1
|
Ghofrani HA, Gomberg-Maitland M, Zhao L, Grimminger F. Mechanisms and treatment of pulmonary arterial hypertension. Nat Rev Cardiol 2025; 22:105-120. [PMID: 39112561 DOI: 10.1038/s41569-024-01064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 11/28/2024]
Abstract
Substantial progress has been made in the management of pulmonary arterial hypertension (PAH) in the past 25 years, but the disease remains life-limiting. Established therapies for PAH are mostly limited to symptomatic relief by correcting the imbalance of vasoactive factors. The tyrosine kinase inhibitor imatinib, the first predominantly non-vasodilatory drug to be tested in patients with PAH, improved exercise capacity and pulmonary haemodynamics compared with placebo but at the expense of adverse events such as subdural haematoma. Given that administration by inhalation might reduce the risk of systemic adverse effects, inhaled formulations of tyrosine kinase inhibitors are currently in clinical development. Other novel therapeutic approaches for PAH include suppression of activin receptor type IIA signalling with sotatercept, which has shown substantial efficacy in clinical trials and was approved for use in the USA in 2024, but the long-term safety of the drug remains unclear. Future advances in the management of PAH will focus on right ventricular function and involve deep phenotyping and the development of a personalized medicine approach. In this Review, we summarize the mechanisms underlying PAH, provide an overview of available PAH therapies and their limitations, describe the development of newer, predominantly non-vasodilatory drugs that are currently being tested in phase II or III clinical trials, and discuss future directions for PAH research.
Collapse
Affiliation(s)
- Hossein-Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| | - Mardi Gomberg-Maitland
- George Washington University School of Medicine and Health Sciences, Department of Medicine, Washington, DC, USA
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Friedrich Grimminger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
2
|
Ge Q, Zhang T, Yu J, Lu X, Xiao S, Zhang T, Qing T, Xiao Z, Zeng L, Luo L. A new perspective on targeting pulmonary arterial hypertension: Programmed cell death pathways (Autophagy, Pyroptosis, Ferroptosis). Biomed Pharmacother 2024; 181:117706. [PMID: 39581144 DOI: 10.1016/j.biopha.2024.117706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiovascular disease characterized by elevated pulmonary vascular resistance, progressive increases in pulmonary artery pressures, ultimately leading to right-sided heart failure, and potentially mortality. Pulmonary vascular remodeling is pivotal in PAH onset and progression. While targeted drug therapies have notably ameliorated PAH prognosis, current medications primarily focus on vascular vasodilation, with limited ability to reverse pulmonary vascular remodeling fundamentally, resulting in suboptimal patient prognoses. Cellular death in pulmonary vasculature, once thought to be confined to apoptosis and necrosis, has evolved with the identification of pyroptosis, autophagy, and ferroptosis, revealing their association with vascular injury in PAH. These novel forms of regulated cellular death impact reactive oxygen species (ROS) generation, calcium stress, and inflammatory cascades, leading to pulmonary vascular cell loss, exacerbating vascular injury, and mediating adverse remodeling, inflammation, immune anomalies, and current emerging mechanisms (such as endothelial-mesenchymal transition, abnormal energy metabolism, and epigenetic regulation) in the pathogenesis of PAH. This review comprehensively delineates the roles of autophagy, pyroptosis, and ferroptosis in PAH, elucidating recent advances in their involvement and regulation of vascular injury. It juxtaposes their distinct functions in PAH and discusses the interplay of these programmed cell deaths in pulmonary vascular injury, highlighting the benefits of combined targeted therapies in mitigating pulmonary arterial hypertension-induced vascular injury, providing novel insights into targeted treatments for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Jiangbiao Yu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Xuelin Lu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Sijie Xiao
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Ting Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tao Qing
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China.
| |
Collapse
|
3
|
Liang X, Zhou J, Wang H, Zhang Z, Yin M, Zhu Y, Li L, Chen C, Wei M, Hu M, Zhao C, Yao J, Li G, Dinh‐Xuan A, Xiao J, Bei Y. miR-30d Attenuates Pulmonary Arterial Hypertension via Targeting MTDH and PDE5A and Modulates the Beneficial Effect of Sildenafil. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407712. [PMID: 39206778 PMCID: PMC11516105 DOI: 10.1002/advs.202407712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary arterial hypertension (PAH) is associated with aberrant pulmonary vascular smooth muscle cell (PASMC) function and vascular remodeling. MiR-30d plays an important role in the pathogenesis of several cardiovascular disorders. However, the function of miR-30d in PAH progression remained unknown. Our study shows that circulating miR-30d level is significantly reduced in the plasma from PAH patients. In miR-30d transgenic (TG) rats, overexpressing miR-30d attenuates monocrotaline (MCT)-induced pulmonary hypertension (PH) and pulmonary vascular remodeling. Increasing miR-30d also inhibits platelet-derived growth factor-bb (PDGF-bb)-induced proliferation and migration of human PASMC. Metadherin (MTDH) and phosphodiesterase 5A (PDE5A) are identified as direct target genes of miR-30d. Meanwhile, nuclear respiratory factor 1 (NRF1) acts as a positive upstream regulator of miR-30d. Using miR-30d knockout (KO) rats treated with sildenafil, a PDE5A inhibitor that is used in clinical PAH therapies, it is further found that suppressing miR-30d partially attenuates the beneficial effect of sildenafil against MCT-induced PH and vascular remodeling. The present study shows a protective effect of miR-30d against PAH and pulmonary vascular remodeling through targeting MTDH and PDE5A and reveals that miR-30d modulates the beneficial effect of sildenafil in treating PAH. MiR-30d should be a prospective target to treat PAH and pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Xuchun Liang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Jingwen Zhou
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Hongyun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Ziyi Zhang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Mingming Yin
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yujiao Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Lin Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Chen Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Meng Wei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Meiyu Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Cuimei Zhao
- Department of CardiologyShanghai Tongji HospitalTongji University School of MedicineShanghai200065China
| | - Jianhua Yao
- Department of CardiologyTenth People's HospitalSchool of MedicineTongji UniversityShanghai200090China
- Department of CardiologyShigatse People's HospitalTibet857000China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Anh‐Tuan Dinh‐Xuan
- Lung Function & Respiratory Physiology UnitsDepartment of Respiratory Physiology and Sleep MedicineCochin & George Pompidou HospitalsAssistance Publique‐Hôpitaux de Paris (APHP) CentreUniversity Paris CitéParis75014France
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yihua Bei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life ScienceShanghai UniversityNantong226011China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education)Shanghai UniversityShanghai200444China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| |
Collapse
|
4
|
Selvakumar SC, Preethi K A, Sekar D. MicroRNA-510-3p regulated vascular dysfunction in Preeclampsia by targeting Vascular Endothelial Growth Factor A (VEGFA) and its signaling axis. Placenta 2024; 153:31-52. [PMID: 38820941 DOI: 10.1016/j.placenta.2024.05.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/11/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is a pregnancy complication associated with multi-organ damage and vascular dysfunction. Meanwhile, microRNAs or miRNAs are crucial regulators of gene expression in various diseases including PE. Our previous studies reported high expression of miR-510 in the PE patients' blood compared to normal. Hence, we hypothesize that miR-510-3p targets Vascular endothelial growth factor A (VEGFA) in the regulation of PI3K/AKT/eNOS/mTOR axis in PE and miR-510-3p could be a potential therapeutic target for PE. METHODS The proliferation, migration, and apoptosis of HTR8/SVNeo and BeWo cells were analyzed by manipulating the miR-510-3p and VEGFA expression. Similarly, the inhibition of miR-510-3p through anti-miR-510-3p was analyzed in PE rat models, and the biochemical, hemodynamic parameters, and histopathology were examined between the groups. Moreover, the expression of miR-510-3p and VEGFA/PI3K/AKT/eNOS/mTOR axis was analyzed using qRT-PCR and Western blot. RESULTS Significant changes were observed in the BP, proteinuria, and other biochemical parameters between PE and control rats. Our results suggest that miR-510-3p targets VEGFA leading to vascular dysfunction in PE, while treatment with anti-miR-510-3p in the PE-induced rat model exhibits a significant change in the expression of miR-510-3p/VEGFA/PI3K/AKT/eNOS/mTOR signaling where miR-510-3p showed lesser expression and vice versa with VEGFA. The gene and protein expression analysis revealed a significant correlation between miR-510-3p and the VEGFA signaling axis in PE. DISCUSSION Thus, our findings from in vitro and in vivo suggest miR-510-3p as a potential therapeutic target and anti-miR-510-3p as a novel therapeutic molecule for PE.
Collapse
Affiliation(s)
- Sushmaa Chandralekha Selvakumar
- RNA Biology Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Auxzilia Preethi K
- RNA Biology Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Durairaj Sekar
- RNA Biology Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| |
Collapse
|
5
|
Ribeuz HL, Willer ASM, Chevalier B, Sancho M, Masson B, Eyries M, Jung V, Guerrera IC, Dutheil M, Jekmek KE, Laubry L, Carpentier G, Perez-Vizcaino F, Tu L, Guignabert C, Chaumais MC, Péchoux C, Humbert M, Hinzpeter A, Mercier O, Capuano V, Montani D, Antigny F. Role of KCNK3 Dysfunction in Dasatinib-associated Pulmonary Arterial Hypertension and Endothelial Cell Dysfunction. Am J Respir Cell Mol Biol 2024; 71:95-109. [PMID: 38546978 DOI: 10.1165/rcmb.2023-0185oc] [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: 05/23/2023] [Accepted: 03/27/2024] [Indexed: 07/02/2024] Open
Abstract
Pulmonary arterial (PA) hypertension (PAH) is a severe cardiopulmonary disease that may be triggered by exposure to drugs such as dasatinib or facilitated by genetic predispositions. The incidence of dasatinib-associated PAH is estimated at 0.45%, suggesting individual predispositions. The mechanisms of dasatinib-associated PAH are still incomplete. We discovered a KCNK3 gene (Potassium channel subfamily K member 3; coding for outward K+ channel) variant in a patient with dasatinib-associated PAH and investigated the impact of this variant on KCNK3 function. Additionally, we assessed the effects of dasatinib exposure on KCNK3 expression. In control human PA smooth muscle cells (hPASMCs) and human pulmonary endothelial cells (hPECs), we evaluated the consequences of KCNK3 knockdown on cell migration, mitochondrial membrane potential, ATP production, and in vitro tube formation. Using mass spectrometry, we determined the KCNK3 interactome. Patch-clamp experiments revealed that the KCNK3 variant represents a loss-of-function variant. Dasatinib contributed to PA constriction by decreasing KCNK3 function and expression. In control hPASMCs, KCNK3 knockdown promotes mitochondrial membrane depolarization and glycolytic shift. Dasatinib exposure or KCNK3 knockdown reduced the number of caveolae in hPECs. Moreover, KCNK3 knockdown in control hPECs reduced migration, proliferation, and in vitro tubulogenesis. Using proximity labeling and mass spectrometry, we identified the KCNK3 interactome, revealing that KCNK3 interacts with various proteins across different cellular compartments. We identified a novel pathogenic variant in KCNK3 and showed that dasatinib downregulates KCNK3, emphasizing the relationship between dasatinib-associated PAH and KCNK3 dysfunction. We demonstrated that a loss of KCNK3-dependent signaling contributes to endothelial dysfunction in PAH and glycolytic switch of hPASMCs.
Collapse
Affiliation(s)
- Hélène Le Ribeuz
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Anaïs Saint-Martin Willer
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Benoit Chevalier
- Paris Cité University, CNRS, INSERM, Institut Necker Enfants Malades-INEM, Paris, France
| | - Maria Sancho
- Department of Physiology and
- Department of Pharmacology, University of Vermont, Burlington, Vermont
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Bastien Masson
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Mélanie Eyries
- Genetics Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vincent Jung
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, Université Paris Cité-Federative Research Structure Necker, Paris, France
| | - Ida Chiara Guerrera
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, Université Paris Cité-Federative Research Structure Necker, Paris, France
| | - Mary Dutheil
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Kristelle El Jekmek
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Loann Laubry
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Gilles Carpentier
- Gly-CRRET Research Unit 4397, Paris-Est Créteil University, Créteil, France
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ly Tu
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Christophe Guignabert
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Marie-Camille Chaumais
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Paris-Saclay University, Faculty of Pharmacy, Orsay, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Christine Péchoux
- Paris-Saclay University, INRAE, AgroparisTech, GABI, Jouy-en-Josas, France
| | - Marc Humbert
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Alexandre Hinzpeter
- Paris Cité University, CNRS, INSERM, Institut Necker Enfants Malades-INEM, Paris, France
| | - Olaf Mercier
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Groupe Hospitalier Paris Saint-Joseph-Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Véronique Capuano
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - David Montani
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| |
Collapse
|
6
|
Feng Y, Yu Z, Tang M, Li J, Peng B, Juaiti M, Tang Y, Liang B, Ouyang M, Liu Q, Song J. Transcriptome-Wide N6-Methyladenosine Alternations in Pulmonary Arteries of Monocrotaline-Induced Pulmonary Arterial Hypertension in Rats and Novel Therapeutic Targets. Biomedicines 2024; 12:364. [PMID: 38397966 PMCID: PMC10886831 DOI: 10.3390/biomedicines12020364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
N6-methyladenosine (m6A) is a post-transcriptional epigenetic change with transcriptional stability and functionality regulated by specific m6A-modifying enzymes. However, the significance of genes modified by m6A and enzymes specific to m6A regulation in the context of pulmonary arterial hypertension (PAH) remains largely unexplored. MeRIP-seq and RNA-seq were applied to explore variances in m6A and RNA expression within the pulmonary artery tissues of control and monocrotaline-induced PAH rats. Functional enrichments were analyzed using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. To screen candidate m6A-related genes, the STRING and Metascape databases were used to construct a protein-protein interaction network followed by a real-time PCR validation of their expression. The expression level of an m6A regulator was further investigated using immunohistochemical staining, immunofluorescence, and Western blot techniques. Additionally, proliferation assays were conducted on primary rat pulmonary artery smooth muscle cells (PASMCs). We identified forty-two differentially expressed genes that exhibited either hypermethylated or hypomethylated m6A. These genes are predominantly related to the extracellular matrix structure, MAPK, and PI3K/AKT pathways. A candidate gene, centromere protein F (CENPF), was detected with increased expression in the PAH group. Additionally, we first identified an m6A reader, leucine rich pentatricopeptide repeat containing (LRPPRC), which was downregulated in the PAH rat model. The in vitro downregulation of Lrpprc mediated by siRNA resulted in the enhanced proliferation and elevated expression of Cenpf mRNA in primary rat PASMCs. Our study revealed a modified transcriptome-wide m6A landscape and associated regulatory mechanisms in the pulmonary arteries of PAH rats, potentially offering a novel target for therapeutic strategies in the future.
Collapse
Affiliation(s)
- Yilu Feng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China; (Y.F.); (Z.Y.); (B.P.); (M.J.); (Y.T.); (B.L.)
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, China; (J.L.); (M.O.)
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zaixin Yu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China; (Y.F.); (Z.Y.); (B.P.); (M.J.); (Y.T.); (B.L.)
| | - Mi Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Jiang Li
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, China; (J.L.); (M.O.)
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Baohua Peng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China; (Y.F.); (Z.Y.); (B.P.); (M.J.); (Y.T.); (B.L.)
| | - Mukamengjiang Juaiti
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China; (Y.F.); (Z.Y.); (B.P.); (M.J.); (Y.T.); (B.L.)
| | - Yiyang Tang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China; (Y.F.); (Z.Y.); (B.P.); (M.J.); (Y.T.); (B.L.)
| | - Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China; (Y.F.); (Z.Y.); (B.P.); (M.J.); (Y.T.); (B.L.)
| | - Mingqi Ouyang
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, China; (J.L.); (M.O.)
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qingqing Liu
- Department of Respiratory and Critical Care, The Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Jie Song
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, China; (J.L.); (M.O.)
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
7
|
Hopkins CD, Wessel C, Chen O, El-Kersh K, Cave MC, Cai L, Huang J. Potential Roles of Metals in the Pathogenesis of Pulmonary and Systemic Hypertension. Int J Biol Sci 2023; 19:5036-5054. [PMID: 37928257 PMCID: PMC10620830 DOI: 10.7150/ijbs.85590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 11/07/2023] Open
Abstract
Pulmonary and systemic hypertension (PH, SH) are characterized by vasoconstriction and vascular remodeling resulting in increased vascular resistance and pulmonary/aortic artery pressures. The chronic stress leads to inflammation, oxidative stress, and infiltration by immune cells. Roles of metals in these diseases, particularly PH are largely unknown. This review first discusses the pathophysiology of PH including vascular oxidative stress, inflammation, and remodeling in PH; mitochondrial dysfunction and metabolic changes in PH; ion channel and its alterations in the pathogenesis of PH as well as PH-associated right ventricular (RV) remodeling and dysfunctions. This review then summarizes metal general features and essentiality for the cardiovascular system and effects of metals on systemic blood pressure. Lastly, this review explores non-essential and essential metals and potential roles of their dyshomeostasis in PH and RV dysfunction. Although it remains early to conclude the role of metals in the pathogenesis of PH, emerging direct and indirect evidence implicates the possible contributions of metal-mediated toxicities in the development of PH. Future research should focus on comprehensive clinical metallomics study in PH patients; mechanistic evaluations to elucidate roles of various metals in PH animal models; and novel therapy clinical trials targeting metals. These important discoveries will significantly advance our understandings of this rare yet fatal disease, PH.
Collapse
Affiliation(s)
- C. Danielle Hopkins
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Caitlin Wessel
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Oscar Chen
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Karim El-Kersh
- Department of Internal Medicine, Division of Pulmonary Critical Care and Sleep Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew C. Cave
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- The Transplant Program at UofL Health - Jewish Hospital Trager Transplant Center, Louisville, KY, USA
| | - Lu Cai
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- The Transplant Program at UofL Health - Jewish Hospital Trager Transplant Center, Louisville, KY, USA
- Cardiovascular Innovation Institute, Department of Cardiovascular and Thoracic Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
8
|
Saint-Martin Willer A, Santos-Gomes J, Adão R, Brás-Silva C, Eyries M, Pérez-Vizcaino F, Capuano V, Montani D, Antigny F. Physiological and pathophysiological roles of the KCNK3 potassium channel in the pulmonary circulation and the heart. J Physiol 2023; 601:3717-3737. [PMID: 37477289 DOI: 10.1113/jp284936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
Potassium channel subfamily K member 3 (KCNK3), encoded by the KCNK3 gene, is part of the two-pore domain potassium channel family, constitutively active at resting membrane potentials in excitable cells, including smooth muscle and cardiac cells. Several physiological and pharmacological mediators, such as intracellular signalling pathways, extracellular pH, hypoxia and anaesthetics, regulate KCNK3 channel function. Recent studies show that modulation of KCNK3 channel expression and function strongly influences pulmonary vascular cell and cardiomyocyte function. The altered activity of KCNK3 in pathological situations such as atrial fibrillation, pulmonary arterial hypertension and right ventricular dysfunction demonstrates the crucial role of KCNK3 in cardiovascular homeostasis. Furthermore, loss of function variants of KCNK3 have been identified in patients suffering from pulmonary arterial hypertension and atrial fibrillation. This review focuses on current knowledge of the role of the KCNK3 channel in pulmonary circulation and the heart, in healthy and pathological conditions.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Joana Santos-Gomes
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Rui Adão
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Mélanie Eyries
- Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
9
|
Masson B, Saint-Martin Willer A, Dutheil M, Penalva L, Le Ribeuz H, El Jekmek K, Ruchon Y, Cohen-Kaminsky S, Sabourin J, Humbert M, Mercier O, Montani D, Capuano V, Antigny F. Contribution of transient receptor potential canonical channels in human and experimental pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2023; 325:L246-L261. [PMID: 37366608 DOI: 10.1152/ajplung.00011.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is due to progressive distal pulmonary artery (PA) obstruction, leading to right ventricular hypertrophy and failure. Exacerbated store-operated Ca2+ entry (SOCE) contributes to PAH pathogenesis, mediating human PA smooth muscle cell (hPASMC) abnormalities. The transient receptor potential canonical channels (TRPC family) are Ca2+-permeable channels contributing to SOCE in different cell types, including PASMCs. However, the properties, signaling pathways, and contribution to Ca2+ signaling of each TRPC isoform are unclear in human PAH. We studied in vitro the impact of TRPC knockdown on control and PAH-hPASMCs function. In vivo, we analyzed the consequences of pharmacological TRPC inhibition using the experimental model of pulmonary hypertension (PH) induced by monocrotaline (MCT) exposure. Compared with control-hPASMCs cells, in PAH-hPASMCs, we found a decreased TRPC4 expression, overexpression of TRPC3 and TRPC6, and unchanged TRPC1 expression. Using the siRNA strategy, we found that the knockdown of TRPC1-C3-C4-C6 reduced the SOCE and the proliferation rate of PAH-hPASMCs. Only TRPC1 knockdown decreased the migration capacity of PAH-hPASMCs. After PAH-hPASMCs exposure to the apoptosis inducer staurosporine, TRPC1-C3-C4-C6 knockdown increased the percentage of apoptotic cells, suggesting that these channels promote apoptosis resistance. Only TRPC3 function contributed to exacerbated calcineurin activity. In the MCT-PH rat model, only TRPC3 protein expression was increased in lungs compared with control rats, and in vivo "curative" administration of a TRPC3 inhibitor attenuated PH development in rats. These results suggest that TRPC channels contribute to PAH-hPASMCs dysfunctions, including SOCE, proliferation, migration, and apoptosis resistance, and could be considered as therapeutic targets in PAH.NEW & NOTEWORTHY TRPC3 is increased in human and experimental pulmonary arterial hypertension (PAH). In PAH pulmonary arterial smooth muscle cells, TRPC3 participates in the aberrant store-operated Ca2+ entry contributing to their pathological cell phenotypes (exacerbated proliferation, enhanced migration, apoptosis resistance, and vasoconstriction). Pharmacological in vivo inhibition of TRPC3 reduces the development of experimental PAH. Even if other TRPC acts on PAH development, our results prove that TRPC3 inhibition could be considered as an innovative treatment for PAH.
Collapse
Affiliation(s)
- Bastien Masson
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Anais Saint-Martin Willer
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Mary Dutheil
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France
| | - Lucille Penalva
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Hélène Le Ribeuz
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Kristelle El Jekmek
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Yann Ruchon
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France
| | - Sylvia Cohen-Kaminsky
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Jessica Sabourin
- INSERM UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Olaf Mercier
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Le Plessis Robinson, France
| | - David Montani
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Véronique Capuano
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
10
|
Novel Molecular Mechanisms Involved in the Medical Treatment of Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24044147. [PMID: 36835558 PMCID: PMC9965798 DOI: 10.3390/ijms24044147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe condition with a high mortality rate despite advances in diagnostic and therapeutic strategies. In recent years, significant scientific progress has been made in the understanding of the underlying pathobiological mechanisms. Since current available treatments mainly target pulmonary vasodilation, but lack an effect on the pathological changes that develop in the pulmonary vasculature, there is need to develop novel therapeutic compounds aimed at antagonizing the pulmonary vascular remodeling. This review presents the main molecular mechanisms involved in the pathobiology of PAH, discusses the new molecular compounds currently being developed for the medical treatment of PAH and assesses their potential future role in the therapeutic algorithms of PAH.
Collapse
|
11
|
Inhibition of JAK1,2 Prevents Fibrotic Remodeling of Pulmonary Vascular Bed and Improves Outcomes in the Rat Model of Chronic Thromboembolic Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms232415646. [PMID: 36555286 PMCID: PMC9779027 DOI: 10.3390/ijms232415646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/15/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare complication of acute pulmonary embolism with poor clinical outcomes. Therapeutic approaches to prevention of fibrotic remodeling of the pulmonary vascular bed in CTEPH are limited. In this work, we tested the hypothesis that Janus kinase 1/2 (JAK1/2) inhibition with ruxolitinib might prevent and attenuate CTEPH in a rat model. CTEPH was induced by repeated embolization of the pulmonary artery with partially biodegradable 180 ± 30 μm alginate microspheres. Two weeks after the last injection of microspheres, ruxolitinib was administered orally at doses of 0.86, 2.58, and 4.28 mg/kg per day for 4 weeks. Prednisolone (1.475 mg/kg, i.m.) was used as a reference drug. Ruxolitinib in all doses as well as prednisolone reduced pulmonary vascular wall hypertrophy. Ruxolitinib at a dose of 2.58 mg/kg and prednisolone reduced vascular wall fibrosis. Prednisolone treatment resulted in decreased right ventricular systolic pressure. Pulmonary vascular resistance was lower in the prednisolone and ruxolitinib (4.28 mg/kg) groups in comparison with the placebo group. The plasma level of brain natriuretic peptide was lower in groups receiving ruxolitinib at doses of 2.58 and 4.28 mg/kg versus placebo. This study demonstrated that JAK1/2 inhibitor ruxolitinib dose-dependently reduced pulmonary vascular remodeling, thereby preventing CTEPH formation in rats.
Collapse
|
12
|
Preethi KA, Selvakumar SC, Ross K, Sekar D. Therapeutic aspect of microRNA inhibition in various types of hypertension and hypertensive complications. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
13
|
Masson B, Le Ribeuz H, Sabourin J, Laubry L, Woodhouse E, Foster R, Ruchon Y, Dutheil M, Boët A, Ghigna MR, De Montpreville VT, Mercier O, Beech DJ, Benitah JP, Bailey MA, Humbert M, Montani D, Capuano V, Antigny F. Orai1 Inhibitors as Potential Treatments for Pulmonary Arterial Hypertension. Circ Res 2022; 131:e102-e119. [PMID: 36164973 DOI: 10.1161/circresaha.122.321041] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by progressive distal pulmonary artery (PA) obstruction, leading to right ventricular hypertrophy and failure. Exacerbated intracellular calcium (Ca2+) signaling contributes to abnormalities in PA smooth muscle cells (PASMCs), including aberrant proliferation, apoptosis resistance, exacerbated migration, and arterial contractility. Store-operated Ca2+ entry is involved in Ca2+ homeostasis in PASMCs, but its properties in PAH are unclear. METHODS Using a combination of Ca2+ imaging, molecular biology, in vitro, ex vivo, and in vivo approaches, we investigated the roles of the Orai1 SOC channel in PA remodeling in PAH and determined the consequences of pharmacological Orai1 inhibition in vivo using experimental models of pulmonary hypertension (PH). RESULTS Store-operated Ca2+ entry and Orai1 mRNA and protein were increased in human PASMCs (hPASMCs) from patients with PAH (PAH-hPASMCs). We found that MEK1/2 (mitogen-activated protein kinase kinase 1/2), NFAT (nuclear factor of activated T cells), and NFκB (nuclear factor-kappa B) contribute to the upregulation of Orai1 expression in PAH-hPASMCs. Using small interfering RNA (siRNA) and Orai1 inhibitors, we found that Orai1 inhibition reduced store-operated Ca2+ entry, mitochondrial Ca2+ uptake, aberrant proliferation, apoptosis resistance, migration, and excessive calcineurin activity in PAH-hPASMCs. Orai1 inhibitors reduced agonist-evoked constriction in human PAs. In experimental rat models of PH evoked by chronic hypoxia, monocrotaline, or Sugen/hypoxia, administration of Orai1 inhibitors (N-{4-[3,5-bis(Trifluoromethyl)-1H-pyrazol-1-yl]phenyl}-4-methyl-1,2,3-thiadiazole-5-carboxamide [BTP2], 4-(2,5-dimethoxyphenyl)-N-[(pyridin-4-yl)methyl]aniline [JPIII], or 5J4) protected against PH. CONCLUSIONS In human PAH and experimental PH, Orai1 expression and activity are increased. Orai1 inhibition normalizes the PAH-hPASMCs phenotype and attenuates PH in rat models. These results suggest that Orai1 should be considered as a relevant therapeutic target for PAH.
Collapse
Affiliation(s)
- Bastien Masson
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| | - Hélène Le Ribeuz
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| | - Jessica Sabourin
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (J.S., J.-P.B.)
| | - Loann Laubry
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Emily Woodhouse
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Richard Foster
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Yann Ruchon
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Mary Dutheil
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Angèle Boët
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Maria-Rosa Ghigna
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| | | | - Olaf Mercier
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Le Plessis Robinson, France (O.M.)
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Jean-Pierre Benitah
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (J.S., J.-P.B.)
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Marc Humbert
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (M.H., D.M.)
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (M.H., D.M.)
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| |
Collapse
|
14
|
Upregulation of miR-335-5p Contributes to Right Ventricular Remodeling via Calumenin in Pulmonary Arterial Hypertension. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9294148. [PMID: 36246958 PMCID: PMC9557250 DOI: 10.1155/2022/9294148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/08/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022]
Abstract
Right ventricular (RV) failure determines the prognosis in pulmonary arterial hypertension (PAH), but the underlying mechanism is still unclear. Growing evidence has shown that microRNAs participate in RV remodeling. This study is undertaken to explore the role of miR-335-5p in regulating RV remodeling induced by PAH. Two PAH models were used in the study, including the monocrotaline rat model and hypoxia/su5416 mouse model. miRNA sequencing and RT-qPCR validation identified that miR-335-5p was elevated in the RV of PAH rats. In vitro, miR-335-5p expression was increased after angiotensin II treatment, and miR-335-5p inhibition relieved angiotensin II-induced cardiomyocyte hypertrophy. The luciferase reporter assay showed that calumenin was a target gene for miR-335-5p. Pretreatment with miR-335-5p inhibitors could rescue calumenin downregulation induced by angiotensin II in H9C2 cells. Moreover, intracellular Ca2+ concentration and apoptosis were increased after angiotensin II treatment, and miR-335-5p inhibition decreased intracellular Ca2+ accumulation and apoptosis. Finally, in vivo miR-335-5p downregulation (antagomir miR-335-5p) attenuated RV remodeling and rescued calumenin downregulation under conditions of hypoxia/su5416 exposure. Our work highlights the role of miR-335-5p and calumenin in RV remodeling and may lead to the development of novel therapeutic strategies for right heart failure.
Collapse
|
15
|
Redel-Traub G, Sampson KJ, Kass RS, Bohnen MS. Potassium Channels as Therapeutic Targets in Pulmonary Arterial Hypertension. Biomolecules 2022; 12:1341. [PMID: 36291551 PMCID: PMC9599705 DOI: 10.3390/biom12101341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 12/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease with high morbidity and mortality. Deleterious remodeling in the pulmonary arterial system leads to irreversible arterial constriction and elevated pulmonary arterial pressures, right heart failure, and eventually death. The difficulty in treating PAH stems in part from the complex nature of disease pathogenesis, with several signaling compounds known to be involved (e.g., endothelin-1, prostacyclins) which are indeed targets of PAH therapy. Over the last decade, potassium channelopathies were established as novel causes of PAH. More specifically, loss-of-function mutations in the KCNK3 gene that encodes the two-pore-domain potassium channel KCNK3 (or TASK-1) and loss-of-function mutations in the ABCC8 gene that encodes a key subunit, SUR1, of the ATP-sensitive potassium channel (KATP) were established as the first two potassium channelopathies in human cohorts with pulmonary arterial hypertension. Moreover, voltage-gated potassium channels (Kv) represent a third family of potassium channels with genetic changes observed in association with PAH. While other ion channel genes have since been reported in association with PAH, this review focuses on KCNK3, KATP, and Kv potassium channels as promising therapeutic targets in PAH, with recent experimental pharmacologic discoveries significantly advancing the field.
Collapse
Affiliation(s)
- Gabriel Redel-Traub
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kevin J. Sampson
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Robert S. Kass
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael S. Bohnen
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
16
|
Pujar M, Vastrad B, Kavatagimath S, Vastrad C, Kotturshetti S. Identification of candidate biomarkers and pathways associated with type 1 diabetes mellitus using bioinformatics analysis. Sci Rep 2022; 12:9157. [PMID: 35650387 PMCID: PMC9160069 DOI: 10.1038/s41598-022-13291-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 05/16/2022] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a metabolic disorder for which the underlying molecular mechanisms remain largely unclear. This investigation aimed to elucidate essential candidate genes and pathways in T1DM by integrated bioinformatics analysis. In this study, differentially expressed genes (DEGs) were analyzed using DESeq2 of R package from GSE162689 of the Gene Expression Omnibus (GEO). Gene ontology (GO) enrichment analysis, REACTOME pathway enrichment analysis, and construction and analysis of protein–protein interaction (PPI) network, modules, miRNA-hub gene regulatory network and TF-hub gene regulatory network, and validation of hub genes were performed. A total of 952 DEGs (477 up regulated and 475 down regulated genes) were identified in T1DM. GO and REACTOME enrichment result results showed that DEGs mainly enriched in multicellular organism development, detection of stimulus, diseases of signal transduction by growth factor receptors and second messengers, and olfactory signaling pathway. The top hub genes such as MYC, EGFR, LNX1, YBX1, HSP90AA1, ESR1, FN1, TK1, ANLN and SMAD9 were screened out as the critical genes among the DEGs from the PPI network, modules, miRNA-hub gene regulatory network and TF-hub gene regulatory network. Receiver operating characteristic curve (ROC) analysis confirmed that these genes were significantly associated with T1DM. In conclusion, the identified DEGs, particularly the hub genes, strengthen the understanding of the advancement and progression of T1DM, and certain genes might be used as candidate target molecules to diagnose, monitor and treat T1DM.
Collapse
Affiliation(s)
- Madhu Pujar
- Department of Pediatrics, J J M Medical College, Davangere, Karnataka, 577004, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag, Karnataka, 582101, India
| | - Satish Kavatagimath
- Department of Pharmacognosy, K.L.E. College of Pharmacy, Belagavi, Karnataka, 590010, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India.
| | - Shivakumar Kotturshetti
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India
| |
Collapse
|
17
|
Wang J, Jiang R, Tan Y, Cheng K. Human pulmonary artery smooth muscle cell dysfunction is regulated by miR-509-5p in hypoxic environment. Cell Cycle 2022; 21:1212-1221. [PMID: 35244512 PMCID: PMC9103279 DOI: 10.1080/15384101.2022.2044147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Reportedly, dysfunction of human pulmonary arterial smooth muscle cells (PASMCs) is associated with the pathogenesis of pulmonary arterial hypertension (PAH). Herein, the role of miR-509-5p in hypoxia-induced PASMCs and the underlying mechanism were explored. PASMCs were cultured under both normoxia and hypoxia conditions. Quantitative real-time polymerase-chain reaction (qPCR) was employed for quantifying the expressions of miR-509-5p and DNMT1 mRNA in the serum of PAH patients and PASMCs. MiR-509-5p mimics and inhibitors were then, respectively, transfected into PAMSCs, and CCK-8 and Transwell assays were utilized to detect PASMCs' proliferation and migration. Flow cytometry was executed for evaluating PASMCs' apoptosis. Interrelation between miR-509-5p and DNMT1 was determined utilizing bioinformatics analysis and dual-luciferase reporter assay. Western blot assay was used to detect the expression of DNMT1 or SOD2. MiR-509-5p in serum samples of patients with PAH as well as hypoxia-induced PASMCs was significantly down-regulated, whereas DNMT1 was markedly up-regulated. MiR-509-5p mimics reduces the proliferation and migration of PASMCs, but promotes the apoptosis; conversely, miR-509-5p inhibitors exerted opposite effects. DNMT1 was identified as a target gene of miR-509-5p, and overexpression of DNMT1 reversed the biological functions of miR-509-5p in regulating the phenotypes of PAMSCs. MiR-509-5p up-regulated the expression of SOD2 by down-regulating DNMT1. MiR-509-5p regulates the proliferation, migration and apoptosis of PASMCs, and restoration of miR-509-5p may be a promising strategy to treat PAH.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Yanlin Tan
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Kuan Cheng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, P.R. China,CONTACT Kuan Cheng Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Xietu Road No.1609, Shanghai200032, P.R.China
| |
Collapse
|
18
|
hsa_circWDR37_016 Regulates Hypoxia-Induced Proliferation of Pulmonary Arterial Smooth Muscle Cells. Cardiovasc Ther 2022; 2022:7292034. [PMID: 35116078 PMCID: PMC8786516 DOI: 10.1155/2022/7292034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/01/2021] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by abnormal remodeling of pulmonary vessel walls caused by excessive pulmonary arterial smooth muscle cell (PASMC) proliferation. Our previous clinical studies have demonstrated the importance of the downregulated circRNA in PAH. However, the role of upregulated circRNAs is still elusive. Here, we identified the upregulated circRNA in PAH patients, hsa_circWDR37_016 (circWDR37), as a key regulator of hypoxic proliferative disorder of pulmonary arterial smooth muscle cells (PASMCs). Quantitative real-time PCR (qRT-PCR) analysis validated that exposure to hypoxia markedly increased the circWDR37 level in cultured human PASMCs. As evidenced by flow cytometry, 5-ethynyl-2′-deoxyuridine (EdU) incorporation, wound healing, and Tunel assay, silencing of endogenous circWDR37 attenuated proliferation and cell-cycle progression in hypoxia-exposed human PASMCs in vitro. Furthermore, bioinformatics and Luciferase assay showed that circWDR37 directly sponged hsa-miR-138-5p (miR-138) and was involved in the immunoregulatory and inflammatory processes of PAH. Together, these studies suggested new insights into circRNA regulated the pathology of PAH, providing a new potential therapeutic target for PAH treatment.
Collapse
|
19
|
Feng X, Zhan F, Luo D, Hu J, Wei G, Hua F, Xu G. LncRNA 4344 promotes NLRP3-related neuroinflammation and cognitive impairment by targeting miR-138-5p. Brain Behav Immun 2021; 98:283-298. [PMID: 34455059 DOI: 10.1016/j.bbi.2021.08.230] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/01/2021] [Accepted: 08/21/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Cognitive impairment is a common neurological disease of which NLRP3-related neuroinflammation has been demonstrated to be an essential mediator. Previous studies have indicated that long non-coding RNAs (lncRNAs) are critical for the development of neurological disorders. However, the roles and functions of lncRNA 4344 in neuroinflammation during cognitive impairment are unknown and need to be further elucidated. METHODS Lipopolysaccharide (LPS)-induced rat cognitive impairment and rat microglia (RM) cell inflammation models were established in vitro and in vivo. The Morris water maze test was used to evaluate the cognitive behavior of the rats. Gene expression was assessed using real-time quantitative polymerase chain reaction, and protein levels using enzyme-linked immunosorbent assay, or western blot analysis. The targeting relationship between lncRNA 4344, miR-138-5p, and NLRP3 was identified using bioinformatics analysis and a dual-luciferase reporter gene assay. Hematoxylin-Eosin and Nissl stainings, terminal deoxynucleotidyl transferase dUTP nick end labeling, or immunofluorescence staining assays were performed to detect pathological changes, neuronal apoptosis, or positive cells in hippocampal tissues, respectively. RESULTS The expression levels of lncRNA 4344 and NLRP3 were upregulated in the hippocampal tissues of LPS-treated rats and RM cells, and showed a strong positive correlation between each other. LncRNA 4344 overexpression further enhanced the expression of NLRP3 and its downstream genes (caspase-1, IL-1β, and IL-18), as well as neuronal apoptosis in LPS-stimulated RM cells, whereas lncRNA 4344 silencing attenuated the inflammatory injuries. Moreover, miR-138-5p was the direct target of lncRNA 4344 and was downregulated in the RM cell inflammation model. We also found that miR-138-5p directly reduced the expression of NLRP3 and its downstream genes. Subsequently, the results of the animal experiments showed that the lncRNA 4344/miR-138-5p/NLRP3 axis plays an essential role in regulating the cognitive behavior, pathological changes and apoptosis of hippocampal neurons, expression of inflammation-related factors (NLRP3, caspase-1, IL-1β, and IL-18), and microglial activation in LPS-induced cognitive impairment rats. CONCLUSION Our results demonstrated for the first time that lncRNA 4344 regulates NLRP3-related neuroinflammation and cognitive impairment by targeting miR-138-5p, providing a possible target for the treatment of diseases characterized by a cognitive deficit.
Collapse
Affiliation(s)
- Xiaojin Feng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang 330006, Jiangxi, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang 330006, Jiangxi, China
| | - Deqiang Luo
- Department of Intensive Care Unit, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jialing Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang 330006, Jiangxi, China.
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
20
|
Ion channels as convergence points in the pathology of pulmonary arterial hypertension. Biochem Soc Trans 2021; 49:1855-1865. [PMID: 34346486 PMCID: PMC8421048 DOI: 10.1042/bst20210538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease of the cardiopulmonary system that lacks curative treatments. The main pathological event in PAH is elevated vascular resistance in the pulmonary circulation, caused by abnormal vasoconstriction and vascular remodelling. Ion channels are key determinants of vascular smooth muscle tone and homeostasis, and four PAH channelopathies (KCNK3, ABCC8, KCNA5, TRPC6) have been identified so far. However, the contribution of ion channels in other forms of PAH, which account for the majority of PAH patients, has been less well characterised. Here we reason that a variety of triggers of PAH (e.g. BMPR2 mutations, hypoxia, anorectic drugs) that impact channel function may contribute to the onset of the disease. We review the molecular mechanisms by which these ‘extrinsic’ factors converge on ion channels and provoke their dysregulation to promote the development of PAH. Ion channels of the pulmonary vasculature are therefore promising therapeutic targets because of the modulation they provide to both vasomotor tone and proliferation of arterial smooth muscle cells.
Collapse
|
21
|
Feng X, Hu J, Zhan F, Luo D, Hua F, Xu G. MicroRNA-138-5p Regulates Hippocampal Neuroinflammation and Cognitive Impairment by NLRP3/Caspase-1 Signaling Pathway in Rats. J Inflamm Res 2021; 14:1125-1143. [PMID: 33814920 PMCID: PMC8009546 DOI: 10.2147/jir.s304461] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/04/2021] [Indexed: 12/23/2022] Open
Abstract
Purpose Neuroinflammation is an essential causative factor in the pathogenesis and progression of cognitive impairment. The present study aims to evaluate the critical role of microRNA-138-5p (miR-138-5p) in hippocampal neuroinflammation and cognitive impairment through the NLRP3/caspase-1 signaling pathway in rats. Material and Methods We established the cognitive impairment rat model and RM (Rat microglia) microglial cellular inflammation model by intracerebroventricular (icv) injection or stimulation of lipopolysaccharide (LPS). Morris water maze (MWM) and Y-maze tests were performed to assess the cognitive behaviors. Quantitative real-time polymerase chain reaction (qRT-PCR), Enzyme-linked immune-sorbent assay (ELISA) and Western blot analysis were utilized to evaluate mRNA or protein expression. Bioinformatic analysis and dual-luciferase reporter gene assay were performed to verify the targeting relationship between NLRP3 and miR-138-5p. Besides, Hematoxylin and eosin (H&E) staining and immunohistochemistry were applied to observe the neuronal morphology and detect the positive cells of the hippocampus, respectively. Results Compared to the control groups, LPS-treated rats exhibited significantly impaired learning and memory in MWM and Y-maze tests. The expression of NLRP3, caspase-1 and pro-inflammation cytokines (IL-1β and IL-18) were upregulated, while miR-138-5p was downregulated both in rat hippocampus and RM cells treated with LPS. MiR-138-5p is downregulated in microarray data of cognitive impairment animals and could directly target the 3ʹ-UTR of NLRP3. Furthermore, upregulation of miR-138-5p improved impaired cognitive functions, while inhibited hippocampal neuroinflammation demonstrated by decreased expression of NLRP3/caspase-1 axis, pro-inflammation cytokines and microglial activation. This study demonstrates for the first time that miR-138-5p suppresses the hippocampal NLRP3/caspase-1 signaling pathway activation in cognition impaired rats. Conclusion The low expression of miR-138-5p after LPS administration may contribute to the activation of the NLRP3/caspase-1 pathway, leading to hippocampal neuroinflammation and cognitive impairment in rat models. These findings indicate a promising therapeutic avenue for cognitive disorders.
Collapse
Affiliation(s)
- Xiaojin Feng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jialing Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Deqiang Luo
- Department of Intensive Care Unit, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi 330006, People's Republic of China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi 330006, People's Republic of China
| |
Collapse
|
22
|
Epigenetic Regulation of Pulmonary Arterial Hypertension-Induced Vascular and Right Ventricular Remodeling: New Opportunities? Int J Mol Sci 2020; 21:ijms21238901. [PMID: 33255338 PMCID: PMC7727715 DOI: 10.3390/ijms21238901] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
Pulmonary artery hypertension (PAH) is a rare chronic disease with high impact on patients’ quality of life and currently no available cure. PAH is characterized by constant remodeling of the pulmonary artery by increased proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), fibroblasts (FBs) and endothelial cells (ECs). This remodeling eventually leads to increased pressure in the right ventricle (RV) and subsequent right ventricle hypertrophy (RVH) which, when left untreated, progresses into right ventricle failure (RVF). PAH can not only originate from heritable mutations, but also develop as a consequence of congenital heart disease, exposure to drugs or toxins, HIV, connective tissue disease or be idiopathic. While much attention was drawn into investigating and developing therapies related to the most well understood signaling pathways in PAH, in the last decade, a shift towards understanding the epigenetic mechanisms driving the disease occurred. In this review, we reflect on the different epigenetic regulatory factors that are associated with the pathology of RV remodeling, and on their relevance towards a better understanding of the disease and subsequently, the development of new and more efficient therapeutic strategies.
Collapse
|