential importance of this pathway in the human condition. CONCLUSIONS Arf6 is a novel effector of CLIC4 and a new therapeutic target in pulmonary hypertension.
Collapse
Affiliation(s)
- Vahitha B Abdul-Salam
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Giusy Russomanno
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Chen Chien-Nien
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Abdul S Mahomed
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Luke A Yates
- Section of Structural Biology (L.A.Y.), Department of Medicine, Imperial College London, United Kingdom
| | - Martin R Wilkins
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Lan Zhao
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Magdalena Gierula
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Oliver Dubois
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| | - Ute Schaeper
- Silence Therapeutics GmbH, Berlin, Germany (U.S., J.E.)
| | | | - Beata Wojciak-Stothard
- From the Centre for Pharmacology and Therapeutics (V.B.A.-S., G.R., C.C.-N., A.S.M., M.R.W., L.Z., M.G., O.D., B.W.-S.), Department of Medicine, Imperial College London, United Kingdom
| |
Collapse
|
28
|
Tian W, Jiang X, Sung YK, Shuffle E, Wu TH, Kao PN, Tu AB, Dorfmüller P, Cao A, Wang L, Peng G, Kim Y, Zhang P, Chappell J, Pasupneti S, Dahms P, Maguire P, Chaib H, Zamanian R, Peters-Golden M, Snyder MP, Voelkel NF, Humbert M, Rabinovitch M, Nicolls MR. Phenotypically Silent Bone Morphogenetic Protein Receptor 2 Mutations Predispose Rats to Inflammation-Induced Pulmonary Arterial Hypertension by Enhancing the Risk for Neointimal Transformation. Circulation 2019; 140:1409-1425. [PMID: 31462075 DOI: 10.1161/circulationaha.119.040629] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bmpr2 (bone morphogenetic protein receptor 2) mutations are critical risk factors for hereditary pulmonary arterial hypertension (PAH) with approximately 20% of carriers developing disease. There is an unmet medical need to understand how environmental factors, such as inflammation, render Bmpr2 mutants susceptible to PAH. Overexpressing 5-LO (5-lipoxygenase) provokes lung inflammation and transient PAH in Bmpr2+/- mice. Accordingly, 5-LO and its metabolite, leukotriene B4, are candidates for the second hit. The purpose of this study was to determine how 5-LO-mediated pulmonary inflammation synergized with phenotypically silent Bmpr2 defects to elicit significant pulmonary vascular disease in rats. METHODS Monoallelic Bmpr2 mutant rats were generated and found phenotypically normal for up to 1 year of observation. To evaluate whether a second hit would elicit disease, animals were exposed to 5-LO-expressing adenovirus, monocrotaline, SU5416, SU5416 with chronic hypoxia, or chronic hypoxia alone. Bmpr2-mutant hereditary PAH patient samples were assessed for neointimal 5-LO expression. Pulmonary artery endothelial cells with impaired BMPR2 signaling were exposed to increased 5-LO-mediated inflammation and were assessed for phenotypic and transcriptomic changes. RESULTS Lung inflammation, induced by intratracheal delivery of 5-LO-expressing adenovirus, elicited severe PAH with intimal remodeling in Bmpr2+/- rats but not in their wild-type littermates. Neointimal lesions in the diseased Bmpr2+/- rats gained endogenous 5-LO expression associated with elevated leukotriene B4 biosynthesis. Bmpr2-mutant hereditary PAH patients similarly expressed 5-LO in the neointimal cells. In vitro, BMPR2 deficiency, compounded by 5-LO-mediated inflammation, generated apoptosis-resistant and proliferative pulmonary artery endothelial cells with mesenchymal characteristics. These transformed cells expressed nuclear envelope-localized 5-LO consistent with induced leukotriene B4 production, as well as a transcriptomic signature similar to clinical disease, including upregulated nuclear factor Kappa B subunit (NF-κB), interleukin-6, and transforming growth factor beta (TGF-β) signaling pathways. The reversal of PAH and vasculopathy in Bmpr2 mutants by TGF-β antagonism suggests that TGF-β is critical for neointimal transformation. CONCLUSIONS In a new 2-hit model of disease, lung inflammation induced severe PAH pathology in Bmpr2+/- rats. Endothelial transformation required the activation of canonical and noncanonical TGF-β signaling pathways and was characterized by 5-LO nuclear envelope translocation with enhanced leukotriene B4 production. This study offers an explanation of how an environmental injury unleashes the destructive potential of an otherwise silent genetic mutation.
Collapse
Affiliation(s)
- Wen Tian
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Xinguo Jiang
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Yon K Sung
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Eric Shuffle
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Ting-Hsuan Wu
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Peter N Kao
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Allen B Tu
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Peter Dorfmüller
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France (P.D., M.H.).,Institut National de la Sante Et de la Recherche Medicale UMR_S 999, Le Plessis-Robinson, France (P.D., M.H.).,Pathology Department, Hôpital Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.)
| | - Aiqin Cao
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Lingli Wang
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Gongyong Peng
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.).,State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (G.P.)
| | - Yesl Kim
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Patrick Zhang
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - James Chappell
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Shravani Pasupneti
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Petra Dahms
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Peter Maguire
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Hassan Chaib
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Roham Zamanian
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | | | - Michael P Snyder
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | | | - Marc Humbert
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France (P.D., M.H.).,Institut National de la Sante Et de la Recherche Medicale UMR_S 999, Le Plessis-Robinson, France (P.D., M.H.).,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Department Hospitalo-Universitaire Thorax Innovation, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France (M.H.)
| | - Marlene Rabinovitch
- Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| | - Mark R Nicolls
- Veterans Affairs Palo Alto Health Care System, CA (W.T, X.J., Y.K.S., E.S., A.B.T., G.P., Y.K., P.Z., S.P., P.D., M.R.N.).,Stanford University School of Medicine, CA (W.T., X.J., Y.K.S., E.S., T.H.W., P.N.K., A.B.T., A.C., L.W., G.P., Y.K., P.Z., J.C., S.P., P.D., P.M., H.C., R.Z., M.P.S., M.R., M.R.N.)
| |
Collapse
|
29
|
Li Q, Zhou X, Zhou X. Downregulation of miR‑98 contributes to hypoxic pulmonary hypertension by targeting ALK1. Mol Med Rep 2019; 20:2167-2176. [PMID: 31322216 PMCID: PMC6691262 DOI: 10.3892/mmr.2019.10482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/31/2019] [Indexed: 12/21/2022] Open
Abstract
Chronic hypoxia is one of the most common causes of secondary pulmonary hypertension, the mechanisms of which remain unclear. MicroRNAs (miRNAs) are small, noncoding RNAs that inhibit the translation or accelerate the degradation of mRNA. Previous studies have demonstrated that deregulated miRNA expression contributes to various cellular processes including cell apoptosis and proliferation, which are mediated by hypoxia. In the present study, the expression of miR‑98 was identified to be decreased in the lung tissue of a hypoxic pulmonary hypertension (HPH) rat model and pulmonary artery (PA) smooth muscle cells (PASMCs), which was induced by hypoxia. By transfecting miR‑98 mimics into PASMCs, the high expression of miR‑98 inhibited cell proliferation, but upregulated hypoxia‑induced PASMCs apoptosis. However, these effects of miR‑98 mimics on PASMCs were reversed by ALK1 (activin receptor‑like kinase‑1) overexpression. ALK1 was identified as a candidate target of miR‑98. In addition, overexpressing miR‑98 markedly decreased the pulmonary artery wall thickness and the right ventricular systolic pressure in rats induced by hypoxia. These results provided clear evidence that miR‑98 was a direct regulator of ALK1, and that the downregulation of miR‑98 contributed to the pathogenesis of HPH. These results provide a novel potential therapeutic strategy for the treatment of HPH.
Collapse
Affiliation(s)
- Qingling Li
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xincan Zhou
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xianghui Zhou
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
30
|
Di R, Yang Z, Xu P, Xu Y. Silencing PDK1 limits hypoxia-induced pulmonary arterial hypertension in mice via the Akt/p70S6K signaling pathway. Exp Ther Med 2019; 18:699-704. [PMID: 31281449 DOI: 10.3892/etm.2019.7627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 06/15/2018] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the effect of phosphoinositide-dependent protein kinase-1 (PDK1) on hypoxia-induced pulmonary arterial hypertension (PAH). A mouse model of hypoxia-induced PAH was generated using normal or PDK1-knockout mice. Histological analysis and hemodynamic evaluations were performed to identify the progression of PAH. The expression and phosphorylation of PDK1/protein kinase B (Akt) signaling pathway associated proteins were detected by western blot analysis. Increased lung vessel thickness, right ventricular (RV) systolic pressure (RVSP), RV hypertrophy index (RVHI) values [the RV weight-to-left ventricular (LV) plus septum (S) weight ratio] and PDK1 expression were observed in the hypoxia-induced PAH model compared with the normal control. The phosphorylation of AktT308, proline-rich Akt1 substrate 1 (PRAS40) and S6KT229 was also notably increased in the PAH model compared with the control. The changes of proteins were not observed in the hypoxia treated PDK1flox/+ : Tie2-Cre mice. Similarly, the RVSP and RVHI values, and PDK1 expression were reduced in the hypoxia treated PDK1flox/+: Tie2-Cre mice to a level comparable with those in the control, suggesting that PDK1 partial knockout significantly limited hypoxia-induced PAH. The results of the present study indicate that PDK1 is essential for hypoxia-induced PAH through the PDK1/Akt/S6K signaling cascades.
Collapse
Affiliation(s)
- Ruomin Di
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, P.R. China
| | - Peng Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yingjia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
31
|
West J, Chen X, Yan L, Gladson S, Loyd J, Rizwan H, Talati M. Adverse effects of BMPR2 suppression in macrophages in animal models of pulmonary hypertension. Pulm Circ 2019; 10:2045894019856483. [PMID: 31124398 PMCID: PMC7074495 DOI: 10.1177/2045894019856483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/17/2019] [Indexed: 01/11/2023] Open
Abstract
Inflammatory cells contribute to irreversible damage in pulmonary arterial hypertension (PAH). We hypothesized that in PAH, dysfunctional BMPR2 signaling in macrophages contributes to pulmonary vascular injury and phenotypic changes via proinflammatory cytokine production. Studies were conducted in: (1) Rosa26-rtTA2 3 X TetO7-Bmpr2delx4 FVB/N mice (mutant Bmpr2 is universally expressed, BMPR2delx4 mice) given a weekly intra-tracheal liposomal clodronate injections for four weeks; and (2) LysM-Cre X floxed BMPR2 X floxed eGFP monocyte lineage-specific BMPR2 knockout (KO) mouse model (Bmpr2 gene expression knockdown in monocytic lineage cells) (BMPR2KO) following three weeks of sugen/hypoxia treatment. In the BMPR2delx4 mice, increased right ventricular systolic pressure (RVSP; P < 0.05) was normalized by clodronate, and in monocyte lineage-specific BMPR2KO mice sugen hypoxia treatment increased (P < 0.05) RVSP compared to control littermates, suggesting that suppressed BMPR2 in macrophages modulate RVSP in animal models of PH. In addition, in these mouse models, muscularized pulmonary vessels were increased (P < 0.05) and surrounded by an increased number of macrophages. Elimination of macrophages in BMPR2delx4 mice reduced the number of muscularized pulmonary vessels and macrophages surrounding these vessels. Further, in monocyte lineage-specific BMPR2KO mice, there was significant increase in proinflammatory cytokines, including C-X-C Motif Chemokine Ligand 12 (CXCL12), complement component 5 a (C5a), Interleukin-16 (IL-16), and secretory ICAM. C5a positive inflammatory cells present in and around the pulmonary vessels in the PAH lung could potentially be involved in pulmonary vessel remodeling. In summary, our data indicate that, in BMPR2-related PAH, macrophages with dysfunctional BMPR2 influence pulmonary vascular remodeling and phenotypic outcomes via proinflammatory cytokine production.
Collapse
Affiliation(s)
- James West
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Xinping Chen
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ling Yan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Santhi Gladson
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James Loyd
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hamid Rizwan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Megha Talati
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
32
|
Nie X, Chen Y, Tan J, Dai Y, Mao W, Qin G, Ye S, Sun J, Yang Z, Chen J. MicroRNA-221-3p promotes pulmonary artery smooth muscle cells proliferation by targeting AXIN2 during pulmonary arterial hypertension. Vascul Pharmacol 2019; 116:24-35. [PMID: 28694128 DOI: 10.1016/j.vph.2017.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 05/13/2017] [Accepted: 07/06/2017] [Indexed: 12/23/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a pathological condition characterized by excessive cell proliferation and migration of pulmonary arterial smooth muscle cells (PASMC). PAH pathogenesis shares similarities with cancers such as excessive cell proliferation and apoptosis resistance. A previous study by our group revealed that decreased expression of a tumor suppressor-AXIN2 (Axis inhibition protein 2) was responsible for enhanced PASMC proliferation and suppressed apoptosis. Nevertheless, the mechanisms that regulate the downregulation of AXIN2 in PAH remain elusive. Data from the present study demonstrated that miR-221-3p acts as an upstream regulator of AXIN2 and functions to induce PASMC proliferation. We first showed that miR-221-3p expression was elevated in lung tissue and PASMC of PAH patients as well as in animal models of PAH. Human PASMC were transfected with a miR-221-3p mimic and miR-221-3p inhibitor, respectively, and their effects on the proliferation and migration was assessed using BrdU incorporation, PCNA staining and wound healing assays. In addition, we investigated the molecular mechanism through which miR-221-3p contributes to cell proliferation in PASMC and identified AXIN2 as a direct target gene of miR-221-3p by dual luciferase reporter gene assays, qRT-qPCR and western blotting. Furthermore, we found that ectopic expression of AXIN2 or pharmacological inhibition of β-catenin by XAV-939 can attenuate the effect of miR-221-3p on cell proliferation in PASMC. Moreover, intravenous injection of miR-221-3p inhibitor attenuated the progression of SU5416-hypoxia-induced PAH in rats. The results of the present study identified a new regulatory axis in which miR-221-3p and AXIN2 regulate the proliferation of PASMC.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adult
- Animals
- Axin Protein/genetics
- Axin Protein/metabolism
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Case-Control Studies
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Female
- Humans
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/prevention & control
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- Vascular Remodeling
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Xiaowei Nie
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China.
| | - Yuan Chen
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Jianxin Tan
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Youai Dai
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Wenjun Mao
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Guowei Qin
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Shugao Ye
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Jie Sun
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, PR China
| | - Zhenkun Yang
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, PR China
| | - Jingyu Chen
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China.
| |
Collapse
|
33
|
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: 313] [Impact Index Per Article: 52.2] [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
|
34
|
Kuebler WM, Nicolls MR, Olschewski A, Abe K, Rabinovitch M, Stewart D, Chan SY, Morrell NW, Archer SL, Spiekerkoetter E. A pro-con debate: current controversies in PAH pathogenesis at the American Thoracic Society International Conference in 2017. Am J Physiol Lung Cell Mol Physiol 2018; 315:L502-L516. [PMID: 29877097 PMCID: PMC6230875 DOI: 10.1152/ajplung.00150.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/22/2018] [Accepted: 06/02/2018] [Indexed: 12/16/2022] Open
Abstract
The following review summarizes the pro-con debate about current controversies regarding the pathogenesis of pulmonary arterial hypertension (PAH) that took place at the American Thoracic Society Conference in May 2017. Leaders in the field of PAH research discussed the importance of the immune system, the role of hemodynamic stress and endothelial apoptosis, as well as bone morphogenetic protein receptor-2 signaling in PAH pathogenesis. Whereas this summary does not intend to resolve obvious conflicts in opinion, we hope that the presented arguments entice further discussions and draw a new generation of enthusiastic researchers into this vibrant field of science to bridge existing gaps for a better understanding and therapy of this fatal disease.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitaetsmedizin Berlin, Berlin , Germany
- Keenan Research Centre for Biomedical Science at Saint Michael's , Toronto, Ontario , Canada
- Department of Surgery, University of Toronto , Toronto, Ontario , Canada
- Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care, Department of Medicine, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University , Stanford, California
| | - Andrea Olschewski
- Ludwig Boltzmann Institute, Lung Vascular Research, Medical University of Graz , Graz , Austria
- Johannes Kepler University Linz, Medicine Rectorate, Linz, Austria
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Marlene Rabinovitch
- Division of Cardiology, Department of Pediatrics, Stanford University School of Medicine , Stanford, California
| | - Duncan Stewart
- Division of Cardiology, Department of Medicine, Ottawa Hospital Research Institute , Ottawa, Ontario , Canada
| | - Stephen Y Chan
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Nicholas W Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge , Cambridge , United Kingdom
| | - Stephen L Archer
- Department of Medicine, Queen's University , Kingston, Ontario , Canada
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care, Department of Medicine, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University , Stanford, California
| |
Collapse
|
35
|
Tamura Y, Phan C, Tu L, Le Hiress M, Thuillet R, Jutant EM, Fadel E, Savale L, Huertas A, Humbert M, Guignabert C. Ectopic upregulation of membrane-bound IL6R drives vascular remodeling in pulmonary arterial hypertension. J Clin Invest 2018; 128:1956-1970. [PMID: 29629897 DOI: 10.1172/jci96462] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by a progressive accumulation of pulmonary artery smooth muscle cells (PA-SMCs) in pulmonary arterioles leading to the narrowing of the lumen, right heart failure, and death. Although most studies have supported the notion of a role for IL-6/glycoprotein 130 (gp130) signaling in PAH, it remains unclear how this signaling pathway determines the progression of the disease. Here, we identify ectopic upregulation of membrane-bound IL-6 receptor (IL6R) on PA-SMCs in PAH patients and in rodent models of pulmonary hypertension (PH) and demonstrate its key role for PA-SMC accumulation in vitro and in vivo. Using Sm22a-Cre Il6rfl/fl, which lack Il6r in SM22A-expressing cells, we found that these animals are protected against chronic hypoxia-induced PH with reduced PA-SMC accumulation, revealing the potent pro-survival potential of membrane-bound IL6R. Moreover, we determine that treatment with IL6R-specific antagonist reverses experimental PH in two rat models. This therapeutic strategy holds promise for future clinical studies in PAH.
Collapse
|
36
|
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
|
37
|
Goumans MJ, Ten Dijke P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022210. [PMID: 28348036 DOI: 10.1101/cshperspect.a022210] [Citation(s) in RCA: 247] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic studies in animals and humans indicate that gene mutations that functionally perturb transforming growth factor β (TGF-β) signaling are linked to specific hereditary vascular syndromes, including Osler-Rendu-Weber disease or hereditary hemorrhagic telangiectasia and Marfan syndrome. Disturbed TGF-β signaling can also cause nonhereditary disorders like atherosclerosis and cardiac fibrosis. Accordingly, cell culture studies using endothelial cells or smooth muscle cells (SMCs), cultured alone or together in two- or three-dimensional cell culture assays, on plastic or embedded in matrix, have shown that TGF-β has a pivotal effect on endothelial and SMC proliferation, differentiation, migration, tube formation, and sprouting. Moreover, TGF-β can stimulate endothelial-to-mesenchymal transition, a process shown to be of key importance in heart valve cushion formation and in various pathological vascular processes. Here, we discuss the roles of TGF-β in vasculogenesis, angiogenesis, and lymphangiogenesis and the deregulation of TGF-β signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
38
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
39
|
Miyazaki S, Ikeda T, Ito G, Inoue M, Nara K, Nishinaga Y, Hasegawa Y. Pulmonary tumor thrombotic microangiopathy successfully treated with corticosteroids: a case report. J Med Case Rep 2017; 11:356. [PMID: 29273073 PMCID: PMC5741930 DOI: 10.1186/s13256-017-1524-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/21/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pulmonary tumor thrombotic microangiopathy is a special type of tumor thromboembolism. We report the case of a patient who developed pulmonary tumor thrombotic microangiopathy with alveolar hemorrhage. Almost all patients with pulmonary tumor thrombotic microangiopathy die within 1 week of the onset of dyspnea; however, the prognosis in this case was better, with 10 weeks of survival from presentation. CASE PRESENTATION A 62-year-old Japanese man was referred to our hospital with a 4-week history of dyspnea on exertion and severe pulmonary hypertension. Five years previously, he had undergone distal gastrectomy for gastric cancer. He was afebrile, normotensive, and hypoxemic. A physical examination was unremarkable except for purpura on his upper extremities and trunk. Blood tests showed anemia and disseminated intravascular coagulation. Chest computed tomography revealed diffuse ground-glass opacities with emphysema in his upper lungs, moderate pleural effusions, mediastinal lymphadenopathy, and enlargement of the right ventricle and main pulmonary artery. A computed tomography pulmonary angiogram showed no evidence of pulmonary embolism. Lung perfusion scintigraphy showed multiple segmental defects. Although recurrence of gastric cancer was confirmed from the results of bone marrow biopsy, bronchoscopy was not performed due to bleeding diathesis. He was treated with corticosteroids, antibiotics, and platelet transfusion, following which resolution of the abnormal lung shadows and right ventricular pressure overload along with partial alleviation of respiratory failure was observed. Because of his poor performance status, he was eventually transited to palliative care and died 6 weeks after admission. Necropsy of the lung confirmed the diagnosis of pulmonary tumor thrombotic microangiopathy with alveolar hemorrhage. CONCLUSIONS Pulmonary tumor thrombotic microangiopathy should be considered in the differential diagnosis of patients with cancer who present with severe pulmonary hypertension. In pulmonary tumor thrombotic microangiopathy, local inflammation in pulmonary microvasculature may contribute to pulmonary hypertension, and regulation of inflammation using corticosteroids may help improve the prognosis.
Collapse
Affiliation(s)
- Shinichi Miyazaki
- Department of Respiratory Medicine, Yokkaichi Municipal Hospital, Sibata2-2-37, Shibata, Yokkaichi-shi, Mie, 510-0822, Japan.
| | - Takuya Ikeda
- Department of Respiratory Medicine, Yokkaichi Municipal Hospital, Sibata2-2-37, Shibata, Yokkaichi-shi, Mie, 510-0822, Japan
| | - Genshi Ito
- Department of Respiratory Medicine, Yokkaichi Municipal Hospital, Sibata2-2-37, Shibata, Yokkaichi-shi, Mie, 510-0822, Japan
| | - Masahide Inoue
- Department of Respiratory Medicine, Yokkaichi Municipal Hospital, Sibata2-2-37, Shibata, Yokkaichi-shi, Mie, 510-0822, Japan
| | - Keiji Nara
- Department of Pathology, Yokkaichi Municipal Hospital, Sibata2-2-37, Shibata, Yokkaichi-shi, Mie, 510-0822, Japan
| | - Yuko Nishinaga
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan
| |
Collapse
|
40
|
Hemnes AR, Humbert M. Pathobiology of pulmonary arterial hypertension: understanding the roads less travelled. Eur Respir Rev 2017; 26:26/146/170093. [DOI: 10.1183/16000617.0093-2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/08/2017] [Indexed: 01/09/2023] Open
Abstract
The pathobiology of pulmonary arterial hypertension (PAH) is complex and incompletely understood. Although three pathogenic pathways have been relatively well characterised, it is widely accepted that dysfunction in a multitude of other cellular processes is likely to play a critical role in driving the development of PAH. Currently available therapies, which all target one of the three well-characterised pathways, provide significant benefits for patients; however, PAH remains a progressive and ultimately fatal disease. The development of drugs to target alternative pathogenic pathways is, therefore, an attractive proposition and one that may complement existing treatment regimens to improve outcomes for patients. Considerable research has been undertaken to identify the role of the less well-understood pathways and in this review we will highlight some of the key discoveries and the potential for utility as therapeutic targets.
Collapse
|
41
|
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
|
42
|
Abstract
Pulmonary arterial hypertension (PAH) remains a mysterious killer that, like cancer, is characterized by tremendous complexity. PAH development occurs under sustained and persistent environmental stress, such as inflammation, shear stress, pseudo-hypoxia, and more. After inducing an initial death of the endothelial cells, these environmental stresses contribute with time to the development of hyper-proliferative and apoptotic resistant clone of cells including pulmonary artery smooth muscle cells, fibroblasts, and even pulmonary artery endothelial cells allowing vascular remodeling and PAH development. Molecularly, these cells exhibit many features common to cancer cells offering the opportunity to exploit therapeutic strategies used in cancer to treat PAH. In this review, we outline the signaling pathways and mechanisms described in cancer that drive PAH cells' survival and proliferation and discuss the therapeutic potential of antineoplastic drugs in PAH.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Department of Medicine, Québec, Canada
| | - Geraldine Vitry
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Department of Medicine, Québec, Canada
| | - Isabelle Trinh
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Department of Medicine, Québec, Canada
| | - Roxane Paulin
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Department of Medicine, Québec, Canada
| | - Steeve Provencher
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Department of Medicine, Québec, Canada
| | - Sebastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Department of Medicine, Québec, Canada
| |
Collapse
|
43
|
Rothman A, Wiencek RG, Davidson S, Evans WN, Restrepo H, Sarukhanov V, Mann D. Challenges in the development of chronic pulmonary hypertension models in large animals. Pulm Circ 2017; 7:156-166. [PMID: 28680575 PMCID: PMC5448539 DOI: 10.1086/690099] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/07/2016] [Indexed: 01/17/2023] Open
Abstract
Pulmonary hypertension (PH) results in significant morbidity and mortality. Chronic PH animal models may advance the study of PH's mechanisms, evolution, and therapy. In this report, we describe the challenges and successes in developing three models of chronic PH in large animals: two models (one canine and one swine) utilized repeated infusions of ceramic microspheres into the pulmonary vascular bed, and the third model employed a surgical aorto-pulmonary shunt. In the canine model, seven dogs underwent microsphere infusions that resulted in progressive elevation of pulmonary arterial pressure over a few months. In this model, pulmonary endoarterial tissue was obtained for histology. In the aorto-pulmonary shunt swine model, 17 pigs developed systemic level pulmonary pressures after 2-3 months. In this model, pulmonary endoarterial tissue was sequentially obtained to assess for changes in gene and microRNA expression. In the swine microsphere infusion model, three pigs developed only a modest chronic increase in pulmonary arterial pressure, despite repeated infusions of microspheres (up to 40 in one animal). The main purpose of this model was for vasodilator testing, which was performed successfully immediately after acute microsphere infusions. Chronic PH in large animal models can be successfully created; however, a model's characteristics need to match the investigational goals.
Collapse
Affiliation(s)
- Abraham Rothman
- Children's Heart Center Nevada, Las Vegas, NV, USA.,University of Nevada, School of Medicine, Department of Pediatrics, Las Vegas, NV, USA
| | - Robert G Wiencek
- Stanford University, Department of Cardiothoracic Surgery, Cardiothoracic Dignity Healthcare, Las Vegas, NV, USA
| | | | - William N Evans
- Children's Heart Center Nevada, Las Vegas, NV, USA.,University of Nevada, School of Medicine, Department of Pediatrics, Las Vegas, NV, USA
| | - Humberto Restrepo
- Children's Heart Center Nevada, Las Vegas, NV, USA.,University of Nevada, School of Medicine, Department of Pediatrics, Las Vegas, NV, USA
| | | | | |
Collapse
|
44
|
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
|
45
|
TNFα drives pulmonary arterial hypertension by suppressing the BMP type-II receptor and altering NOTCH signalling. Nat Commun 2017; 8:14079. [PMID: 28084316 PMCID: PMC5241886 DOI: 10.1038/ncomms14079] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023] Open
Abstract
Heterozygous germ-line mutations in the bone morphogenetic protein type-II receptor (BMPR-II) gene underlie heritable pulmonary arterial hypertension (HPAH). Although inflammation promotes PAH, the mechanisms by which inflammation and BMPR-II dysfunction conspire to cause disease remain unknown. Here we identify that tumour necrosis factor-α (TNFα) selectively reduces BMPR-II transcription and mediates post-translational BMPR-II cleavage via the sheddases, ADAM10 and ADAM17 in pulmonary artery smooth muscle cells (PASMCs). TNFα-mediated suppression of BMPR-II subverts BMP signalling, leading to BMP6-mediated PASMC proliferation via preferential activation of an ALK2/ACTR-IIA signalling axis. Furthermore, TNFα, via SRC family kinases, increases pro-proliferative NOTCH2 signalling in HPAH PASMCs with reduced BMPR-II expression. We confirm this signalling switch in rodent models of PAH and demonstrate that anti-TNFα immunotherapy reverses disease progression, restoring normal BMP/NOTCH signalling. Collectively, these findings identify mechanisms by which BMP and TNFα signalling contribute to disease, and suggest a tractable approach for therapeutic intervention in PAH. Reduced BMP receptor II signalling underlies pulmonary arterial hypertension (PAH). Here, Hurst et al. show that TNFα subverts BMP signalling by increasing BMP6 expression and signalling via an alternative BMP receptor, ALK2, in pulmonary artery smooth muscle cells to drive abnormal proliferation and PAH.
Collapse
|
46
|
Guignabert C, Bailly S, Humbert M. Restoring BMPRII functions in pulmonary arterial hypertension: opportunities, challenges and limitations. Expert Opin Ther Targets 2016; 21:181-190. [DOI: 10.1080/14728222.2017.1275567] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Univ. Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Sabine Bailly
- INSERM U1036, Grenoble, France
- Laboratoire Biologie du Cancer et de l’Infection, Commissariat à l’Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
- Université Grenoble-Alpes, Grenoble, France
| | - Marc Humbert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Univ. Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France
| |
Collapse
|
47
|
Nie X, Tan J, Dai Y, Mao W, Chen Y, Qin G, Li G, Shen C, Zhao J, Chen J. Nur77 downregulation triggers pulmonary artery smooth muscle cell proliferation and migration in mice with hypoxic pulmonary hypertension via the Axin2-β-catenin signaling pathway. Vascul Pharmacol 2016; 87:230-241. [PMID: 27871853 DOI: 10.1016/j.vph.2016.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 11/03/2016] [Indexed: 01/15/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by remodeling of the pulmonary vasculature, including marked proliferation and reduced apoptosis of pulmonary artery smooth muscle cells (PASMCs). Members of the nuclear receptor 4A (NR4A) subfamily are involved in a variety of biological events, such as cell apoptosis, proliferation, inflammation, and metabolism. Activation of Nur77 (an orphan nuclear receptor that belongs to NR4A subfamily) has recently been reported to be as a beneficial agent in the treatment of cardiovascular and metabolic diseases. In the present study, we investigated the effects of NR4A on human PASMCs function in vitro and determined the underlying mechanisms. We found a robust expression of NR4A receptors in lung tissues of PAH patients and hypoxic mice but a highly significant downregulation within pulmonary arteries (PAs) as assessed by quantitative polymerase chain reaction, immunoblotting, and immunohistochemistry. In vitro, NR4A receptors were found significantly decreased in PASMCs derived from PAH patients. To explore the pathological effects of decreased Nur77 in PASMCs, PASMCs were transduced with siRNA against Nur77. The siRNA-mediated knockdown of Nur77 significantly augmented PASMCs proliferation and migration. In contrast, Nur77 overexpression prevented PASMCs from proliferation and migration. Mechanistically, overexpression of Axis inhibition protein 2 (Axin2) or inhibition of β-catenin signaling was shown to be responsible for Nur77 knockdown-induced proliferation of PASMCs. Following hypoxia-induced angiogenesis of the pulmonary artery in C57BL/6 mice, small-molecule Nur77 agonists-Octaketide Cytosporone B (Csn-B) can significantly decreased thickness of vascular wall and markedly attenuated the development of chronic hypoxia-induced PAH in vivo. Therefore, reconstitution of Nur77 levels represents a promising therapeutic option to prevent vascular remodeling processes.
Collapse
Affiliation(s)
- Xiaowei Nie
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China.
| | - Jianxin Tan
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Youai Dai
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Wenjun Mao
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Yuan Chen
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Guowei Qin
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Guirong Li
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Chenyou Shen
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Jingjing Zhao
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Jingyu Chen
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China.
| |
Collapse
|
48
|
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
|
49
|
Ghigna MR, Guignabert C, Montani D, Girerd B, Jaïs X, Savale L, Hervé P, Thomas de Montpréville V, Mercier O, Sitbon O, Soubrier F, Fadel E, Simonneau G, Humbert M, Dorfmüller P. BMPR2 mutation status influences bronchial vascular changes in pulmonary arterial hypertension. Eur Respir J 2016; 48:1668-1681. [PMID: 27811071 DOI: 10.1183/13993003.00464-2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022]
Abstract
The impact of bone morphogenetic protein receptor 2 (BMPR2) gene mutations on vascular remodelling in pulmonary arterial hypertension (PAH) is unknown. We sought to identify a histological profile of BMPR2 mutation carriers.Clinical data and lung histology from 44 PAH patients were subjected to systematic analysis and morphometry.Bronchial artery hypertrophy/dilatation and bronchial angiogenesis, as well as muscular remodelling of septal veins were significantly increased in PAH lungs carrying BMPR2 mutations. We found that patients displaying increased bronchial artery remodelling and bronchial microvessel density, irrespective of the mutation status, were more likely to suffer from severe haemoptysis. History of substantial haemoptysis (>50 mL) was significantly more frequent in BMPR2 mutation carriers. 43.5% of BMPR2 mutation carriers, as opposed to 9.5% of noncarriers, displayed singular large fibrovascular lesions, which appear to be closely related to the systemic lung vasculature.Our analysis provides evidence for the involvement of the pulmonary systemic circulation in BMPR2 mutation-related PAH. We show that BMPR2 mutation carriers are more prone to haemoptysis and that haemoptysis is closely correlated to bronchial arterial remodelling and angiogenesis; in turn, pronounced changes in the systemic vasculature correlate with increased pulmonary venous remodelling, creating a distinctive profile in PAH patients harbouring a BMPR2 mutation.
Collapse
Affiliation(s)
- Maria-Rosa Ghigna
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Dept of Pathology, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Christophe Guignabert
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - David Montani
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Barbara Girerd
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Xavier Jaïs
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Laurent Savale
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Philippe Hervé
- Dept of Thoracic and Vascular Surgery, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | | | - Olaf Mercier
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Dept of Thoracic and Vascular Surgery, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Olivier Sitbon
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Florent Soubrier
- AP-HP, Dept of Genetics, Pitié-Salpétrière Hospital, Université Pierre et Marie Curie, Paris, France
| | - Elie Fadel
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Dept of Thoracic and Vascular Surgery, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Gérald Simonneau
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France.,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,AP-HP, Dept of Pulmonology, DHU Thorax Innovation, Bicêtre Hospital, Kremlin-Bicêtre, France
| | - Peter Dorfmüller
- INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France .,School of Medicine, Paris South University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Dept of Pathology, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| |
Collapse
|
50
|
Barnes JW, Kucera ET, Tian L, Mellor NE, Dvorina N, Baldwin WW, Aldred MA, Farver CF, Comhair SAA, Aytekin M, Dweik RA. Bone Morphogenic Protein Type 2 Receptor Mutation-Independent Mechanisms of Disrupted Bone Morphogenetic Protein Signaling in Idiopathic Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2016; 55:564-575. [PMID: 27187737 DOI: 10.1165/rcmb.2015-0402oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Altered bone morphogenic protein (BMP) signaling, independent of BMPR2 mutations, can result in idiopathic pulmonary arterial hypertension (IPAH). Glucose dysregulation can regulate multiple processes in IPAH. However, the role of glucose in BMP antagonist expression in IPAH has not been characterized. We hypothesized that glucose uptake regulates BMP signaling through stimulation of BMP antagonist expression in IPAH. Using human plasma, lung tissue, and primary pulmonary arterial smooth muscle cells (PASMCs), we examined the protein expression of BMP2, BMP-regulated Smads, and Smurf-1 in patients with IPAH and control subjects. Gremlin-1 levels were elevated in patients with IPAH compared with control subjects, whereas expression of BMP2 was not different. We demonstrate increased Smad polyubiquitination in IPAH lung tissue and PASMCs that was further enhanced with proteasomal inhibition. Examination of the Smad ubiquitin-ligase, Smurf-1, showed increased protein expression in IPAH lung tissue and localization in the smooth muscle of the pulmonary artery. Glucose dose dependently increased Smurf-1 protein expression in control PASMCs, whereas Smurf-1 in IPAH PASMCs was increased and sustained. Conversely, phospho-Smad1/5/8 levels were reduced in IPAH compared with control PASMCs at physiological glucose concentrations. Interestingly, high glucose concentrations decreased phosphorylation of Smad1/5/8 in control PASMCs. Blocking glucose uptake had opposing effects in IPAH PASMCs, and inhibition of Smurf-1 activity resulted in partial rescue of Smad1/5/8 activation and cell migration rates. Collectively, these data suggest that BMP signaling can be regulated through BMPR2 mutation-independent mechanisms. Gremlin-1 (synonym: induced-in-high-glucose-2 protein) and Smurf-1 may function to inhibit BMP signaling as a consequence of the glucose dysregulation described in IPAH.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Micheala A Aldred
- 3 Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio; and
| | | | | | - Metin Aytekin
- Departments of 1 Pathobiology and.,5 Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Raed A Dweik
- Departments of 1 Pathobiology and.,6 Pulmonary and Critical Care Medicine, Respiratory Institute
| |
Collapse
|