1
|
Wu H, Liu Q, Shen Y, Qi H, Zhao J, Li Q, Xia H, Xu RA, Shi L. Drug Interactions of Imperatorin and Curcumin on Macitentan in vitro and in vivo. Drug Des Devel Ther 2025; 19:3459-3475. [PMID: 40322036 PMCID: PMC12049132 DOI: 10.2147/dddt.s505960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose The purpose of this study was to establish an in vitro incubation system and an in vivo model to investigate the potential kinetic interactions of macitentan with imperatorin and curcumin, and to validate the potential inhibitory mechanisms using molecular docking. Methods In vitro, the enzyme kinetic profile of macitentan was explored in rat liver microsomes (RLM) and human liver microsomes (HLM). Furthermore, molecular docking technique was used to study the sites of action of macitentan, imperatorin, and curcumin with CYP 3A4. In vivo, the pharmacokinetic parameters of macitentan were investigated in Sprague-Dawley (SD) rats administered the drug orally, both as a single agent and in combination with imperatorin and curcumin. Results In vitro, the results indicated that imperatorin and curcumin could inhibit the metabolism of macitentan, with IC50 values of 6.58 μM and 10.86 μM in RLM and 6.97 μM and 5.71 μM in HLM, respectively. And in the study of inhibition type, in RLM, the inhibition types of imperatorin and curcumin on macitentan were mixed and non-competitive, respectively; in HLM, the inhibition types of imperatorin and curcumin on macitentan were both mixed. Furthermore, additional molecular docking studies demonstrated that both imperatorin and curcumin occupied the CYP3A4 site. In vivo, the result showed significant increases in AUC(0-t), AUC(0-∞), Tmax, t1/2, and Cmax for macitentan while a decrease in CLz/F when combined with imperatorin. The metabolite ACT-132577 exhibited substantial increases in t1/2, Tmax, and Cmax. Combined with curcumin, the AUC(0-∞) and Tmax of macitentan were significantly increased, while CLz/F was significantly decreased. Conversely, the metabolite ACT-132577 exhibited a substantial decrease in CLz/F, accompanied by notable increases in AUC(0-∞) and Tmax. Conclusion In vitro and in vivo studies revealed that imperatorin and curcumin exhibited inhibitory effects on the metabolism of macitentan. Furthermore, molecular docking revealed that the metabolic inhibition of macitentan by imperatorin and curcumin occurred through binding to the site on CYP3A4. However, further investigation is necessary to ascertain whether this phenomenon will occur in humans.
Collapse
Affiliation(s)
- Hualu Wu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Qian Liu
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, People’s Republic of China
| | - Yuxin Shen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - He Qi
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Jiade Zhao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Qiaoying Li
- Lishui Liandu District Adverse Drug Reaction Monitoring Station, Lishui, Zhejiang, People’s Republic of China
| | - Hailun Xia
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Ren-Ai Xu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Lu Shi
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
2
|
Batool Z, Amjad Kamal M, Shen B. Advanced treatment strategies for high-altitude pulmonary hypertension employing natural medicines: A review. J Pharm Anal 2025; 15:101129. [PMID: 40161446 PMCID: PMC11953983 DOI: 10.1016/j.jpha.2024.101129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 04/02/2025] Open
Abstract
High-altitude pulmonary hypertension (HAPH) occurs when blood pressure in the pulmonary arteries rises due to exposure to high altitudes above 2,500 m. At these elevations, reduced atmospheric pressure leads to lower oxygen levels, triggering a series of physiological responses, including pulmonary artery constriction, which elevates blood pressure. This review explored the complex pathophysiological mechanisms of HAPH and reviewed current pharmaceutical interventions for its management. Meanwhile, this review particularly emphasized on the emerging research concerning Chinese medicinal plants as potential treatments for HAPH. Traditional Chinese medicines are rich in diverse natural ingredients that show significant promise in alleviating HAPH symptoms. We reviewed both in vitro and in vivo studies to assess the efficacy, safety, and mechanisms of these natural medicines, along with their potential adverse effects. Additionally, this review highlighted new alternative natural remedies, underscoring the need for ongoing research to expand available treatment options for HAPH.
Collapse
Affiliation(s)
- Zahra Batool
- Center of High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mohammad Amjad Kamal
- Center of High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh
- Centre for Global Health Research, Saveetha Medical College and Hospital, Chennai, Tamil Nadu, 600001, India
| | - Bairong Shen
- Center of High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
Corboz MR, Nguyen TL, Stautberg A, Cipolla D, Perkins WR, Chapman RW. Current Overview of the Biology and Pharmacology in Sugen/Hypoxia-Induced Pulmonary Hypertension in Rats. J Aerosol Med Pulm Drug Deliv 2024; 37:241-283. [PMID: 39388691 PMCID: PMC11502635 DOI: 10.1089/jamp.2024.0016] [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: 04/03/2024] [Accepted: 07/03/2024] [Indexed: 10/12/2024] Open
Abstract
The Sugen 5416/hypoxia (Su/Hx) rat model of pulmonary arterial hypertension (PAH) demonstrates most of the distinguishing features of PAH in humans, including increased wall thickness and obstruction of the small pulmonary arteries along with plexiform lesion formation. Recently, significant advancement has been made describing the epidemiology, genomics, biochemistry, physiology, and pharmacology in Su/Hx challenge in rats. For example, there are differences in the overall reactivity to Su/Hx challenge in different rat strains and only female rats respond to estrogen treatments. These conditions are also encountered in human subjects with PAH. Also, there is a good translation in both the biochemical and metabolic pathways in the pulmonary vasculature and right heart between Su/Hx rats and humans, particularly during the transition from the adaptive to the nonadaptive phase of right heart failure. Noninvasive techniques such as echocardiography and magnetic resonance imaging have recently been used to evaluate the progression of the pulmonary vascular and cardiac hemodynamics, which are important parameters to monitor the efficacy of drug treatment over time. From a pharmacological perspective, most of the compounds approved clinically for the treatment of PAH are efficacious in Su/Hx rats. Several compounds that show efficacy in Su/Hx rats have advanced into phase II/phase III studies in humans with positive results. Results from these drug trials, if successful, will provide additional treatment options for patients with PAH and will also further validate the excellent translation that currently exists between Su/Hx rats and the human PAH condition.
Collapse
|
4
|
Williams TL, Nyimanu D, Kuc RE, Foster R, Glen RC, Maguire JJ, Davenport AP. The biased apelin receptor agonist, MM07, reverses Sugen/hypoxia-induced pulmonary arterial hypertension as effectively as the endothelin antagonist macitentan. Front Pharmacol 2024; 15:1369489. [PMID: 38655187 PMCID: PMC11035786 DOI: 10.3389/fphar.2024.1369489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction: Pulmonary arterial hypertension (PAH) is characterised by endothelial dysfunction and pathological vascular remodelling, resulting in the occlusion of pulmonary arteries and arterioles, right ventricular hypertrophy, and eventually fatal heart failure. Targeting the apelin receptor with the novel, G protein-biased peptide agonist, MM07, is hypothesised to reverse the developed symptoms of elevated right ventricular systolic pressure and right ventricular hypertrophy. Here, the effects of MM07 were compared with the clinical standard-of-care endothelin receptor antagonist macitentan. Methods: Male Sprague-Dawley rats were randomised and treated with either normoxia/saline, or Sugen/hypoxia (SuHx) to induce an established model of PAH, before subsequent treatment with either saline, macitentan (30 mg/kg), or MM07 (10 mg/kg). Rats were then anaesthetised and catheterised for haemodynamic measurements, and tissues collected for histopathological assessment. Results: The SuHx/saline group presented with significant increases in right ventricular hypertrophy, right ventricular systolic pressure, and muscularization of pulmonary arteries compared to normoxic/saline controls. Critically, MM07 was as at least as effective as macitentan in significantly reversing detrimental structural and haemodynamic changes after 4 weeks of treatment. Discussion: These results support the development of G protein-biased apelin receptor agonists with improved pharmacokinetic profiles for use in human disease.
Collapse
Affiliation(s)
- Thomas L. Williams
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Duuamene Nyimanu
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Rhoda E. Kuc
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Richard Foster
- School of Chemistry, Astbury Centre for Structural Biology, University of Leeds, Leeds, United Kingdom
| | - Robert C. Glen
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Cambridge, United Kingdom
- Department of Surgery and Cancer, Biomolecular Medicine, Imperial College London, London, United Kingdom
| | - Janet J. Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Anthony P. Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
5
|
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
|
6
|
Albinni S, Heno J, Pavo I, Kitzmueller E, Marx M, Michel-Behnke I. Macitentan in the Young-Mid-term Outcomes of Patients with Pulmonary Hypertensive Vascular Disease treated in a Pediatric Tertiary Care Center. Paediatr Drugs 2023; 25:467-481. [PMID: 37269500 PMCID: PMC10284929 DOI: 10.1007/s40272-023-00573-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a severe hemodynamic condition with high morbidity and mortality. Approved targeted therapies are limited for pediatric subjects, and treatments are widely adopted from adult algorithms. Macitentan is a safe and effective drug used for adult PH, but data on pediatric patients are limited. In this prospective single-center study, we investigated mid- and long-term effects of macitentan in children with advanced pulmonary hypertensive vascular disease. METHODS Twenty-four patients were enrolled in the study for treatment with macitentan. Efficacy was determined by echo parameters and brain natriuretic peptide levels (BNP) at 3 months and 1 year. For detailed analysis, the entire cohort was subgrouped into patients with congenital heart disease-related PH (CHD-PH) and non-CHD-PH patients, respectively. RESULTS Mean age of the patients was 10.7 ± 7.6 years; median observation period was 36 months. Twenty of 24 patients were on additional sildenafil and/or prostacyclins. Two of 24 patients discontinued because of peripheral edema. Within the entire cohort, BNP levels and all echo measures such as right ventricular systolic pressure (RVSP), right ventricular end-diastolic diameter (RVED), tricuspid annular plane systolic excursion (TAPSE), pulmonary velocity time integral (VTI), and pulmonary artery acceleration time (PAAT) improved significantly after 3 months (p ≤ 0.01), whereas in the long term significant improvement persisted for BNP levels (-16%), VTI (+14%) and PAAT (+11%) (p < 0.05). By subgroup analysis, non-CHD PH patients showed significant improvements in BNP levels (-57%) and all echo measures (TAPSE +21%, VTI +13%, PAAT +37%, RVSP -24%, RVED -12%) at 3 months (p ≤ 0.01), whereas at 12 months, improvements persisted (p < 0.05) except for RVSP and RVED (nonsignificant). In CHD-PH patients, none of the measures changed (nonsignificant). 6-MWD (distance walked in 6 minutes) slightly increased but was not statistically evaluated. CONCLUSION Data presented herein account for the largest cohort of severely affected pediatric patients receiving macitentan. Overall, macitentan was safe and associated with significant beneficial effects and sustained positive signals after 1 year, albeit in the long term disease progression remains a major concern. Our data suggest limited efficacy in CHD-related PH, whereas favorable outcomes were mainly driven by improvements in patients with PH not related to CHD. Larger studies are needed to verify these preliminary results and to prove efficacy of this drug in different pediatric PH entities.
Collapse
Affiliation(s)
- Sulaima Albinni
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Cardiology, Pediatric Heart Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Julian Heno
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Cardiology, Pediatric Heart Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Imre Pavo
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Cardiology, Pediatric Heart Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Erwin Kitzmueller
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Cardiology, Pediatric Heart Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Manfred Marx
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Cardiology, Pediatric Heart Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Ina Michel-Behnke
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Cardiology, Pediatric Heart Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| |
Collapse
|
7
|
Murphy G, Jayasekera G, Mullin J, Gallagher L, Welsh DJ. Exploring the failing right ventricle in pulmonary hypertension by cardiac magnetic resonance: An in vivo study utilizing Macitentan. Pulm Circ 2022; 12:e12124. [PMID: 36092794 PMCID: PMC9438403 DOI: 10.1002/pul2.12124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2022] [Accepted: 07/30/2022] [Indexed: 11/24/2022] Open
Abstract
Cardiac magnetic resonance (CMR) imaging is used to assess the right ventricle (RV) of pulmonary hypertensive (PH) patients and more recently to track changes in response to therapy. We wished to investigate if repeat CMRs could be used to assess ventricular changes in the Sugen 5416 hypoxic (Su/Hx) rat model of PH treated with the dual endothelin receptor antagonist Macitentan. Male Sprague Dawley Su/Hx rats were dosed for 3 weeks with either vehicle or Macitentan (30 mg/kg) daily, control rats received only vehicle. All rats underwent three CMR scans; before treatment, 2 weeks into treatment, and end of the study. A separate group of Su/Hx and control rats, treated as above, underwent terminal hemodynamic measurements. Using terminal and CMR measurements, Macitentan was found to lower RV systolic pressure pulmonary artery remodeling and increase RV ejection fraction but not change RV hypertrophy (RVH). Repeat CMRs determined that Su/Hx rats treated with Macitentan had significantly reversed RVH via reducing RV mass as well as reducing elevated left ventricular eccentricity index; reductions in RV mass were also observed in Su/Hx vehicle rats exposed to normoxic conditions. We have demonstrated that repeat CMRs can be used to assess the volume and structural changes in the ventricles of the Su/Hx rat model. Using repeat CMRs has allowed us to build a more complete picture of the response of the RV and the left ventricle to treatment. It is unknown if these effects are a consequence of direct action on the RV or secondary to improvements in the lung vasculature.
Collapse
Affiliation(s)
- Gerard Murphy
- Scottish Pulmonary Vascular UnitGlasgow Caledonian UniversityGlasgowUK
| | | | - James Mullin
- Institute of Neuroscience & PsychologyUniversity of GlasgowGlasgowUK
| | - Lindsay Gallagher
- Institute of Neuroscience & PsychologyUniversity of GlasgowGlasgowUK
| | - David J. Welsh
- Scottish Pulmonary Vascular UnitGlasgow Caledonian UniversityGlasgowUK
| |
Collapse
|
8
|
Baumann P, Greco F, Wiegert S, Wellmann S, Pellegrini G, Cannizzaro V. Macitentan attenuates cardiovascular remodelling in infant rats with chronic lung disease. J Transl Med 2022; 20:77. [PMID: 35123510 PMCID: PMC8818179 DOI: 10.1186/s12967-022-03281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cardiovascular impairment contributes to increased mortality in preterm infants with chronic lung disease. Macitentan, an endothelin-1 receptor antagonist, has the potential to attenuate pulmonary and cardiovascular remodelling.
Methods
In a prospective randomized placebo-controlled intervention trial, Sprague–Dawley rats were exposed to 0.21 or 1.0 fraction of inspired oxygen (FiO2) for 19 postnatal days. Rats were treated via gavage with placebo or macitentan from days of life 5 to 19. Alveoli, pulmonary vessels, α-smooth muscle actin content in pulmonary arterioles, size of cardiomyocytes, right to left ventricular wall diameter ratio, and endothelin-1 plasma concentrations were assessed.
Results
FiO2 1.0 induced typical features of chronic lung disease with significant alveolar enlargement (p = 0.012), alveolar (p = 0.048) and pulmonary vessel rarefaction (p = 0.024), higher α-smooth muscle actin content in pulmonary arterioles (p = 0.009), higher right to left ventricular wall diameter ratio (p = 0.02), and larger cardiomyocyte cross-sectional area (p < 0.001). Macitentan treatment significantly increased pulmonary vessel count (p = 0.004) and decreased right to left ventricular wall diameter ratios (p = 0.002). Endothelin-1 plasma concentrations were higher compared to placebo (p = 0.015). Alveolar number and size, α-smooth muscle actin, and the cardiomyocyte cross-sectional area remained unchanged (all p > 0.05).
Conclusion
The endothelin-1 receptor antagonist macitentan attenuated cardiovascular remodelling in an infant rat model for preterm chronic lung disease. This study underscores the potential of macitentan to reduce cardiovascular morbidity in preterm infants with chronic lung disease.
Collapse
|
9
|
Botros L, Jansen SM, Ashek A, Spruijt OA, Tramper J, Noordegraaf AV, Aman J, Harms H, de Man FS, Huisman MC, Zhao L, Bogaard HJ. Application of [18F]FLT-PET in pulmonary arterial hypertension: a clinical study in pulmonary arterial hypertension patients and unaffected bone morphogenetic protein receptor type 2 mutation carriers. Pulm Circ 2021; 11:20458940211028017. [PMID: 34276963 PMCID: PMC8256252 DOI: 10.1177/20458940211028017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/05/2021] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension is a heterogeneous group of diseases characterized by vascular cell proliferation leading to pulmonary vascular remodelling and ultimately right heart failure. Previous data indicated that 3'-deoxy-3'-[18F]-fluorothymidine (18FLT) positron emission tomography (PET) scanning was increased in pulmonary arterial hypertension patients, hence providing a possible biomarker for pulmonary arterial hypertension as it reflects vascular cell hyperproliferation in the lung. This study sought to validate 18FLT-PET in an expanded cohort of pulmonary arterial hypertension patients in comparison to matched healthy controls and unaffected bone morphogenetic protein receptor type 2 mutation carriers. 18FLT-PET scanning was performed in 21 pulmonary arterial hypertension patients (15 hereditary pulmonary arterial hypertension and 6 idiopathic pulmonary arterial hypertension), 11 unaffected mutation carriers and 9 healthy control subjects. In-depth kinetic analysis indicated that there were no differences in lung 18FLT k3 phosphorylation among pulmonary arterial hypertension patients, unaffected bone morphogenetic protein receptor type 2 mutation carriers and healthy controls. Lung 18FLT uptake did not correlate with haemodynamic or clinical parameters in pulmonary arterial hypertension patients. Sequential 18FLT-PET scanning in three patients demonstrated uneven regional distribution in 18FLT uptake by 3D parametric mapping of the lung, although this did not follow the clinical course of the patient. We did not detect significantly increased lung 18FLT uptake in pulmonary arterial hypertension patients, nor in the unaffected bone morphogenetic protein receptor type 2 mutation carriers, as compared to healthy subjects. The conflicting results with our preliminary human 18FLT report may be explained by a small sample size previously and we observed large variation of lung 18FLT signals between patients, challenging the application of 18FLT-PET as a biomarker in the pulmonary arterial hypertension clinic.
Collapse
Affiliation(s)
- Liza Botros
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Samara M.A. Jansen
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Ali Ashek
- Faculty of MedicineNational Heart and Lung InstituteImperial College LondonHammersmith HospitalLondonUK
| | - Onno A. Spruijt
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Jelco Tramper
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Anton V. Noordegraaf
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Jurjan Aman
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Hans Harms
- Cardiovascular Imaging ProgramDepartments of Radiology and Medicine; Division of Nuclear Medicine and Molecular ImagingBrigham and Women’s Hospitaland Harvard Medical SchoolBostonMAUSA
- Institute of Clinical MedicineAarhus University HospitalAarhusDenmark
| | - Frances S. de Man
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Marc C. Huisman
- Department of Radiology and Nuclear MedicineAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Lan Zhao
- Faculty of MedicineNational Heart and Lung InstituteImperial College LondonHammersmith HospitalLondonUK
| | - Harm J. Bogaard
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
10
|
Malenfant S, Lebret M, Breton-Gagnon É, Potus F, Paulin R, Bonnet S, Provencher S. Exercise intolerance in pulmonary arterial hypertension: insight into central and peripheral pathophysiological mechanisms. Eur Respir Rev 2021; 30:200284. [PMID: 33853885 PMCID: PMC9488698 DOI: 10.1183/16000617.0284-2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/08/2020] [Indexed: 11/05/2022] Open
Abstract
Exercise intolerance is a cardinal symptom of pulmonary arterial hypertension (PAH) and strongly impacts patients' quality of life (QoL). Although central cardiopulmonary impairments limit peak oxygen consumption (V' O2peak ) in patients with PAH, several peripheral abnormalities have been described over the recent decade as key determinants in exercise intolerance, including impaired skeletal muscle (SKM) morphology, convective O2 transport, capillarity and metabolism indicating that peripheral abnormalities play a greater role in limiting exercise capacity than previously thought. More recently, cerebrovascular alterations potentially contributing to exercise intolerance in patients with PAH were also documented. Currently, only cardiopulmonary rehabilitation has been shown to efficiently improve the peripheral components of exercise intolerance in patients with PAH. However, more extensive studies are needed to identify targeted interventions that would ultimately improve patients' exercise tolerance and QoL. The present review offers a broad and comprehensive analysis of the present literature about the complex mechanisms and their interactions limiting exercise in patients and suggests several gaps in knowledge that need to be addressed in the future for a better understanding of exercise intolerance in patients with PAH.
Collapse
Affiliation(s)
- Simon Malenfant
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
- Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Marius Lebret
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
- Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Émilie Breton-Gagnon
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
- Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - François Potus
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
| | - Roxane Paulin
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
- Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
- Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Steeve Provencher
- Pulmonary Hypertension and Vascular Biology Research Group, Quebec Heart and Lung Institute Research Center, Quebec City, Canada
- Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
11
|
Brito J, Siques P, Pena E. Long-term chronic intermittent hypoxia: a particular form of chronic high-altitude pulmonary hypertension. Pulm Circ 2020; 10:5-12. [PMID: 33110494 PMCID: PMC7557688 DOI: 10.1177/2045894020934625] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
In some subjects, high-altitude hypobaric hypoxia leads to high-altitude pulmonary
hypertension. The threshold for the diagnosis of high-altitude pulmonary hypertension is a
mean pulmonary artery pressure of 30 mmHg, even though for general pulmonary hypertension
is ≥25 mmHg. High-altitude pulmonary hypertension has been associated with high hematocrit
findings (chronic mountain sickness), and although these are two separate entities, they
have a synergistic effect that should be considered. In recent years, a new condition
associated with high altitude was described in South America named long-term chronic
intermittent hypoxia and has appeared in individuals who commute to work at high altitude
but live and rest at sea level. In this review, we discuss the initial epidemiological
pattern from the early studies done in Chile, the clinical presentation and possible
molecular mechanism and a discussion of the potential management of this condition.
Collapse
Affiliation(s)
- Julio Brito
- Institute of Health Studies, Universidad Arturo Prat, Iquique, Chile.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany
| | - Patricia Siques
- Institute of Health Studies, Universidad Arturo Prat, Iquique, Chile.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany
| | - Eduardo Pena
- Institute of Health Studies, Universidad Arturo Prat, Iquique, Chile.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany
| |
Collapse
|
12
|
Oxidative Stress, Kinase Activity and Inflammatory Implications in Right Ventricular Hypertrophy and Heart Failure under Hypobaric Hypoxia. Int J Mol Sci 2020; 21:ijms21176421. [PMID: 32899304 PMCID: PMC7503689 DOI: 10.3390/ijms21176421] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
High altitude (hypobaric hypoxia) triggers several mechanisms to compensate for the decrease in oxygen bioavailability. One of them is pulmonary artery vasoconstriction and its subsequent pulmonary arterial remodeling. These changes can lead to pulmonary hypertension and the development of right ventricular hypertrophy (RVH), right heart failure (RHF) and, ultimately to death. The aim of this review is to describe the most recent molecular pathways involved in the above conditions under this type of hypobaric hypoxia, including oxidative stress, inflammation, protein kinases activation and fibrosis, and the current therapeutic approaches for these conditions. This review also includes the current knowledge of long-term chronic intermittent hypobaric hypoxia. Furthermore, this review highlights the signaling pathways related to oxidative stress (Nox-derived O2.- and H2O2), protein kinase (ERK5, p38α and PKCα) activation, inflammatory molecules (IL-1β, IL-6, TNF-α and NF-kB) and hypoxia condition (HIF-1α). On the other hand, recent therapeutic approaches have focused on abolishing hypoxia-induced RVH and RHF via attenuation of oxidative stress and inflammatory (IL-1β, MCP-1, SDF-1 and CXCR-4) pathways through phytotherapy and pharmacological trials. Nevertheless, further studies are necessary.
Collapse
|
13
|
Gao X, Zhang Z, Li X, Li C, Hao J, Luo Y, Lei M, Li J, Liu C, He K. Macitentan Attenuates Chronic Mountain Sickness in Rats by Regulating Arginine and Purine Metabolism. J Proteome Res 2020; 19:3302-3314. [PMID: 32640793 DOI: 10.1021/acs.jproteome.0c00219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaojian Gao
- Department of Cardiovascular, Chinese PLA General Hospital, Beijing 100853, China
| | - Zeyu Zhang
- Department of Cardiovascular, Chinese PLA General Hospital, Beijing 100853, China
| | - Xin Li
- Core Laboratory of Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Chen Li
- Core Laboratory of Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Jianxiu Hao
- Core Laboratory of Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Yunfu Luo
- WuXi AppTec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Maoyi Lei
- WuXi AppTec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Junmiao Li
- WuXi AppTec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Chunlei Liu
- Core Laboratory of Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Kunlun He
- Core Laboratory of Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
14
|
McCullough DJ, Kue N, Mancini T, Vang A, Clements RT, Choudhary G. Endurance exercise training in pulmonary hypertension increases skeletal muscle electron transport chain supercomplex assembly. Pulm Circ 2020; 10:2045894020925762. [PMID: 32523689 PMCID: PMC7235683 DOI: 10.1177/2045894020925762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/20/2020] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension is associated with pronounced exercise intolerance (decreased V ċ O2 max) that can significantly impact quality of life. The cause of exercise intolerance in pulmonary hypertension remains unclear. Mitochondrial supercomplexes are large respiratory assemblies of individual electron transport chain complexes which can promote more efficient respiration. In this study, we examined pulmonary hypertension and exercise-induced changes in skeletal muscle electron transport chain protein expression and supercomplex assembly. Pulmonary arterial hypertension was induced in rats with the Sugen/Hypoxia model (10% FiO2, three weeks). Pulmonary arterial hypertension and control rats were assigned to an exercise training protocol group or kept sedentary for one month. Cardiac function and V ċ O2 max were assessed at the beginning and end of exercise training. Red (Type 1—oxidative muscle) and white (Type 2—glycolytic muscle) gastrocnemius were assessed for changes in electron transport chain complex protein expression and supercomplex assembly via SDS- and Blue Native-PAGE. Results showed that pulmonary arterial hypertension caused a significant decrease in V ċ O2 max via treadmill testing that was improved with exercise (P < 0.01). Decreases in cardiac output and pulmonary acceleration time due to pulmonary arterial hypertension were not improved with exercise. Pulmonary arterial hypertension reduced expression in individual electron transport chain complex protein expression (NDUFB8 (CI), SDHB (CII), Cox IV (CIV), but not UQCRC2 (CIII), or ATP5a (CV)) in red gastrocnemius muscle. Both red gastrocnemius and white gastrocnemius electron transport chain expression was unaffected by exercise. However, non-denaturing Blue Native-PAGE analysis of mitochondrial supercomplexes demonstrated increases with exercise training in pulmonary arterial hypertension in the red gastrocnemius but not white gastrocnemius muscle. Pulmonary arterial hypertension-induced exercise intolerance is improved with exercise and is associated with muscle type specific alteration in mitochondrial supercomplex assembly and expression of mitochondrial electron transport chain proteins.
Collapse
Affiliation(s)
- Danielle J McCullough
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA.,Edward Via College of Osteopathic Medicine, Auburn Campus, Auburn, AL, USA
| | - Nouaying Kue
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA
| | - Thomas Mancini
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA
| | - Alexander Vang
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA
| | - Richard T Clements
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA.,Department of Surgery, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence RI, USA.,Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island College of Pharmacy, Kingston, RI, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA.,Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
15
|
Endothelin-1 Downregulates Sulfur Dioxide/Aspartate Aminotransferase Pathway via Reactive Oxygen Species to Promote the Proliferation and Migration of Vascular Smooth Muscle Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9367673. [PMID: 32089786 PMCID: PMC7008293 DOI: 10.1155/2020/9367673] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 01/06/2020] [Indexed: 01/08/2023]
Abstract
The regulatory mechanisms for proliferation and migration of vascular smooth muscle cells have not yet been clear. The present study was designed to investigate whether and how endothelin-1 (ET-1) impacted the generation of endogenous sulfur dioxide (SO2) in rat vascular smooth muscle cell (VSMC) proliferation and migration. Primary VSMCs and purified aspartate aminotransferase (AAT) protein were used in this study. We found that in the presence of ET-1, the expression of PCNA and Ki-67 was upregulated and the migration of VSMCs was promoted, while the AAT activity and SO2 levels in VSMCs were reduced without any changes in AAT1 and AAT2 expression. SO2 supplementation successfully prevented the ET-1-facilitated expression of PCNA and Ki-67 and the migration of VSMCs. Interestingly, ET-1 significantly increased reactive oxygen species (ROS) production in association with SO2/AAT pathway downregulation in VSMCs compared with controls, while the ROS scavenger N-acetyl-L-cysteine (NAC) and the antioxidant glutathione (GSH) significantly abolished the ET-1-stimulated downregulation of the SO2/AAT pathway. Moreover, the AAT activity was reduced in purified protein after the treatment for 2 h. However, NAC and GSH blocked the hydrogen peroxide-induced AAT activity reduction. In conclusion, our results suggest that ET-1 results in the downregulation of the endogenous SO2/AAT pathway via ROS generation to enhance the proliferation and migration of VSMCs.
Collapse
|
16
|
Yamamoto K, Nishimura R, Kato F, Naito A, Suda R, Sekine A, Jujo T, Shigeta A, Sakao S, Tanabe N, Tatsumi K. Protective role of endothelial progenitor cells stimulated by riociguat in chronic thromboembolic pulmonary hypertension. Int J Cardiol 2020; 299:263-270. [DOI: 10.1016/j.ijcard.2019.07.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/06/2019] [Accepted: 07/04/2019] [Indexed: 12/18/2022]
|
17
|
Yuan T, Zhang H, Chen D, Chen Y, Lyu Y, Fang L, Du G. Puerarin protects pulmonary arteries from hypoxic injury through the BMPRII and PPARγ signaling pathways in endothelial cells. Pharmacol Rep 2019; 71:855-861. [PMID: 31408784 DOI: 10.1016/j.pharep.2019.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/17/2019] [Accepted: 05/06/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recent evidence indicates that Puerarin has a protective effect on pulmonary arteries. In the present study, we aimed to investigate whether Puerarin could protect pulmonary arterial endothelial cells from hypoxic injury and determine its potential targets. METHODS In our study, human pulmonary arterial endothelial cells (HPAECs) were injured by hypoxic (1% O2) incubation. Cell viability was detected by a cell counting kit (CCK8). The production of nitric oxide (NO) was detected by Griess reagent and endothelin-1 (ET-1) was detected by the ELISA method. Oxidative stress was measured by a fluorescence microscope via the fluorescent probe DCFH-DA. Western blotting was employed for studying the mechanism. RESULTS The results show that Puerarin protects HPAECs from hypoxia-induced apoptosis and slightly improves cell viability. Puerarin increases NO and decreases ET-1 to prevent the imbalance between vasoactive substances induced by hypoxia in HPAECs. Puerarin also inhibits the oxidative stress induced by hypoxia. The results from the Western blot show that Puerarin activates the BMPRII/Smad and PPARγ/PI3K/Akt signaling pathways. CONCLUSION In conclusion, Puerarin protects HPAECs from hypoxic injury through the inhibition of oxidative stress and the activation of the BMPRII and PPARγ signaling pathways. This work provides insight into the development of Puerarin as a treatment for hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Tianyi Yuan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Huifang Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Di Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Yucai Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Yang Lyu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Polymorphic Drugs, Beijing, China
| | - Lianhua Fang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China.
| |
Collapse
|
18
|
Bonnet S, Boucherat O, Paulin R, Wu D, Hindmarch CCT, Archer SL, Song R, Moore JB, Provencher S, Zhang L, Uchida S. Clinical value of non-coding RNAs in cardiovascular, pulmonary, and muscle diseases. Am J Physiol Cell Physiol 2019; 318:C1-C28. [PMID: 31483703 DOI: 10.1152/ajpcell.00078.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although a majority of the mammalian genome is transcribed to RNA, mounting evidence indicates that only a minor proportion of these transcriptional products are actually translated into proteins. Since the discovery of the first non-coding RNA (ncRNA) in the 1980s, the field has gone on to recognize ncRNAs as important molecular regulators of RNA activity and protein function, knowledge of which has stimulated the expansion of a scientific field that quests to understand the role of ncRNAs in cellular physiology, tissue homeostasis, and human disease. Although our knowledge of these molecules has significantly improved over the years, we have limited understanding of their precise functions, protein interacting partners, and tissue-specific activities. Adding to this complexity, it remains unknown exactly how many ncRNAs there are in existence. The increased use of high-throughput transcriptomics techniques has rapidly expanded the list of ncRNAs, which now includes classical ncRNAs (e.g., ribosomal RNAs and transfer RNAs), microRNAs, and long ncRNAs. In addition, splicing by-products of protein-coding genes and ncRNAs, so-called circular RNAs, are now being investigated. Because there is substantial heterogeneity in the functions of ncRNAs, we have summarized the present state of knowledge regarding the functions of ncRNAs in heart, lungs, and skeletal muscle. This review highlights the pathophysiologic relevance of these ncRNAs in the context of human cardiovascular, pulmonary, and muscle diseases.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Roxane Paulin
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Joseph B Moore
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Steeve Provencher
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Shizuka Uchida
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
19
|
Fu J, Bai P, Chen Y, Yu T, Li F. Inhibition of miR-495 Improves Both Vascular Remodeling and Angiogenesis in Pulmonary Hypertension. J Vasc Res 2019; 56:97-106. [PMID: 31030195 DOI: 10.1159/000500024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 03/29/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND AIMS Pulmonary hypertension (PH) is a chronic progressing vascular disease characterized by pulmonary arteriole remodeling and loss of pulmonary microvasculature. The aim of this study was to investigate a potential role for the miR-495 in PH pathogenesis and to explore its therapeutic potential in PH. METHODS Male C57BL/6J mice were injected with SU5416 weekly during 3 weeks of exposure to 10% oxygen to cause PH. We first tested the effects of adeno-associated virus 9 (AAV9) delivery which was specifically designed to block miR-495 in the lungs of the PH model. Then, the biological function of miR-495 was analyzed in cultured pulmonary arterial endothelial cells (PAECs) under hypoxic condition. RESULTS The inhibition of miR-495 improves hemodynamics and vascular remodeling in PH. At the same time, these effects were associated with increases in angiogenic transcription factor VEZF1 and marked upregulation of other angiogenic genes such as Angpt-1 and IGF1. In vitro, cultured mouse PAECs were transfected with miR-495 inhibitor or miR-495 mimics. Both the flow cytometry results and CCK8 assay showed that miR-495 inhibitor increased the percentage of cells in the G2/M+S phase, and the wound healing assays indicated that the migration capacity of PAECs transfected with miR-495 inhibitor was increased compared to the inhibitor-NC cells. CONCLUSIONS Our results indicate that AAV9-TuD-miR-495 delivery improves hemodynamic and pulmonary vascular structural changes in PH mice.
Collapse
Affiliation(s)
- Jie Fu
- Department of Cardiology, Shanghai Children's Medical Center Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peiyuan Bai
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiwei Chen
- Department of Cardiology, Shanghai Children's Medical Center Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tingting Yu
- Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fen Li
- Department of Cardiology, Shanghai Children's Medical Center Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China, .,Shanghai Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China,
| |
Collapse
|
20
|
Giordano N, Corallo C, Chirico C, Brazzi A, Marinetti A, Fioravanti A, Valenti R, Nuti R, Pecetti G. Pulmonary arterial hypertension in systemic sclerosis: Diagnosis and treatment according to the European Society of Cardiology and European Respiratory Society 2015 guidelines. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2019; 4:35-42. [PMID: 35382146 PMCID: PMC8922580 DOI: 10.1177/2397198318808998] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/30/2018] [Indexed: 09/29/2023]
Abstract
Scleroderma (systemic sclerosis) is an autoimmune connective tissue disease which presents endothelial dysfunction and fibroblast dysregulation, resulting in vascular and fibrotic disorders. Pulmonary hypertension is frequent in patients with systemic sclerosis: the natural evolution of the disease can induce the development of different forms of pulmonary hypertension, representing one of the main causes of death. Among the different forms of pulmonary hypertension in systemic sclerosis, pulmonary arterial hypertension is the most frequent one (rate of occurrence is estimated between 7% and 12%). This pulmonary vascular complication should be treated with a combination of drugs that is able to counteract endothelial dysfunction, antagonizing the endothelin-1 system and replacing prostaglandin I2 and nitric oxide activity. A correct diagnosis is mandatory, because it is possible only for pulmonary arterial hypertension to use specific drugs that are able to control the symptomatic condition and the evolution of the disease. According to the most recent guidelines, for the patients with systemic sclerosis, also without pulmonary hypertension symptoms, echocardiography screening for the detection of pulmonary hypertension is recommended. Pulmonary arterial hypertension screening programs in systemic sclerosis patients is able to identify milder forms of the disease, allowing earlier management and better long-term outcome.
Collapse
Affiliation(s)
- Nicola Giordano
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Claudio Corallo
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Chiara Chirico
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Angelica Brazzi
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Adriana Marinetti
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | - Roberto Valenti
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Ranuccio Nuti
- Scleroderma Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Gianluca Pecetti
- Medical and Scientific Direction, Actelion Pharmaceuticals Italia s.r.l., Imola, Italy
| |
Collapse
|
21
|
Akagi S, Matsubara H, Nakamura K, Ito H. Modern treatment to reduce pulmonary arterial pressure in pulmonary arterial hypertension. J Cardiol 2018; 72:466-472. [DOI: 10.1016/j.jjcc.2018.04.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 11/30/2022]
|
22
|
Graham BB, Kumar R, Mickael C, Kassa B, Koyanagi D, Sanders L, Zhang L, Perez M, Hernandez-Saavedra D, Valencia C, Dixon K, Harral J, Loomis Z, Irwin D, Nemkov T, D’Alessandro A, Stenmark KR, Tuder RM. Vascular Adaptation of the Right Ventricle in Experimental Pulmonary Hypertension. Am J Respir Cell Mol Biol 2018; 59:479-489. [PMID: 29851508 PMCID: PMC6178158 DOI: 10.1165/rcmb.2018-0095oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/31/2018] [Indexed: 01/25/2023] Open
Abstract
Optimal right ventricular (RV) function in pulmonary hypertension (PH) requires structural and functional coupling between the RV cardiomyocyte and its adjacent capillary network. Prior investigations have indicated that RV vascular rarefaction occurs in PH, which could contribute to RV failure by reduced delivery of oxygen or other metabolic substrates. However, it has not been determined if rarefaction results from relative underproliferation in the setting of tissue hypertrophy or from actual loss of vessels. It is also unknown if rarefaction results in inadequate substrate delivery to the RV tissue. In the present study, PH was induced in rats by SU5416-hypoxia-normoxia exposure. The vasculature in the RV free wall was assessed using stereology. Steady-state metabolomics of the RV tissue was performed by mass spectrometry. Complementary studies were performed in hypoxia-exposed mice and rats. Rats with severe PH had evidence of RV failure by decreased cardiac output and systemic hypotension. By stereology, there was significant RV hypertrophy and increased total vascular length in the RV free wall in close proportion, with evidence of vessel proliferation but no evidence of endothelial cell apoptosis. There was a modest increase in the radius of tissue served per vessel, with decreased arterial delivery of metabolic substrates. Metabolomics revealed major metabolic alterations and metabolic reprogramming; however, metabolic substrate delivery was functionally preserved, without evidence of either tissue hypoxia or depletion of key metabolic substrates. Hypoxia-treated rats and mice had similar but milder alterations. There is significant homeostatic vascular adaptation in the right ventricle of rodents with PH.
Collapse
Affiliation(s)
- Brian B. Graham
- Program in Translation Lung Research, Department of Medicine
| | - Rahul Kumar
- Program in Translation Lung Research, Department of Medicine
| | - Claudia Mickael
- Program in Translation Lung Research, Department of Medicine
| | - Biruk Kassa
- Program in Translation Lung Research, Department of Medicine
| | - Dan Koyanagi
- Program in Translation Lung Research, Department of Medicine
| | - Linda Sanders
- Program in Translation Lung Research, Department of Medicine
| | - Li Zhang
- Program in Translation Lung Research, Department of Medicine
| | - Mario Perez
- Program in Translation Lung Research, Department of Medicine
| | | | | | | | | | | | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, Colorado
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, Colorado
| | | | - Rubin M. Tuder
- Program in Translation Lung Research, Department of Medicine
| |
Collapse
|
23
|
Simakova MA, Goncharova NS, Karelkina EV, Moiseeva OM. EXPERIENCE OF USING THE NONSELECTIVE ENDOTHELIN RECEPTOR ANTAGONIST MACITENTAN IN PATIENTS WITH PULMONARY ARTERIAL HYPERTENSION. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2018. [DOI: 10.15829/1728-8800-2018-2-35-42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. To present the results of treatment with macitentan of patients included in the clinical trial SERAPHIN in Almazov National Medical Research Centre and define target therapy of pulmonary arterial hypertension (PAH) in real clinical practice.Material and methods. The article presents the results of the 151 patients with PAH followedup in Almazov National Medical Research Centre from 2009 to 2016.Results. Fiveyear survival of PAH patients included in the register of Almazov National Medical Research Centre reached 77% for idiopathic PAH, 52% for PAH associated with systemic scleroderma, 81% for PAH associated with nonrepaired congenital heart disease, 82% for patients with repaired congenital shunts and 100% for PAH associated with human immunodeficiency virus infection. The group that received PAH specific monotherapy consisted of 47% (n=71) of patients among which phosphodiesterase type 5 inhibitor (PDE5i) sildenafil was undoubtedly the most prescribed drug — 74% (n=53). The group that received combined PAH specific therapy consisted of 66 (44%) patients: 48 patients received various twocomponent therapy, 18 patients — threecomponent therapy with endothelin receptor antagonist in combination with PDE5i and prostanoids. 11 patients with PAH were included in SERAPHIN study of which 9 patients had been already receiving generic PDE5i therapy. In macitentan group, a statistically significant increase in the 6minute walk distance (+50 meters) and a decrease in hemodynamic parameters such as mean right atrial pressure (2,3 mm Hg) and pulmonary vascular resistance (445 dynsec/cm5) were observed after 6 months. No patient had a clinically significant increase in liver transaminases or a decrease in hemoglobin levels.Conclusion. Improvement of prognosis in PAH patients according to the register of the Centre is connected both with early detection of the disease, thanks to the development of specialized healthcare, and more frequent use of combination therapy. Macitentan proved its longterm efficacy and safety as monotherapy and in combination with PDE5i.
Collapse
Affiliation(s)
- M. A. Simakova
- FSBI “Almazov National Medical Research Centre” of the Ministry of Health of the Russian Federation
| | - N. S. Goncharova
- FSBI “Almazov National Medical Research Centre” of the Ministry of Health of the Russian Federation
| | - E. V. Karelkina
- FSBI “Almazov National Medical Research Centre” of the Ministry of Health of the Russian Federation
| | - O. M. Moiseeva
- FSBI “Almazov National Medical Research Centre” of the Ministry of Health of the Russian Federation
| |
Collapse
|