1
|
Willmer T, Mabasa L, Sharma J, Muller CJF, Johnson R. Blood-Based DNA Methylation Biomarkers to Identify Risk and Progression of Cardiovascular Disease. Int J Mol Sci 2025; 26:2355. [PMID: 40076974 PMCID: PMC11900213 DOI: 10.3390/ijms26052355] [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: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
Non-communicable diseases (NCDs) are the leading cause of death worldwide, with cardiovascular disease (CVD) accounting for half of all NCD-related deaths. The biological onset of CVD may occur long before the development of clinical symptoms, hence the urgent need to understand the molecular alterations underpinning CVD, which would facilitate intervention strategies to prevent or delay the onset of the disease. There is evidence to suggest that CVD develops through a complex interplay between genetic, lifestyle, and environmental factors. Epigenetic modifications, including DNA methylation, serve as proxies linking genetics and the environment to phenotypes and diseases. In the past decade, a growing list of studies has implicated DNA methylation in the early events of CVD pathogenesis. In this regard, screening for these epigenetic marks in asymptomatic individuals may assist in the early detection of CVD and serve to predict the response to therapeutic interventions. This review discusses the current literature on the relationship between blood-based DNA methylation alterations and CVD in humans. We highlight a set of differentially methylated genes that show promise as candidates for diagnostic and prognostic CVD biomarkers, which should be prioritized and replicated in future studies across additional populations. Finally, we discuss key limitations in DNA methylation studies, including genetic diversity, interpatient variability, cellular heterogeneity, study confounders, different methodological approaches used to isolate and measure DNA methylation, sample sizes, and cross-sectional study design.
Collapse
Affiliation(s)
- Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Jyoti Sharma
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa 3886, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| |
Collapse
|
2
|
Taizhanova D, Nurpissova T, Abildinova G, Martynyuk T, Kulmyrzayeva N, Zholdybayeva E. Hemodynamic and Genetic Associations with the Risk of Idiopathic Pulmonary Arterial Hypertension Development in an Ethnic Cohort of Kazakhs. Diagnostics (Basel) 2024; 14:2687. [PMID: 39682595 DOI: 10.3390/diagnostics14232687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
INTRODUCTION Idiopathic pulmonary arterial hypertension (IPAH) is a progressive and fatal disease. The aim of this study was to evaluate the association of polymorphism of the type 2 bone morphogenetic protein receptor gene (BMPR2) with the risk of IPAH development in an ethnic group of Kazakhs. We also describe the clinical and hemodynamic characteristics and outcomes of patients with and without carriers of BMPR2 gene mutations in IPAH. No available research highlights this problem in an ethnic group of Kazakhs. MATERIALS AND METHODS A total of 53 patients of only Kazakh nationality with IPAH participated in the study. Clinical, functional, and hemodynamic characteristics, as well as the outcome of the disease, were compared among carriers and non-carriers of the BMPR2 mutation. RESULTS When receiving IPAH diagnosis, the average age of patients was 40.0 (32.0-48.0) years. Women predominated among the patients (86.8%). Of these, 17 (32.0%) were carriers of the gene mutation, and 36 (68.0%) did not have this mutation. The results of our research demonstrate that the Rs17199249 variant in BMPR2 contributed to increased susceptibility to IPAH. The T allele was associated with an increased risk of IPAH, with T = 75 (70.75%), G = 31 (29.24%), MAF-0.2925, x2-0.001, and HWE p-0.975. Carriers of the BMPR2 mutation were predominantly women (80.0%), and they had higher pulmonary vascular resistance (8.7-14.9 vs. 5.9-12.6 WU; p = 0.038), a low cardiac index (1.9-2.6 vs. 2.3-3.1 L/min per m2; p = 0.027), and a shorter time to death (p = 0.022). CONCLUSIONS This is the first study of the genetic causes of IPAH that demonstrates the genetic polymorphism of BMPR2 is associated with an increased risk of IPAH developing with worse hemodynamic parameters and clinical outcomes.
Collapse
Affiliation(s)
- Dana Taizhanova
- Department of Internal Diseases, Karaganda Medical University Non-Commercial Joint Stock Company, Karaganda 100000, Kazakhstan
| | - Togzhan Nurpissova
- Department of Internal Diseases, Karaganda Medical University Non-Commercial Joint Stock Company, Karaganda 100000, Kazakhstan
- Department of Therapy No. 7, Medical Center Hospital of the President's Affairs Administration of the Republic of Kazakhstan, Astana 010000, Kazakhstan
| | - Gulshara Abildinova
- Laboratory of Personalized Genomic Diagnostics, Medical Center Hospital of the President's Affairs Administration of the Republic of Kazakhstan, Astana 010000, Kazakhstan
| | - Tamilla Martynyuk
- Institution «National Medical Cardiology Research Center Named After Academician Ye. I. Chazov» of the Ministry of Health of the Russian Federation, Moscow 105064, Russia
| | - Nazgul Kulmyrzayeva
- Department of Therapy No. 7, Medical Center Hospital of the President's Affairs Administration of the Republic of Kazakhstan, Astana 010000, Kazakhstan
| | - Elena Zholdybayeva
- National Scientific Shared Laboratory of Biotechnology, National Center of Biotechnology Limited Liability Partnership, Astana 010000, Kazakhstan
| |
Collapse
|
3
|
Zhang S, Wang J, Wen J, Xin Q, Wang J, Ju Z, Luan Y. MSC-derived exosomes attenuates pulmonary hypertension via inhibiting pulmonary vascular remodeling. Exp Cell Res 2024; 442:114256. [PMID: 39299482 DOI: 10.1016/j.yexcr.2024.114256] [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: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a serious cardiopulmonary disease with significant morbidity and mortality. Vascular obstruction leads to a continuous increase in pulmonary vascular resistance, vascular remodeling, and right ventricular hypertrophy and failure, which are the main pathological features of PH. Currently, the treatments for PH are very limited, so new methods are urgently needed. Msenchymal stem cells-derived exosomes have been shown to have significant therapeutic effects in PH, however, the mechanism still very blurry. Here, we investigated the possible mechanism by which umbilical cord mesenchymal stem cell-derived exosomes (hUC-MSC-EXO) inhibited monocrotaline (MCT)-induced pulmonary vascular remodeling in a rat model of PH by regulating the NF-κB/BMP signaling pathway. Our data revealed that hUC-MSC-EXO could significantly attenuate MCT-induced PH and right ventricular hypertrophy. Moreover, the protein expression level of BMPR2, BMP-4, BMP-9 and ID1 was significantly increased, but NF-κB p65, p-NF-κB-p65 and BMP antagonists Gremlin-1 was increased in vitro and vivo. Collectively, this study revealed that the mechanism of hUC-MSC-EXO attenuates pulmonary hypertension may be related to inhibition of NF-κB signaling to further activation of BMP signaling. The present study provided a promising therapeutic strategy for PH vascular remodeling.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Emergency, The Second Hospital of Shandong University, PR China
| | - Junfu Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, PR China
| | - Jiang Wen
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Qian Xin
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Jue Wang
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Zhiye Ju
- Department of Ultrasound, Shandong Provincial Public Health Clinical Center, No. 46, Lishan Road, Jinan, 250000, PR China.
| | - Yun Luan
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China.
| |
Collapse
|
4
|
Todesco A, Grynblat J, Akoumia KKF, Bonnet D, Mendes‐Ferreira P, Morisset S, Chemla D, Levy M, Méot M, Malekzadeh‐Milani S, Tielemans B, Decante B, Vastel‐Amzallag C, Habert P, Ghigna M, Humbert M, Montani D, Boulate D, Perros F. Pulmonary Hypertension Induced by Right Pulmonary Artery Occlusion: Hemodynamic Consequences of Bmpr2 Mutation. J Am Heart Assoc 2024; 13:e034621. [PMID: 38979789 PMCID: PMC11292755 DOI: 10.1161/jaha.124.034621] [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: 02/17/2024] [Accepted: 05/28/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The primary genetic risk factor for heritable pulmonary arterial hypertension is the presence of monoallelic mutations in the BMPR2 gene. The incomplete penetrance of BMPR2 mutations implies that additional triggers are necessary for pulmonary arterial hypertension occurrence. Pulmonary artery stenosis directly raises pulmonary artery pressure, and the redirection of blood flow to unobstructed arteries leads to endothelial dysfunction and vascular remodeling. We hypothesized that right pulmonary artery occlusion (RPAO) triggers pulmonary hypertension (PH) in rats with Bmpr2 mutations. METHODS AND RESULTS Male and female rats with a 71 bp monoallelic deletion in exon 1 of Bmpr2 and their wild-type siblings underwent acute and chronic RPAO. They were subjected to full high-fidelity hemodynamic characterization. We also examined how chronic RPAO can mimic the pulmonary gene expression pattern associated with installed PH in unobstructed territories. RPAO induced precapillary PH in male and female rats, both acutely and chronically. Bmpr2 mutant and male rats manifested more severe PH compared with their counterparts. Although wild-type rats adapted to RPAO, Bmpr2 mutant rats experienced heightened mortality. RPAO induced a decline in cardiac contractility index, particularly pronounced in male Bmpr2 rats. Chronic RPAO resulted in elevated pulmonary IL-6 (interleukin-6) expression and decreased Gdf2 expression (corrected P value<0.05 and log2 fold change>1). In this context, male rats expressed higher pulmonary levels of endothelin-1 and IL-6 than females. CONCLUSIONS Our novel 2-hit rat model presents a promising avenue to explore the adaptation of the right ventricle and pulmonary vasculature to PH, shedding light on pertinent sex- and gene-related effects.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Female
- Male
- Pulmonary Artery/physiopathology
- Pulmonary Artery/metabolism
- Hemodynamics
- Disease Models, Animal
- Mutation
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Rats
- Rats, Sprague-Dawley
- Vascular Remodeling/genetics
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/etiology
- Stenosis, Pulmonary Artery/genetics
- Stenosis, Pulmonary Artery/physiopathology
- Stenosis, Pulmonary Artery/metabolism
- Arterial Pressure
- Myocardial Contraction/physiology
Collapse
Affiliation(s)
- Alban Todesco
- Department of Thoracic Surgery, Diseases of the Esophagus and Lung Transplantation, North HospitalAix Marseille University, Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
| | - Julien Grynblat
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
- M3C‐Necker, Hôpital Necker‐Enfants maladesAP‐HP Université de Paris Cité, Cardiologie Congénitale et PédiatriqueParisFrance
- Faculty of Medicine Le Kremlin‐BicêtreUniversité Paris‐SaclayBures‐sur‐YvetteFrance
| | - Kouamé Kan Firmin Akoumia
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
| | - Damien Bonnet
- M3C‐Necker, Hôpital Necker‐Enfants maladesAP‐HP Université de Paris Cité, Cardiologie Congénitale et PédiatriqueParisFrance
| | - Pedro Mendes‐Ferreira
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and PhysiologyFaculty of Medicine of the University of PortoPortoPortugal
- Paris‐Porto Pulmonary Hypertension Collaborative Laboratory (3PH), UMR_S 999, INSERMUniversité Paris‐SaclayParisFrance
| | | | - Denis Chemla
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
| | - Marilyne Levy
- M3C‐Necker, Hôpital Necker‐Enfants maladesAP‐HP Université de Paris Cité, Cardiologie Congénitale et PédiatriqueParisFrance
| | - Mathilde Méot
- M3C‐Necker, Hôpital Necker‐Enfants maladesAP‐HP Université de Paris Cité, Cardiologie Congénitale et PédiatriqueParisFrance
| | - Sophie‐Guiti Malekzadeh‐Milani
- M3C‐Necker, Hôpital Necker‐Enfants maladesAP‐HP Université de Paris Cité, Cardiologie Congénitale et PédiatriqueParisFrance
| | - Birger Tielemans
- Department of Imaging and Pathology, Biomedical MRI unit/MosaicKU LeuvenLeuvenBelgium
| | - Benoit Decante
- Preclinical Research Laboratory, Pulmonary Hypertension National Referral Center, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint JosephParis‐Saclay UniversityLe Plessis RobinsonFrance
| | - Carine Vastel‐Amzallag
- Paediatric Cardiology, Centre de Spécialités Pédiatriques de l’Est Parisien, CSPEPCréteilFrance
| | - Paul Habert
- Department of RadiologyNorth Hospital, Assistance Publique–Hôpitaux de MarseilleMarseilleFrance
- Aix Marseille Univ, LIIEMarseilleFrance
| | - Maria‐Rosa Ghigna
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
- Department of PathologyInstitut Gustave RoussyVillejuifFrance
| | - Marc Humbert
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
- Faculty of Medicine Le Kremlin‐BicêtreUniversité Paris‐SaclayBures‐sur‐YvetteFrance
- AP‐HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral CentreDMU 5 Thorinno, Hôpital BicêtreLe Kremlin‐BicêtreFrance
| | - David Montani
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
- Faculty of Medicine Le Kremlin‐BicêtreUniversité Paris‐SaclayBures‐sur‐YvetteFrance
- AP‐HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral CentreDMU 5 Thorinno, Hôpital BicêtreLe Kremlin‐BicêtreFrance
| | - David Boulate
- Department of Thoracic Surgery, Diseases of the Esophagus and Lung Transplantation, North HospitalAix Marseille University, Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
- COMPutational Pharmacology and clinical Oncology (COMPO), INRIA‐INSERMAix Marseille UniversityMarseilleFrance
| | - Frédéric Perros
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel TherapiesLe Plessis RobinsonFrance
- Paris‐Porto Pulmonary Hypertension Collaborative Laboratory (3PH), UMR_S 999, INSERMUniversité Paris‐SaclayParisFrance
- CarMeN Laboratory, INSERM U1060, INRAE U1397Université Claude Bernard Lyon1Pierre‐BéniteFrance
| |
Collapse
|
5
|
Favoino E, Prete M, Liakouli V, Leone P, Sisto A, Navarini L, Vomero M, Ciccia F, Ruscitti P, Racanelli V, Giacomelli R, Perosa F. Idiopathic and connective tissue disease-associated pulmonary arterial hypertension (PAH): Similarities, differences and the role of autoimmunity. Autoimmun Rev 2024; 23:103514. [PMID: 38181859 DOI: 10.1016/j.autrev.2024.103514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
Pre-capillary pulmonary arterial hypertension (PAH) is hemodynamically characterized by a mean pulmonary arterial pressure (mPAP) ≥ 20 mmHg, pulmonary capillary wedge pressure (PAWP) ≤15 mmHg and pulmonary vascular resistance (PVR) > 2. PAH is classified in six clinical subgroups, including idiopathic PAH (IPAH) and PAH associated to connective tissue diseases (CTD-PAH), that will be the main object of this review. The aim is to compare these two PAH subgroups in terms of epidemiology, histological and pathogenic findings in an attempt to define disease-specific features, including autoimmunity, that may explain the heterogeneity of response to therapy between IPAH and CTD-PAH.
Collapse
Affiliation(s)
- Elvira Favoino
- Laboratory of Cellular and Molecular Immunology, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy.
| | - Marcella Prete
- Internal Medicine Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy
| | - Vasiliki Liakouli
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Patrizia Leone
- Internal Medicine Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy
| | - Adriana Sisto
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy
| | - Luca Navarini
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Marta Vomero
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Francesco Ciccia
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vito Racanelli
- Centre for Medical Sciences, University of Trento and Internal Medicine Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| | - Roberto Giacomelli
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
6
|
Boucetta H, Zhang L, Sosnik A, He W. Pulmonary arterial hypertension nanotherapeutics: New pharmacological targets and drug delivery strategies. J Control Release 2024; 365:236-258. [PMID: 37972767 DOI: 10.1016/j.jconrel.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare, serious, and incurable disease characterized by high lung pressure. PAH-approved drugs based on conventional pathways are still not exhibiting favorable therapeutic outcomes. Drawbacks like short half-lives, toxicity, and teratogenicity hamper effectiveness, clinical conventionality, and long-term safety. Hence, approaches like repurposing drugs targeting various and new pharmacological cascades and/or loaded in non-toxic/efficient nanocarrier systems are being investigated lately. This review summarizes the status of conventional, repurposed, either in vitro, in vivo, and/or in clinical trials of PAH treatment. In-depth description, discussion, and classification of the new pharmacological targets and nanomedicine strategies with a description of all the nanocarriers that showed promising efficiency in delivering drugs are discussed. Ultimately, an illustration of the different nucleic acids tailored and nanoencapsulated within different types of nanocarriers to restore the pathways affected by this disease is presented.
Collapse
Affiliation(s)
- Hamza Boucetta
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
7
|
Tielemans B, Wagenaar A, Belge C, Delcroix M, Quarck R. Pulmonary arterial hypertension drugs can partially restore altered angiogenic capacities in bmpr2-silenced human lung microvascular endothelial cells. Pulm Circ 2023; 13:e12293. [PMID: 37790139 PMCID: PMC10543474 DOI: 10.1002/pul2.12293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/31/2023] [Accepted: 09/17/2023] [Indexed: 10/05/2023] Open
Abstract
Mutations in the bone morphogenetic protein receptor type 2 (bmpr2) gene and signaling pathway impairment are observed in heritable and idiopathic pulmonary arterial hypertension (PAH). In PAH, endothelial dysfunction is currently handled by drugs targeting the endothelin-1 (ET-1), nitric oxide (NO), and prostacyclin (PGI2) pathways. The role of angiogenesis in the disease process and the effect of PAH therapies on dysregulated angiogenesis remain inconclusive. We aim to investigate in vitro whether (i) bmpr2 silencing can impair angiogenic capacity of human lung microvascular endothelial cells (HLMVECs) and (ii) PAH therapies can restore them. The effects of macitentan (ET-1), tadalafil (NO), and selexipag (PGI2), on BMPRII pathway activation, endothelial barrier function, and angiogenesis were investigated in bmpr2-silenced HLMVECs. Stable bmpr2 silencing resulted in impaired migration and tube formation in vitro capacity. Inhibition of ET-1 pathway was able to partially restore tube formation in bmpr2-silenced HLMVECs, whereas none of the therapies was able to restore endothelial barrier function, no deleterious effects were observed. Our findings highlight the potential role of BMPRII signaling pathway in driving pulmonary endothelial cell angiogenesis. In addition, PAH drugs display limited effects on endothelial function when BMPRII is impaired, suggesting that innovative therapeutic strategies targeting BMPRII signaling are needed to better rescue endothelial dysfunction in PAH.
Collapse
Affiliation(s)
- Birger Tielemans
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Biomedical MRI, Department of Imaging and PathologyUniversity of LeuvenLeuvenBelgium
| | - Allard Wagenaar
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)University of LeuvenLeuvenBelgium
| | - Catharina Belge
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Clinical Department of Respiratory Diseases, University HospitalsUniversity of LeuvenLeuvenBelgium
| | - Marion Delcroix
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Clinical Department of Respiratory Diseases, University HospitalsUniversity of LeuvenLeuvenBelgium
| | - Rozenn Quarck
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Clinical Department of Respiratory Diseases, University HospitalsUniversity of LeuvenLeuvenBelgium
| |
Collapse
|
8
|
Quist-Løkken I, Andersson-Rusch C, Kastnes MH, Kolos JM, Jatzlau J, Hella H, Olsen OE, Sundan A, Knaus P, Hausch F, Holien T. FKBP12 is a major regulator of ALK2 activity in multiple myeloma cells. Cell Commun Signal 2023; 21:25. [PMID: 36717825 PMCID: PMC9885706 DOI: 10.1186/s12964-022-01033-9] [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: 06/24/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The immunophilin FKBP12 binds to TGF-β family type I receptors, including the BMP type I receptor ALK2. FKBP12 keeps the type I receptor in an inactive state and controls signaling activity. Removal of FKBP12 with drugs such as the FKBP-ligand FK506 enhances BMP activity in various cell types. In multiple myeloma cells, activation of SMAD1/5/8 leads to apoptosis. We hypothesized that removing FKBP12 from ALK2 in myeloma cells would potentiate BMP-induced ALK2-SMAD1/5/8 activity and in consequence cell death. METHODS Multiple myeloma cell lines were treated with FK506, or other FKBP-binding compounds, combined with different BMPs before analyzing SMAD1/5/8 activity and cell viability. SMAD1/5/8 activity was also investigated using a reporter cell line, INA-6 BRE-luc. To characterize the functional signaling receptor complex, we genetically manipulated receptor expression by siRNA, shRNA and CRISPR/Cas9 technology. RESULTS FK506 potentiated BMP-induced SMAD1/5/8 activation and apoptosis in multiple myeloma cell lines. By using FKBP-binding compounds with different affinity profiles, and siRNA targeting FKBP12, we show that the FK506 effect is mediated by binding to FKBP12. Ligands that typically signal via ALK3 in myeloma cells, BMP2, BMP4, and BMP10, did not induce apoptosis in cells lacking ALK3. Notably, BMP10 competed with BMP6 and BMP9 and antagonized their activity via ALK2. However, upon addition of FK506, we saw a surprising shift in specificity, as the ALK3 ligands gained the ability to signal via ALK2 and induce apoptosis. This indicates that the receptor complex can switch from an inactive non-signaling complex (NSC) to an active one by adding FK506. This gain of activity was also seen in other cell types, indicating that the observed effects have broader relevance. BMP2, BMP4 and BMP10 depended on BMPR2 as type II receptor to signal, which contrasts with BMP6 and BMP9, that activate ALK2 more potently when BMPR2 is knocked down. CONCLUSIONS In summary, our data suggest that FKBP12 is a major regulator of ALK2 activity in multiple myeloma cells, partly by switching an NSC into an active signaling complex. FKBP12 targeting compounds devoid of immunosuppressing activity could have potential in novel treatment strategies aiming at reducing multiple myeloma tumor load. Video Abstract.
Collapse
Affiliation(s)
- Ingrid Quist-Løkken
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Clara Andersson-Rusch
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Martin Haugrud Kastnes
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Centre of Molecular Inflammation Research, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Jürgen Markus Kolos
- grid.6546.10000 0001 0940 1669Department of Chemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jerome Jatzlau
- grid.14095.390000 0000 9116 4836Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Hanne Hella
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Oddrun Elise Olsen
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Anders Sundan
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Centre of Molecular Inflammation Research, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Hematology, St. Olav’s University Hospital, Trondheim, Norway
| | - Petra Knaus
- grid.14095.390000 0000 9116 4836Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Felix Hausch
- grid.6546.10000 0001 0940 1669Department of Chemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Toril Holien
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Immunology and Transfusion Medicine, St. Olav’s University Hospital, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Hematology, St. Olav’s University Hospital, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Department of Biomedical Laboratory Science, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| |
Collapse
|
9
|
Zhao J, Wang Q, Deng X, Qian J, Tian Z, Liu Y, Li M, Zeng X. The treatment strategy of connective tissue disease associated pulmonary arterial hypertension: Evolving into the future. Pharmacol Ther 2022; 239:108192. [DOI: 10.1016/j.pharmthera.2022.108192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
|
10
|
Important Functions and Molecular Mechanisms of Mitochondrial Redox Signaling in Pulmonary Hypertension. Antioxidants (Basel) 2022; 11:antiox11030473. [PMID: 35326123 PMCID: PMC8944689 DOI: 10.3390/antiox11030473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are important organelles that act as a primary site to produce reactive oxygen species (ROS). Additionally, mitochondria play a pivotal role in the regulation of Ca2+ signaling, fatty acid oxidation, and ketone synthesis. Dysfunction of these signaling molecules leads to the development of pulmonary hypertension (PH), atherosclerosis, and other vascular diseases. Features of PH include vasoconstriction and pulmonary artery (PA) remodeling, which can result from abnormal proliferation, apoptosis, and migration of PA smooth muscle cells (PASMCs). These responses are mediated by increased Rieske iron–sulfur protein (RISP)-dependent mitochondrial ROS production and increased mitochondrial Ca2+ levels. Mitochondrial ROS and Ca2+ can both synergistically activate nuclear factor κB (NF-κB) to trigger inflammatory responses leading to PH, right ventricular failure, and death. Evidence suggests that increased mitochondrial ROS and Ca2+ signaling leads to abnormal synthesis of ketones, which play a critical role in the development of PH. In this review, we discuss some of the recent findings on the important interactive role and molecular mechanisms of mitochondrial ROS and Ca2+ in the development and progression of PH. We also address the contributions of NF-κB-dependent inflammatory responses and ketone-mediated oxidative stress due to abnormal regulation of mitochondrial ROS and Ca2+ signaling in PH.
Collapse
|
11
|
Cheron C, McBride SA, Antigny F, Girerd B, Chouchana M, Chaumais MC, Jaïs X, Bertoletti L, Sitbon O, Weatherald J, Humbert M, Montani D. Sex and gender in pulmonary arterial hypertension. Eur Respir Rev 2021; 30:30/162/200330. [PMID: 34750113 DOI: 10.1183/16000617.0330-2020] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterised by pulmonary vascular remodelling and elevated pulmonary pressure, which eventually leads to right heart failure and death. Registries worldwide have noted a female predominance of the disease, spurring particular interest in hormonal involvement in the disease pathobiology. Several experimental models have shown both protective and deleterious effects of oestrogens, suggesting that complex mechanisms participate in PAH pathogenesis. In fact, oestrogen metabolites as well as receptors and enzymes implicated in oestrogen signalling pathways and associated conditions such as BMPR2 mutation contribute to PAH penetrance more specifically in women. Conversely, females have better right ventricular function, translating to a better prognosis. Along with right ventricular adaptation, women tend to respond to PAH treatment differently from men. As some young women suffer from PAH, contraception is of particular importance, considering that pregnancy in patients with PAH is strongly discouraged due to high risk of death. When contraception measures fail, pregnant women need a multidisciplinary team-based approach. This article aims to review epidemiology, mechanisms underlying the higher female predominance, but better prognosis and the intricacies in management of women affected by PAH.
Collapse
Affiliation(s)
- Céline Cheron
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Susan Ainslie McBride
- Internal Medicine Residency Program, Dept of Medicine, University of Calgary, Calgary, Canada
| | - Fabrice Antigny
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Barbara Girerd
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Margot Chouchana
- Assistance Publique Hôpitaux de Paris, Service de Pharmacie Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Marie-Camille Chaumais
- Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,Assistance Publique Hôpitaux de Paris, Service de Pharmacie Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Université Paris-Saclay, Faculté de Pharmacie, Chatenay Malabry, France
| | - Xavier Jaïs
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Laurent Bertoletti
- Centre Hospitalier Universitaire de Saint-Etienne, Service de Médecine Vasculaire et Thérapeutique, Saint-Etienne, France.,INSERM U1059 et CIC1408, Université Jean-Monnet, Saint-Etienne, France
| | - Olivier Sitbon
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Jason Weatherald
- Division of Respirology, Dept of Medicine, University of Calgary, Calgary, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Canada
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France .,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
12
|
Xu B, Xu G, Yu Y, Lin J. The role of TGF-β or BMPR2 signaling pathway-related miRNA in pulmonary arterial hypertension and systemic sclerosis. Arthritis Res Ther 2021; 23:288. [PMID: 34819148 PMCID: PMC8613994 DOI: 10.1186/s13075-021-02678-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/07/2021] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe complication of connective tissue disease (CTD), causing death in systemic sclerosis (SSc). The past decade has yielded many scientific insights into microRNA (miRNAs) in PAH and SSc. This growth of knowledge has well-illustrated the complexity of microRNA (miRNA)-based regulation of gene expression in PAH. However, few miRNA-related SSc-PAH were elucidated. This review firstly discusses the role of transforming growth factor-beta (TGF-β) signaling and bone morphogenetic protein receptor type II (BMPR2) in PAH and SSc. Secondly, the miRNAs relating to TGF-β and BMPR2 signaling pathways in PAH and SSc or merely PAH were subsequently summarized. Finally, future studies might develop early diagnostic biomarkers and target-oriented therapeutic strategies for SSc-PAH and PAH treatment.
Collapse
Affiliation(s)
- Bei Xu
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003
| | - Guanhua Xu
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003
| | - Ye Yu
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003
| | - Jin Lin
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003.
| |
Collapse
|
13
|
BMPR2 promoter methylation and its expression in valvular heart disease complicated with pulmonary artery hypertension. Aging (Albany NY) 2021; 13:24580-24604. [PMID: 34793329 PMCID: PMC8660616 DOI: 10.18632/aging.203690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022]
Abstract
Valvular heart disease (VHD) is a common heart disease that affects blood flow. It usually requires heart surgery. Valvular heart disease complicated with pulmonary artery hypertension (VHD-PAH) may be lethal due to heart failure that results from increased heart burden. It is important for these patients to seek early treatment in order to minimize the heart damage. However, there is no reliable diagnosis method in VHD. In this study, we found DNA methylation was increased at the promoter of BMPR2 gene in the VHD patients compared with the healthy controls. This finding was confirmed by an independent cohort study of VHD patients and healthy controls. In addition, BMPR2 mRNA levels were reduced in the plasma of the VHD patients. There is strong correlation between BMPR2 promoter DNA methylation and the severity of VHD. Indeed, we found that both BMPR2 promoter DNA methylation and BMPR2 mRNA levels in the plasma are good biomarkers of VHD by themselves, with the respective AUC value of 0.879 and 0.725, respectively. When they were used in combination, the diagnostic value was even better, with the AUC value of 0.93. Consistent with the results in the VHD patients, we observed decreased BMPR2 and increased fibrosis in the lung of a PAH model mouse. BMPR2 was also decreased in the hearts of the PAH mice, whereas BMP4 was increased. Furthermore, BMPR2 was reduced in the heart valve tissue samples of human VHD patients after valve replacement with moderate/severe PAH compared with those with mild PAH. There was also increased apoptosis in the hearts of the PAH mice. BMPR2 promoter DNA methylation and its expression appear to be good biomarkers for VHD. Our results also suggest that DNA methylation may cause PAH through deregulation of BMP signaling and increased apoptosis.
Collapse
|
14
|
Lechartier B, Berrebeh N, Huertas A, Humbert M, Guignabert C, Tu L. Phenotypic Diversity of Vascular Smooth Muscle Cells in Pulmonary Arterial Hypertension: Implications for Therapy. Chest 2021; 161:219-231. [PMID: 34391758 DOI: 10.1016/j.chest.2021.08.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive incurable condition that is characterized by extensive remodelling of the pulmonary circulation, leading to severe right heart failure and death. Similar to other vascular contractile cells, pulmonary arterial smooth muscle cells (PA-SMCs) play central roles in physiological and pathological vascular remodelling due to their remarkable ability to dynamically modulate their phenotype to ensure contractile and synthetic functions. The dysfunction and molecular mechanisms underlying their contribution to the various pulmonary vascular lesions associated with PAH have been a major focus of research. The aim of this review is to describe the medial and non-medial origins of contractile cells in the pulmonary vascular wall and present evidence of how they contribute to the onset and progression of PAH. We also highlight specific potential target molecules and discuss future directions that are being explored to widen the therapeutic options for the treatment of PAH.
Collapse
Affiliation(s)
- Benoit Lechartier
- Pulmonary Division, Lausanne University Hospital, Lausanne, Switzerland; Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Nihel Berrebeh
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Alice Huertas
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Ly Tu
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.
| |
Collapse
|
15
|
Novel Genetic and Molecular Pathways in Pulmonary Arterial Hypertension Associated with Connective Tissue Disease. Cells 2021; 10:cells10061488. [PMID: 34199176 PMCID: PMC8231632 DOI: 10.3390/cells10061488] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Pulmonary Arterial Hypertension (PAH) is a severe complication of Connective Tissue Disease (CTD), with remarkable morbidity and mortality. However, the molecular and genetic basis of CTD-PAH remains incompletely understood. This study aimed to screen for genetic defects in a cohort of patients with CTD-PAH, using a PAH-specific panel of 35 genes. During recruitment, 79 patients were studied, including 59 Systemic Sclerosis patients (SSc) and 69 females. Disease-associated variants were observed in nine patients: 4 pathogenic/likely pathogenic variants in 4 different genes (TBX4, ABCC8, KCNA5 and GDF2/BMP9) and 5 Variants of Unknown Significance (VUS) in 4 genes (ABCC8, NOTCH3, TOPBP1 and CTCFL). One patient with mixed CTD had a frameshift pathogenic variant in TBX4. Two patients with SSc-PAH carried variants in ABCC8. A patient diagnosed with Systemic Lupus Erythematous (SLE) presented a pathogenic nonsense variant in GDF2/BMP9. Another patient with SSc-PAH presented a pathogenic variant in KCNA5. Four patients with SSc-PAH carried a VUS in NOTCH1, CTCFL, CTCFL and TOPBP1, respectively. These findings suggest that genetic factors may contribute to Pulmonary Vascular Disease (PVD) in CTD patients.
Collapse
|
16
|
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
|
17
|
Bruni C, Guignabert C, Manetti M, Cerinic MM, Humbert M. The multifaceted problem of pulmonary arterial hypertension in systemic sclerosis. THE LANCET. RHEUMATOLOGY 2021; 3:e149-e159. [PMID: 38279370 DOI: 10.1016/s2665-9913(20)30356-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 01/16/2023]
Abstract
Cardiopulmonary complications are a leading cause of death in systemic sclerosis. Pulmonary hypertension in particular carries a high mortality and morbidity burden. Patients with systemic sclerosis can suffer from all of the clinical groups of pulmonary hypertension, particularly pulmonary arterial hypertension and pulmonary hypertension related to interstitial lung disease. Despite a similar pathogenetic background with idiopathic pulmonary arterial hypertension, different mechanisms determine a worse prognostic outcome for patients with systemic sclerosis. In this Viewpoint, we will consider the link between pathogenetic and potential therapeutic targets for the treatment of pulmonary hypertension in the context of systemic sclerosis, with a focus on the current unmet needs, such as the importance of early screening and detection, the absence of agreed criteria to distinguish pulmonary arterial hypertension with interstitial lung disease from pulmonary hypertension due to lung fibrosis, and the need for a holistic treatment approach to target all the vascular, immunological, and inflammatory components of the disease.
Collapse
Affiliation(s)
- Cosimo Bruni
- Division of Rheumatology, and Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Christophe Guignabert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France; Department of Pulmonary Hypertension, Pathophysiology, and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Mirko Manetti
- Section of Anatomy and Histology, and Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marco Matucci Cerinic
- Division of Rheumatology, and Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France; Department of Pulmonary Hypertension, Pathophysiology, and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
18
|
Ricard N, Bailly S, Guignabert C, Simons M. The quiescent endothelium: signalling pathways regulating organ-specific endothelial normalcy. Nat Rev Cardiol 2021; 18:565-580. [PMID: 33627876 PMCID: PMC7903932 DOI: 10.1038/s41569-021-00517-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Endothelial cells are at the interface between circulating blood and tissues. This position confers on them a crucial role in controlling oxygen and nutrient exchange and cellular trafficking between blood and the perfused organs. The endothelium adopts a structure that is specific to the needs and function of each tissue and organ and is subject to tissue-specific signalling input. In adults, endothelial cells are quiescent, meaning that they are not proliferating. Quiescence was considered to be a state in which endothelial cells are not stimulated but are instead slumbering and awaiting activating signals. However, new evidence shows that quiescent endothelium is fully awake, that it constantly receives and initiates functionally important signalling inputs and that this state is actively regulated. Signalling pathways involved in the maintenance of functionally quiescent endothelia are starting to be identified and are a combination of endocrine, autocrine, paracrine and mechanical inputs. The paracrine pathways confer a microenvironment on the endothelial cells that is specific to the perfused organs and tissues. In this Review, we present the current knowledge of organ-specific signalling pathways involved in the maintenance of endothelial quiescence and the pathologies associated with their disruption. Linking organ-specific pathways and human vascular pathologies will pave the way towards the development of innovative preventive strategies and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Nicolas Ricard
- grid.47100.320000000419368710Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Sabine Bailly
- grid.457348.9Université Grenoble Alpes, INSERM, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France
| | - Christophe Guignabert
- grid.414221.0INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Michael Simons
- grid.47100.320000000419368710Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA ,grid.47100.320000000419368710Department of Cell Biology, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
19
|
Huertas A, Tu L, Humbert M, Guignabert C. Chronic inflammation within the vascular wall in pulmonary arterial hypertension: more than a spectator. Cardiovasc Res 2020; 116:885-893. [PMID: 31813986 DOI: 10.1093/cvr/cvz308] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/08/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
This review seeks to provide an update of preclinical findings and available clinical data on the chronic persistent inflammation and its direct role on the pulmonary arterial hypertension (PAH) progression. We reviewed the different mechanisms by which the inflammatory and immune pathways contribute to the structural and functional changes occurring in the three vascular compartments: the tunica intima, tunica media, and tunica adventitia. We also discussed how these inflammatory mediator changes may serve as a biomarker of the PAH progression and summarize unanswered questions and opportunities for future studies in this area.
Collapse
Affiliation(s)
- Alice Huertas
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France.,Service de Pneumologie, AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France.,Service de Pneumologie, AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
20
|
Brittain EL, Thennapan T, Maron BA, Chan SY, Austin ED, Spiekerkoetter E, Bogaard HJ, Guignabert C, Paulin R, Machado RF, Yu PB. Update in Pulmonary Vascular Disease 2016 and 2017. Am J Respir Crit Care Med 2019. [PMID: 29533671 DOI: 10.1164/rccm.201801-0062up] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Evan L Brittain
- 1 Division of Cardiovascular Medicine, Department of Medicine.,2 Vanderbilt Translational and Clinical Cardiovascular Research Center.,3 Pulmonary Vascular Center, Department of Medicine, and
| | | | - Bradley A Maron
- 5 Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,6 Department of Cardiology, Boston VA Healthcare System, Boston, Massachusetts
| | - Stephen Y Chan
- 7 Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Eric D Austin
- 3 Pulmonary Vascular Center, Department of Medicine, and.,8 Pediatric Pulmonary Hypertension Program, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Edda Spiekerkoetter
- 9 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and.,10 Vera Moulton Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, California
| | - Harm J Bogaard
- 11 Pulmonary Hypertension Expert Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Christophe Guignabert
- 12 INSERM UMR-S 999, Le Plessis-Robinson, France.,13 Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Roxane Paulin
- 14 Quebec Heart and Lung Institute, Laval University, Quebec, Quebec, Canada; and
| | - Roberto F Machado
- 15 Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Paul B Yu
- 5 Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Mercurio V, Bianco A, Campi G, Cuomo A, Diab N, Mancini A, Parrella P, Petretta M, Hassoun PM, Bonaduce D. New Drugs, Therapeutic Strategies, and Future Direction for the Treatment of Pulmonary Arterial Hypertension. Curr Med Chem 2019; 26:2844-2864. [DOI: 10.2174/0929867325666180201095743] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/21/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
Despite recent advances in Pulmonary Arterial Hypertension (PAH) treatment, this condition is still characterized by an extremely poor prognosis. In this review, we discuss the use of newly-approved drugs for PAH treatment with already known mechanisms of action (macitentan), innovative targets (riociguat and selexipag), and novel therapeutic approaches with initial up-front combination therapy. Secondly, we describe new potential signaling pathways and investigational drugs with promising role in the treatment of PAH.
Collapse
Affiliation(s)
- Valentina Mercurio
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Anna Bianco
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Giacomo Campi
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Alessandra Cuomo
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Nermin Diab
- University of Ottawa, Department of Medicine, Ottawa, ON, Canada
| | - Angela Mancini
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paolo Parrella
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Mario Petretta
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paul M. Hassoun
- Johns Hopkins University, Division of Pulmonary and Critical Care Medicine, Baltimore, MD, United States
| | - Domenico Bonaduce
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| |
Collapse
|
22
|
Bertero T, Perk D, Chan SY. The molecular rationale for therapeutic targeting of glutamine metabolism in pulmonary hypertension. Expert Opin Ther Targets 2019; 23:511-524. [PMID: 31055988 DOI: 10.1080/14728222.2019.1615438] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Pulmonary hypertension (PH) is a deadly enigmatic disease with increasing prevalence. Cellular pathologic hallmarks of PH are driven at least partly by metabolic rewiring, but details are just emerging. The discovery that vascular matrix stiffening can mechanically activate the glutaminase (GLS) enzyme and serve as a pathogenic mechanism of PH has advanced our understanding of the complex role of glutamine in PH. It has also offered a novel therapeutic target for development as a next-generation drug for this disease. Area covered: This review discusses the cellular contribution of glutamine metabolism to PH together with the possible therapeutic application of pharmacologic GLS inhibitors in this disease. Expert opinion: Despite advances in our understanding of glutamine metabolism in PH, questions remain unanswered regarding the development of therapies targeting glutamine in PH. The comprehensive mechanisms by which glutamine metabolism rewiring influences pulmonary vascular cell behavior to drive PH are incompletely understood. Because glutamine metabolism exhibits a variety of functions in organ repair and homeostasis, a better understanding of the overall risk-benefit ratio of these strategies with long-term follow-up is needed. This knowledge should pave the way for the design of new strategies to prevent and hopefully even regress PH.
Collapse
Affiliation(s)
- Thomas Bertero
- a Institute of Molecular and Cellular Pharmacology , Université Côte d'Azur , Valbonne , France
| | - Dror Perk
- b Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine , University of Pittsburgh Medical Center , Pittsburgh , PA , USA
| | - Stephen Y Chan
- b Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine , University of Pittsburgh Medical Center , Pittsburgh , PA , USA
| |
Collapse
|
23
|
Guignabert C, Tu L, Feige JJ, Humbert M, Bailly S. Response by Guignabert et al to Letter Regarding Article, “Selective BMP-9 Inhibition Partially Protects Against Experimental Pulmonary Hypertension”. Circ Res 2019; 124:e82-e83. [DOI: 10.1161/circresaha.119.315053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Christophe Guignabert
- INSERM UMR_S 999, Université Paris-Sud, Université Paris-Saclay, AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S 999, Université Paris-Sud, Université Paris-Saclay, AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Jean-Jacques Feige
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France
| | - Marc Humbert
- INSERM UMR_S 999, Université Paris-Sud, Université Paris-Saclay, AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Sabine Bailly
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France
| |
Collapse
|
24
|
Bordenave J, Tu L, Savale L, Huertas A, Humbert M, Guignabert C. [New insights in the pathogenesis of pulmonary arterial hypertension]. Rev Mal Respir 2019; 36:433-437. [PMID: 31010759 DOI: 10.1016/j.rmr.2019.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 11/26/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a severe and incurable cardiopulmonary disorder. Research from the past 10 years illustrates the complex and multifactorial aspects of PAH pathophysiology. Furthermore, latest advances in the field have led to a better understanding of the key components underlying this inadequate accumulation of pulmonary vascular cells within the pulmonary arterial walls, leading to pulmonary vascular remodelling. Among the underlying molecular and cellular mechanisms, pulmonary endothelial dysfunction, alterations of the inter-cell communications within the pulmonary arterial walls as well as defects of the inflammatory component and the loss of BMPRII activity play critical roles in the pathogenesis of the disease.
Collapse
Affiliation(s)
- J Bordenave
- Inserm UMR_S 999, hôpital Marie Lannelongue, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France; Faculté de Médecine, université Paris-Sud, université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| | - L Tu
- Inserm UMR_S 999, hôpital Marie Lannelongue, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France; Faculté de Médecine, université Paris-Sud, université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| | - L Savale
- Inserm UMR_S 999, hôpital Marie Lannelongue, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France; Faculté de Médecine, université Paris-Sud, université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; Service de pneumologie, centre de référence de l'hypertension pulmonaire sévère, DHU Thorax Innovation, hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - A Huertas
- Inserm UMR_S 999, hôpital Marie Lannelongue, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France; Faculté de Médecine, université Paris-Sud, université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; Service de pneumologie, centre de référence de l'hypertension pulmonaire sévère, DHU Thorax Innovation, hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - M Humbert
- Inserm UMR_S 999, hôpital Marie Lannelongue, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France; Faculté de Médecine, université Paris-Sud, université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; Service de pneumologie, centre de référence de l'hypertension pulmonaire sévère, DHU Thorax Innovation, hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - C Guignabert
- Inserm UMR_S 999, hôpital Marie Lannelongue, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France; Faculté de Médecine, université Paris-Sud, université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France.
| |
Collapse
|
25
|
Tu L, Desroches-Castan A, Mallet C, Guyon L, Cumont A, Phan C, Robert F, Thuillet R, Bordenave J, Sekine A, Huertas A, Ritvos O, Savale L, Feige JJ, Humbert M, Bailly S, Guignabert C. Selective BMP-9 Inhibition Partially Protects Against Experimental Pulmonary Hypertension. Circ Res 2019; 124:846-855. [DOI: 10.1161/circresaha.118.313356] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Ly Tu
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
| | - Agnès Desroches-Castan
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France (A.D.-C., C.M., L.G., F.R., J.-J.F., S.B.)
| | - Christine Mallet
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France (A.D.-C., C.M., L.G., F.R., J.-J.F., S.B.)
| | - Laurent Guyon
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France (A.D.-C., C.M., L.G., F.R., J.-J.F., S.B.)
| | - Amélie Cumont
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
| | - Carole Phan
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
| | - Florian Robert
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France (A.D.-C., C.M., L.G., F.R., J.-J.F., S.B.)
| | - Raphaël Thuillet
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
| | - Jennifer Bordenave
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
| | - Ayumi Sekine
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (A.S., A.H., L.S., M.H.)
| | - Alice Huertas
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (A.S., A.H., L.S., M.H.)
| | - Olli Ritvos
- Department of Bacteriology and Immunology and Department of Physiology, Faculty of Medicine, University of Helsinki, Finland (O.R.)
| | - Laurent Savale
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (A.S., A.H., L.S., M.H.)
| | - Jean-Jacques Feige
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France (A.D.-C., C.M., L.G., F.R., J.-J.F., S.B.)
| | - Marc Humbert
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (A.S., A.H., L.S., M.H.)
| | - Sabine Bailly
- Université Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France (A.D.-C., C.M., L.G., F.R., J.-J.F., S.B.)
| | - Christophe Guignabert
- From the INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, France (L.T., A.C., C.P., R.T., J.B., A.S., A.H., L.S., M.H., C.G.)
| |
Collapse
|
26
|
Lambert M, Capuano V, Olschewski A, Sabourin J, Nagaraj C, Girerd B, Weatherald J, Humbert M, Antigny F. Ion Channels in Pulmonary Hypertension: A Therapeutic Interest? Int J Mol Sci 2018; 19:ijms19103162. [PMID: 30322215 PMCID: PMC6214085 DOI: 10.3390/ijms19103162] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K⁺, Ca2+, Na⁺ and Cl-). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Véronique Capuano
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
- Department of Physiology, Medical University Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria.
| | - Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, UMRS 1180, Univ. Paris-Sud, INSERM, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
| | - Barbara Girerd
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Jason Weatherald
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
- Division of Respirology, Department of Medicine, University of Calgary, Calgary, AB T1Y 6J4, Canada.
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB T1Y 6J4, Canada.
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| |
Collapse
|
27
|
Hu CJ, Zhang H, Laux A, Pullamsetti SS, Stenmark KR. Mechanisms contributing to persistently activated cell phenotypes in pulmonary hypertension. J Physiol 2018; 597:1103-1119. [PMID: 29920674 PMCID: PMC6375873 DOI: 10.1113/jp275857] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/16/2018] [Indexed: 12/24/2022] Open
Abstract
Chronic pulmonary hypertension (PH) is characterized by the accumulation of persistently activated cell types in the pulmonary vessel exhibiting aberrant expression of genes involved in apoptosis resistance, proliferation, inflammation and extracellular matrix (ECM) remodelling. Current therapies for PH, focusing on vasodilatation, do not normalize these activated phenotypes. Furthermore, current approaches to define additional therapeutic targets have focused on determining the initiating signals and their downstream effectors that are important in PH onset and development. Although these approaches have produced a large number of compelling PH treatment targets, many promising human drugs have failed in PH clinical trials. Herein, we propose that one contributing factor to these failures is that processes important in PH development may not be good treatment targets in the established phase of chronic PH. We hypothesize that this is due to alterations of chromatin structure in PH cells, resulting in functional differences between the same factor or pathway in normal or early PH cells versus cells in chronic PH. We propose that the high expression of genes involved in the persistently activated phenotype of PH vascular cells is perpetuated by an open chromatin structure and multiple transcription factors (TFs) via the recruitment of high levels of epigenetic regulators including the histone acetylases P300/CBP, histone acetylation readers including BRDs, the Mediator complex and the positive transcription elongation factor (Abstract figure). Thus, determining how gene expression is controlled by examining chromatin structure, TFs and epigenetic regulators associated with aberrantly expressed genes in pulmonary vascular cells in chronic PH, may uncover new PH therapeutic targets.
![]()
Collapse
Affiliation(s)
- Cheng-Jun Hu
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aya Laux
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Soni S Pullamsetti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Bad Nauheim, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), member of the DZL, Justus-Liebig University, Giessen, Germany
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
28
|
Savale L, Guignabert C, Weatherald J, Humbert M. Precision medicine and personalising therapy in pulmonary hypertension: seeing the light from the dawn of a new era. Eur Respir Rev 2018; 27:27/148/180004. [PMID: 29653948 PMCID: PMC9488842 DOI: 10.1183/16000617.0004-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023] Open
Abstract
Pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH) include different cardiopulmonary disorders in which the interaction of multiple genes with environmental and behavioural factors modulates the onset and the progression of these severe conditions. Although the development of therapeutic agents that modulate abnormalities in three major pathobiological pathways for PAH has revolutionised our approach to the treatment of PAH, the long-term survival rate remains unsatisfactory. Accumulating evidence has underlined that clinical outcomes and responses to therapy in PAH are modified by multiple factors, including genetic variations, which will be different for each individual. Since precision medicine, also known as stratified medicine or personalised medicine, aims to better target intervention to the individual while maximising benefit and minimising harm, it has significant potential advantages. This article aims to assemble and discuss the different initiatives that are currently underway in the PH/PAH fields together with the opportunities and prospects for their use in the near future. Development of precision medicine strategies will be the next frontier in the evolution of PAH treatmenthttp://ow.ly/8T8730j7e36
Collapse
|
29
|
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
|
30
|
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
|
31
|
Nie X, Tan J, Dai Y, Liu Y, Zou J, Sun J, Ye S, Shen C, Fan L, Chen J, Bian JS. CCL5 deficiency rescues pulmonary vascular dysfunction, and reverses pulmonary hypertension via caveolin-1-dependent BMPR2 activation. J Mol Cell Cardiol 2018; 116:41-56. [PMID: 29374556 DOI: 10.1016/j.yjmcc.2018.01.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 12/13/2017] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating cardiopulmonary disorder characterized by pulmonary arterial remodeling mainly due to excess cellular proliferation and apoptosis resistance of pulmonary arterial smooth muscle cells (PASMCs). Reduced bone morphogenetic protein receptor 2 (BMPR2) expression in patients with PAH impairs pulmonary arterial endothelial cells (PAECs) function. This can adversely affect PAEC survival and promote PASMCs proliferation. We hypothesized that interventions to normalize the expression of genes that are targets of the BMPR2 signaling could restore PAECs function and prevent or reverse PAH. Here we characterized for the first time, in human PAECs, chemokine (C-C motif) ligand 5 (CCL5/RANTES) deficiency restore BMP-mediated PAECs function. In the cell culture experiments, we found that CCL5 deficiency increased apoptosis and tube formation of PAECs, but suppressed proliferation and migration of PASMCs. Silencing CCL5 expression in PAH PAECs restored bone morphogenetic protein (BMP) signaling responses and promoted phosphorylation of SMADs and transcription of ID genes. Moreover, CCL5 deficiency inhibited angiogenesis by increasing pSMAD-dependent and-independent BMPR2 signaling. This was linked mechanistically to enhanced interaction of BMPR2 with caveolin-1 via CCL5 deficiency-mediated stabilization of endothelial surface caveolin-1. Consistent with these functions, deletion of CCL5 significantly attenuated development of Sugen5416/hypoxia-induced PAH by restoring BMPR2 signaling in mice. Taken together, our findings suggest that CCL5 deficiency could reverse obliterative changes in pulmonary arteries via caveolin-1-dependent amplification of BMPR2 signaling. Our results shed light on better understanding of the disease pathobiology and provide a possible novel target for the treatment of PAH.
Collapse
Affiliation(s)
- Xiaowei Nie
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China.
| | - Jianxin Tan
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Youai Dai
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Yun Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222000, PR China
| | - Jian Zou
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Jie Sun
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Shugao Ye
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Chenyou Shen
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Li Fan
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Jingyu Chen
- Center of Clincical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
32
|
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
|
33
|
Suzuki T, Carrier EJ, Talati MH, Rathinasabapathy A, Chen X, Nishimura R, Tada Y, Tatsumi K, West J. Isolation and characterization of endothelial-to-mesenchymal transition cells in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2017; 314:L118-L126. [PMID: 28935639 DOI: 10.1152/ajplung.00296.2017] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) is a process in which endothelial cells lose polarity and cell-to cell contacts, and undergo a dramatic remodeling of the cytoskeleton. It has been implicated in initiation and progression of pulmonary arterial hypertension (PAH). However, the characteristics of cells which have undergone EndMT cells in vivo have not been reported and so remain unclear. To study this, sugen5416 and hypoxia (SuHx)-induced PAH was established in Cdh5-Cre/Gt(ROSA)26Sortm4(ACTB-tdTomato,EGFP)Luo/J double transgenic mice, in which GFP was stably expressed in pan-endothelial cells. After 3 wk of SuHx, flow cytometry and immunohistochemistry demonstrated CD144-negative and GFP-positive cells (complete EndMT cells) possessed higher proliferative and migratory activity compared with other mesenchymal cells. While CD144-positive and α-smooth muscle actin (α-SMA)-positive cells (partial EndMT cells) continued to express endothelial progenitor cell markers, complete EndMT cells were Sca-1-rich mesenchymal cells with high proliferative and migratory ability. When transferred in fibronectin-coated chamber slides containing smooth muscle media, α-SMA robustly expressed in these cells compared with cEndMT cells that were grown in maintenance media. Demonstrating additional paracrine effects, conditioned medium from isolated complete EndMT cells induced enhanced mesenchymal proliferation and migration and increased angiogenesis compared with conditioned medium from resident mesenchymal cells. Overall, these findings show that EndMT cells could contribute to the pathogenesis of PAH both directly, by transformation into smooth muscle-like cells with higher proliferative and migratory potency, and indirectly, through paracrine effects on vascular intimal and medial proliferation.
Collapse
Affiliation(s)
- Toshio Suzuki
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Erica J Carrier
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Megha H Talati
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Anandharajan Rathinasabapathy
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Xinping Chen
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Rintaro Nishimura
- Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan.,Department of Advanced Medicine in Pulmonary Hypertension, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Yuji Tada
- Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - James West
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| |
Collapse
|
34
|
|
35
|
BMP type II receptor as a therapeutic target in pulmonary arterial hypertension. Cell Mol Life Sci 2017; 74:2979-2995. [PMID: 28447104 PMCID: PMC5501910 DOI: 10.1007/s00018-017-2510-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/09/2017] [Accepted: 03/17/2017] [Indexed: 12/30/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disease characterized by a progressive elevation in mean pulmonary arterial pressure. This occurs due to abnormal remodeling of small peripheral lung vasculature resulting in progressive occlusion of the artery lumen that eventually causes right heart failure and death. The most common cause of PAH is inactivating mutations in the gene encoding a bone morphogenetic protein type II receptor (BMPRII). Current therapeutic options for PAH are limited and focused mainly on reversal of pulmonary vasoconstriction and proliferation of vascular cells. Although these treatments can relieve disease symptoms, PAH remains a progressive lethal disease. Emerging data suggest that restoration of BMPRII signaling in PAH is a promising alternative that could prevent and reverse pulmonary vascular remodeling. Here we will focus on recent advances in rescuing BMPRII expression, function or signaling to prevent and reverse pulmonary vascular remodeling in PAH and its feasibility for clinical translation. Furthermore, we summarize the role of described miRNAs that directly target the BMPR2 gene in blood vessels. We discuss the therapeutic potential and the limitations of promising new approaches to restore BMPRII signaling in PAH patients. Different mutations in BMPR2 and environmental/genetic factors make PAH a heterogeneous disease and it is thus likely that the best approach will be patient-tailored therapies.
Collapse
|