1
|
Kianzad A, Baccelli A, Braams NJ, Andersen S, van Wezenbeek J, Wessels JN, Celant LR, Vos AE, Davies R, Lo Giudice F, Haji G, Rinaldo RF, Vigo B, Gopalan D, Symersky P, Winkelman JA, Boonstra A, Nossent EJ, Tim Marcus J, Vonk Noordegraaf A, Meijboom LJ, de Man FS, Andersen A, Howard LS, Bogaard HJ. Long-term effects of pulmonary endarterectomy on pulmonary hemodynamics, cardiac function, and exercise capacity in chronic thromboembolic pulmonary hypertension. J Heart Lung Transplant 2024; 43:580-593. [PMID: 38000764 DOI: 10.1016/j.healun.2023.11.011] [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: 06/06/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Long-term changes in exercise capacity and cardiopulmonary hemodynamics after pulmonary endarterectomy (PEA) for chronic thromboembolic pulmonary hypertension (CTEPH) have been poorly described. METHODS We analyzed the data from 2 prospective surgical CTEPH cohorts in Hammersmith Hospital, London, and Amsterdam UMC. A structured multimodal follow-up was adopted, consisting of right heart catheterization, cardiac magnetic resonance imaging, and cardiopulmonary exercise testing before and after PEA. Preoperative predictors of residual pulmonary hypertension (PH; mean pulmonary artery pressure >20 mm Hg and pulmonary vascular resistance ≥2 WU) and long-term exercise intolerance (VO2max <80%) at 18 months were analyzed. RESULTS A total of 118 patients (61 from London and 57 from Amsterdam) were included in the analysis. Both cohorts displayed a significant improvement of pulmonary hemodynamics, right ventricular (RV) function, and exercise capacity 6 months after PEA. Between 6 and 18 months after PEA, there were no further improvements in hemodynamics and RV function, but the proportion of patients with impaired exercise capacity was high and slightly increased over time (52%-59% from 6 to 18 months). Long-term exercise intolerance was common and associated with preoperative diffusion capacity for carbon monoxide (DLCO), preoperative mixed venous oxygen saturation, and postoperative PH and right ventricular ejection fraction (RVEF). Clinically significant RV deterioration (RVEF decline >3%; 5 [9%] of 57 patients) and recurrent PH (5 [14%] of 36 patients) rarely occurred beyond 6 months after PEA. Age and preoperative DLCO were predictors of residual PH post-PEA. CONCLUSIONS Restoration in exercise tolerance, cardiopulmonary hemodynamics, and RV function occurs within 6 months. No substantial changes occurred between 6 and 18 months after PEA in the Amsterdam cohort. Nevertheless, long-term exercise intolerance is common and associated with postoperative RV function.
Collapse
Affiliation(s)
- Azar Kianzad
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Andrea Baccelli
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Unit, ASST Santi Paolo e Carlo, San Paolo Hospital, Department of Health Sciences, University of Milan, Milan, Italy
| | - Natalia J Braams
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Stine Andersen
- Aarhus University Hospital, Department of Cardiology, Aarhus, Denmark
| | - Jessie van Wezenbeek
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Jeroen N Wessels
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Lucas R Celant
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Anna E Vos
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands
| | - Rachel Davies
- National Pulmonary Hypertension Service, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Francesco Lo Giudice
- National Pulmonary Hypertension Service, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Gulammehdi Haji
- National Pulmonary Hypertension Service, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Rocco F Rinaldo
- Respiratory Unit, ASST Santi Paolo e Carlo, San Paolo Hospital, Department of Health Sciences, University of Milan, Milan, Italy
| | - Beatrice Vigo
- Respiratory Unit, ASST Santi Paolo e Carlo, San Carlo Hospital, Department of Health Sciences, University of Milan, Milan, Italy
| | - Deepa Gopalan
- Department of Radiology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Petr Symersky
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Cardiothoracic Surgery, Amsterdam, the Netherlands
| | - Jacobus A Winkelman
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Cardiothoracic Surgery, Amsterdam, the Netherlands
| | - Anco Boonstra
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Esther J Nossent
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - J Tim Marcus
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands; Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands
| | - Anton Vonk Noordegraaf
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Lilian J Meijboom
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands; Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands
| | - Frances S de Man
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - Asger Andersen
- Aarhus University Hospital, Department of Cardiology, Aarhus, Denmark
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; National Pulmonary Hypertension Service, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Harm Jan Bogaard
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Scarlata S, Di Matteo E, Finamore P, Perri G, Mancini D, Sogaro L, Grandi T, Brando E, Travaglino F, Sambuco F, Antonelli Incalzi R. Diaphragmatic ultrasound evaluation in acute heart failure: clinical and functional associations. Intern Emerg Med 2024; 19:705-711. [PMID: 38363523 DOI: 10.1007/s11739-024-03531-9] [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: 07/31/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024]
Abstract
Heart failure patients often experience respiratory symptoms due to diaphragmatic involvement, but the diaphragmatic motion in heart failure remains understudied. This research aimed to investigate the correlation between ultrasonographically assessed diaphragmatic motion and thickness with cardiac performance indexes in an emergency setting. Seventy-two acutely decompensated heart failure patients and 100 non-heart failure individuals were enrolled. Diaphragmatic motion and thickness were assessed via ultrasound. Cardiac and respiratory parameters were recorded, and regression analysis was performed. Heart failure patients exhibited reduced diaphragmatic motion at total lung capacity compared to controls, and an inverse association was found between motion and heart failure severity (NYHA stage). Diaphragmatic thickness was also higher in heart failure patients at tidal volume and total lung capacity. Notably, diaphragmatic motion inversely correlated with systolic pulmonary artery pressure. The study highlights diaphragmatic dysfunction in acutely decompensated heart failure, with reduced motion and increased thickness. These changes were associated with cardio-respiratory parameters, specifically systolic pulmonary artery pressure. Monitoring diaphragmatic motion via ultrasound may aid in evaluating heart failure severity and prognosis in emergency settings. Additionally, interventions targeting diaphragmatic function could improve heart failure management. Further research is warranted to enhance heart failure management and patient outcomes.
Collapse
Affiliation(s)
- Simone Scarlata
- Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Via Alvaro del Portillo, 200-00128, Rome, Italy.
- Research Unit of Internal Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.
| | - Evelyn Di Matteo
- Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Via Alvaro del Portillo, 200-00128, Rome, Italy
| | - Panaiotis Finamore
- Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Via Alvaro del Portillo, 200-00128, Rome, Italy
| | - Giuseppe Perri
- Geriatrics Unit, Ospedale Santa Maria della Scaletta, Imola, BO, Italy
| | | | - Luigi Sogaro
- Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Via Alvaro del Portillo, 200-00128, Rome, Italy
| | - Tommaso Grandi
- Unit of Emergency and Critical Care Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
| | - Elisa Brando
- Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Via Alvaro del Portillo, 200-00128, Rome, Italy
| | - Francesco Travaglino
- Unit of Emergency and Critical Care Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
| | - Federica Sambuco
- Unit of Emergency and Critical Care Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
| | - Raffaele Antonelli Incalzi
- Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Via Alvaro del Portillo, 200-00128, Rome, Italy
- Research Unit of Internal Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
3
|
Singh N, Al-Naamani N, Brown MB, Long GM, Thenappan T, Umar S, Ventetuolo CE, Lahm T. Extrapulmonary manifestations of pulmonary arterial hypertension. Expert Rev Respir Med 2024; 18:189-205. [PMID: 38801029 DOI: 10.1080/17476348.2024.2361037] [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: 12/05/2023] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Extrapulmonary manifestations of pulmonary arterial hypertension (PAH) may play a critical pathobiological role and a deeper understanding will advance insight into mechanisms and novel therapeutic targets. This manuscript reviews our understanding of extrapulmonary manifestations of PAH. AREAS COVERED A group of experts was assembled and a complimentary PubMed search performed (October 2023 - March 2024). Inflammation is observed throughout the central nervous system and attempts at manipulation are an encouraging step toward novel therapeutics. Retinal vascular imaging holds promise as a noninvasive method of detecting early disease and monitoring treatment responses. PAH patients have gut flora alterations and dysbiosis likely plays a role in systemic inflammation. Despite inconsistent observations, the roles of obesity, insulin resistance and dysregulated metabolism may be illuminated by deep phenotyping of body composition. Skeletal muscle dysfunction is perpetuated by metabolic dysfunction, inflammation, and hypoperfusion, but exercise training shows benefit. Renal, hepatic, and bone marrow abnormalities are observed in PAH and may represent both end-organ damage and disease modifiers. EXPERT OPINION Insights into systemic manifestations of PAH will illuminate disease mechanisms and novel therapeutic targets. Additional study is needed to understand whether extrapulmonary manifestations are a cause or effect of PAH and how manipulation may affect outcomes.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | - Nadine Al-Naamani
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Gary Marshall Long
- Department of Kinesiology, Health and Sport Sciences, University of Indianapolis, Indianapolis, IN, USA
| | - Thenappan Thenappan
- Section of Advanced Heart Failure and Pulmonary Hypertension, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Corey E Ventetuolo
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, USA
- Department of Health Services, Policy and Practice, Brown University, Providence, RI, USA
| | - Tim Lahm
- Department of Medicine, National Jewish Health, Denver, CO, USA
- Department of Medicine, University of Colorado, Aurora, CO, USA
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| |
Collapse
|
4
|
Nakayama M, Konishi M, Sugano T, Okamura M, Gohbara M, Iwata K, Nakayama N, Akiyama E, Komura N, Nitta M, Kawaura N, Ishigami T, Hibi K, Ishikawa T, Nakamura T, Tamura K, Kimura K. Association between sarcopenia and exercise capacity in patients with pulmonary hypertension without left heart disease. Int J Cardiol 2023; 387:131115. [PMID: 37302419 DOI: 10.1016/j.ijcard.2023.06.006] [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: 10/25/2022] [Revised: 03/25/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) has recently been described as a complex clinical syndrome affecting multiple organ systems, including the heart, lungs, and skeletal muscle, each of which plays an important role in exercise capacity. However, the relationship between exercise capacity and skeletal muscle abnormalities in patients with PH has not been fully elucidated. METHODS We retrospectively analysed the exercise capacity and measures of skeletal muscle of 107 patients with PH without left heart disease (mean age 63 ± 15 years, 32.7% males, n = 30/6/66/5 in the clinical classification Group 1/3/4/5). RESULTS Sarcopenia, low appendicular skeletal muscle mass index, low grip strength, and slow gait speed, determined by international criteria, were found in 15 (14.0%), 16 (15.0%), 62 (57.9%), and 41 (38.3%) patients, respectively. The mean 6-min walk distance of all patients was 436 ± 134 m and was independently associated with sarcopenia (standardised β = -0.292, p < 0.001). All patients with sarcopenia showed reduced exercise capacity defined as 6-min walk distance < 440 m. Multivariable logistic regression analysis showed that each of the components of sarcopenia was associated with reduced exercise capacity (adjusted odds ratio and 95% confidence interval of appendicular skeletal muscle mass index: 0.39 [0.24-0.63] per 1 kg/m2, p = 0.006, grip strength: 0.83 [0.74-0.94] per 1 kg, p = 0.003, and gait speed: 0.31 [0.18-0.51] per 0.1 m/s, p < 0.001). CONCLUSIONS Sarcopenia and its components are associated with reduced exercise capacity in patients with PH. A multifaceted evaluation may be important in the management of reduced exercise capacity in patients with PH.
Collapse
Affiliation(s)
- Mina Nakayama
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaaki Konishi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Teruyasu Sugano
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masatsugu Okamura
- Department of Rehabilitation Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Berlin Institute of Health Center forRegenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Masaomi Gohbara
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kiwamu Iwata
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naoki Nakayama
- Department of Cardiology, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Eiichi Akiyama
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Naohiro Komura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Manabu Nitta
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Center for Novel and Exploratory Clinical Trials (Y-NEXT), Yokohama City University Hospital, Yokohama, Japan
| | - Noriyuki Kawaura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Toshiyuki Ishikawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Nakamura
- Department of Rehabilitation Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuo Kimura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
5
|
Evaluation of primary and accessory respiratory muscles and their influence on exercise capacity and dyspnea in pulmonary arterial hypertension. Heart Lung 2023; 57:173-179. [PMID: 36219922 DOI: 10.1016/j.hrtlng.2022.09.015] [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: 07/08/2022] [Revised: 08/11/2022] [Accepted: 09/25/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Skeletal and respiratory muscle disfunction has been described in pulmonary arterial hypertension (PAH), however, involvement of accessory respiratory muscles and their association with symptomatology in PAH is unclear. OBJECTIVES To assess the primary and accessory respiratory muscles and their influence on exercise tolerance and dyspnea. METHODS 27 patients and 27 healthy controls were included. Serratus anterior (SA), pectoralis muscles (PM) and sternocleidomastoid (SCM) muscle strength were evaluated as accessory respiratory muscles, maximal inspiratory (MIP) and expiratory pressures (MEP) as primary respiratory muscles, and quadriceps as peripheral muscle. Exercise capacity was evaluated with 6-min walk test (6MWT), dyspnea with modified Medical Council Research (MMRC) and London Chest Activity of Daily Living (LCADL) scales. RESULTS All evaluated muscles, except SCM, and 6MWT were decreased in patient group (p < 0.01). SA was the most affected muscle among primary and accessory respiratory muscles (Cohen's-d = 1.35). All evaluated muscles significantly correlated to 6MWT (r = 0.428-0.525). A multivariate model including SA, SCM and MIP was the best model for predicting 6MWT (R = 0.606; R2 = 0.368; p = 0.013) and SA strength had the most impact on the 6MWT (B = 1.242; β = 0.340). None of the models including respiratory muscles were able to predict dyspnea, however PM and SA strength correlated to LCADL total (r = -0.493) and MMRC (r = -0.523), respectively. CONCLUSION SCM may be excessively used in PAH since it retains its strength. Considering the relationship of accessory respiratory muscles with exercise tolerance and dyspnea, monitoring the strength of these muscles in the clinical practice may help providing better management for PAH.
Collapse
|
6
|
Miao R, Dong X, Gong J, Li Y, Guo X, Wang J, Huang Q, Wang Y, Li J, Yang S, Kuang T, Liu M, Wan J, Zhai Z, Zhong J, Yang Y. Single-cell RNA-sequencing and microarray analyses to explore the pathological mechanisms of chronic thromboembolic pulmonary hypertension. Front Cardiovasc Med 2022; 9:900353. [PMID: 36440052 PMCID: PMC9684175 DOI: 10.3389/fcvm.2022.900353] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 10/21/2022] [Indexed: 08/25/2023] Open
Abstract
OBJECTIVE The present study aimed to explore the pathological mechanisms of chronic thromboembolic pulmonary hypertension (CTEPH) using a gene chip array and single-cell RNA-sequencing (scRNA-seq). MATERIALS AND METHODS The mRNA expression profile GSE130391 was downloaded from the Gene Expression Omnibus database. The peripheral blood samples of five CTEPH patients and five healthy controls were used to prepare the Affymetrix microRNA (miRNA) chip and the Agilent circular RNA (circRNA) chip. The pulmonary endarterectomized tissues from five CTEPH patients were analyzed by scRNA-seq. Cells were clustered and annotated, followed by the identification of highly expressed genes. The gene chip data were used to identify disease-related mRNAs and differentially expressed miRNAs and circRNAs. The protein-protein interaction (PPI) network and the circRNA-miRNA-mRNA network were constructed for each cell type. RESULTS A total of 11 cell types were identified. Intersection analysis of highly expressed genes in each cell type and differentially expressed mRNAs were performed to obtain disease-related genes in each cell type. TP53, ICAM1, APP, ITGB2, MYC, and ZYX showed the highest degree of connectivity in the PPI network of different types of cells. In addition, the circRNA-miRNA-mRNA network for each cell type was constructed. CONCLUSION For the first time, the key mRNAs, miRNAs, and circRNAs, as well as their possible regulatory relationships, during the progression of CTEPH were analyzed using both gene chip and scRNA-seq data. These findings may contribute to a better understanding of the pathological mechanisms of CTEPH.
Collapse
Affiliation(s)
- Ran Miao
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xingbei Dong
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juanni Gong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yidan Li
- Department of Echocardiography, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Guo
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jianfeng Wang
- Department of Interventional Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qiang Huang
- Department of Interventional Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jifeng Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Suqiao Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Tuguang Kuang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Jun Wan
- Department of Respiration, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhenguo Zhai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yuanhua Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Long GM, Troutman AD, Gray DA, Fisher AJ, Lahm T, Coggan AR, Brown MB. Skeletal muscle blood flow during exercise is reduced in a rat model of pulmonary hypertension. Am J Physiol Regul Integr Comp Physiol 2022; 323:R561-R570. [PMID: 36036455 PMCID: PMC9602702 DOI: 10.1152/ajpregu.00327.2021] [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: 12/21/2021] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by exercise intolerance. Muscle blood flow may be reduced during exercise in PAH; however, this has not been directly measured. Therefore, we investigated blood flow during exercise in a rat model of monocrotaline (MCT)-induced pulmonary hypertension (PH). Male Sprague-Dawley rats (∼200 g) were injected with 60 mg/kg MCT (MCT, n = 23) and vehicle control (saline; CON, n = 16). Maximal rate of oxygen consumption (V̇o2max) and voluntary running were measured before PH induction. Right ventricle (RV) morphology and function were assessed via echocardiography and invasive hemodynamic measures. Treadmill running at 50% V̇o2max was performed by a subgroup of rats (MCT, n = 8; CON, n = 7). Injection of fluorescent microspheres determined muscle blood flow via photo spectroscopy. MCT demonstrated a severe phenotype via RV hypertrophy (Fulton index, 0.61 vs. 0.31; P < 0.001), high RV systolic pressure (51.5 vs. 22.4 mmHg; P < 0.001), and lower V̇o2max (53.2 vs. 71.8 mL·min-1·kg-1; P < 0.0001) compared with CON. Two-way ANOVA revealed exercising skeletal muscle blood flow relative to power output was reduced in MCT compared with CON (P < 0.001), and plasma lactate was increased in MCT (10.8 vs. 4.5 mmol/L; P = 0.002). Significant relationships between skeletal blood flow and blood lactate during exercise were observed for individual muscles (r = -0.58 to -0.74; P < 0.05). No differences in capillarization were identified. Skeletal muscle blood flow is significantly reduced in experimental PH. Reduced blood flow during exercise may be, at least in part, consequent to reduced exercise intensity in PH. This adds further evidence of peripheral muscle dysfunction and exercise intolerance in PAH.
Collapse
Affiliation(s)
- Gary Marshall Long
- Department of Kinesiology, University of Indianapolis, Indianapolis, Indiana
| | - Ashley D Troutman
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Derrick A Gray
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Amanda J Fisher
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Tim Lahm
- Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Indiana University, Indianapolis, Indiana
- Richard L. Roudebush Veteran Affairs Medical Center, Indianapolis, Indiana
| | - Andrew R Coggan
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
8
|
Prevalence of musculoskeletal pain and its impact on quality of life and functional exercise capacity in patients with pulmonary arterial hypertension. Respir Med 2022; 193:106759. [DOI: 10.1016/j.rmed.2022.106759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 01/01/2023]
|
9
|
Krishnamachary B, Mahajan A, Kumar A, Agarwal S, Mohan A, Chen L, Hsue PY, Chalise P, Morris A, Dhillon NK. Extracellular Vesicle TGF-β1 Is Linked to Cardiopulmonary Dysfunction in Human Immunodeficiency Virus. Am J Respir Cell Mol Biol 2021; 65:413-429. [PMID: 34014809 PMCID: PMC8525206 DOI: 10.1165/rcmb.2021-0010oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/18/2021] [Indexed: 01/21/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as important mediators in cell-cell communication; however, their relevance in pulmonary hypertension (PH) secondary to human immunodeficiency virus (HIV) infection is yet to be explored. Considering that circulating monocytes are the source of the increased number of perivascular macrophages surrounding the remodeled vessels in PH, this study aimed to identify the role of circulating small EVs and EVs released by HIV-infected human monocyte-derived macrophages in the development of PH. We report significantly higher numbers of plasma-derived EVs carrying higher levels of TGF-β1 (transforming growth factor-β1) in HIV-positive individuals with PH compared with individuals without PH. Importantly, levels of these TGF-β1-loaded, plasma-derived EVs correlated with pulmonary arterial systolic pressures and CD4 counts but did not correlate with the Dl CO or viral load. Correspondingly, enhanced TGF-β1-dependent pulmonary endothelial injury and smooth muscle hyperplasia were observed. HIV-1 infection of monocyte-derived macrophages in the presence of cocaine resulted in an increased number of TGF-β1-high EVs, and intravenous injection of these EVs in rats led to increased right ventricle systolic pressure accompanied by myocardial injury and increased levels of serum ET-1 (endothelin-1), TNF-α, and cardiac troponin-I. Conversely, pretreatment of rats with TGF-β receptor 1 inhibitor prevented these EV-mediated changes. Findings define the ability of macrophage-derived small EVs to cause pulmonary vascular modeling and PH via modulation of TGF-β signaling and suggest clinical implications of circulating TGF-β-high EVs as a potential biomarker of HIV-associated PH.
Collapse
Affiliation(s)
- Balaji Krishnamachary
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Aatish Mahajan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Ashok Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Aradhana Mohan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Ling Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Priscilla Y. Hsue
- Department of Medicine, University of California San Francisco, San Francisco, California; and
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Alison Morris
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Navneet K. Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| |
Collapse
|
10
|
Cannon DT, Nogueira L, Gutierrez-Gonzalez AK, Gilmore NK, Bigby TD, Breen EC. Role of IL-33 receptor (ST2) deletion in diaphragm contractile and mitochondrial function in the Sugen5416/hypoxia model of pulmonary hypertension. Respir Physiol Neurobiol 2021; 295:103783. [PMID: 34508866 DOI: 10.1016/j.resp.2021.103783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 12/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of the pulmonary vasculature that leads to right ventricular failure. Skeletal muscle maladaptations limit physical activity and may contribute to disease progression. The role of alarmin/inflammatory signaling in PAH respiratory muscle dysfunction is unknown. We hypothesized that diaphragm mitochondrial and contractile functions are impaired in SU5416/hypoxia-induced pulmonary hypertension due to increased systemic IL-33 signaling. We induced pulmonary hypertension in adult C57Bl/6 J (WT) and ST2 (IL1RL1) gene ablated mice by SU5416/hypoxia (SuHx). We measured diaphragm fiber mitochondrial respiration, inflammatory markers, and contractile function ex vivo. SuHx reduced coupled and uncoupled permeabilized myofiber respiration by ∼40 %. During coupled respiration with complex I substrates, ST2-/- attenuated SuHx inhibition of mitochondrial respiration (genotype × treatment interaction F[1,67] = 3.3, p = 0.07, η2 = 0.04). Flux control ratio and coupling efficiency were not affected by SuHx or genotype. A higher substrate control ratio for succinate was observed in SuHx fibers and attenuated in ST2-/- fibers (F[1,67] = 5.3, p < 0.05, η2 = 0.07). Diaphragm TNFα, but not IL-33 or NFkB, was increased in SuHx vs. DMSO in both genotypes (F[1,43] = 4.7, p < 0.05, η2 = 0.1). Diaphragm force-frequency relationships were right-shifted in SuHx vs. WT (F[3,440] = 8.4, p < 0.05, η2 = 0.0025). There was no effect of ST2-/- on the force-frequency relationship. Force decay during a fatigue protocol at 100 Hz, but not at 40 Hz, was attenuated by SuHx vs. DMSO in both genotypes (F[1,41] = 5.6, p < 0.05, η2 = 0.11). SuHx mice exhibit a modest compensation in diaphragm contractility and mitochondrial dysfunction during coupled respiration; the latter partially regulated through ST2 signaling.
Collapse
Affiliation(s)
- Daniel T Cannon
- School of Exercise & Nutritional Sciences, San Diego State University, United States.
| | - Leonardo Nogueira
- Department of Medicine, University of California, San Diego, United States; Instituto de Bioquímica Médica Leopoldo de Meis, Federal University of Rio de Janeiro, Brazil
| | | | - Natalie K Gilmore
- Department of Medicine, University of California, San Diego, United States
| | - Timothy D Bigby
- Department of Medicine, University of California, San Diego, United States
| | - Ellen C Breen
- Department of Medicine, University of California, San Diego, United States
| |
Collapse
|
11
|
Factors contributing to exercise capacity in chronic thromboembolic pulmonary hypertension with near-normal hemodynamics. J Heart Lung Transplant 2021; 40:677-686. [PMID: 33879384 DOI: 10.1016/j.healun.2021.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Despite improved survival for patients with chronic thromboembolic pulmonary hypertension (CTEPH) due to progressive medical and interventional treatment, impaired exercise capacity remains common due to poorly understood mechanisms. We aimed to clarify the exercise capacity of CTEPH patients with near-normal pulmonary hemodynamics and evaluate its determinants among the hemodynamic, peripheral (e.g., oxygen use by the peripheral tissues), and muscular (e.g., skeletal muscle strength) factors. METHODS Three hundred and twenty-nine patients with CTEPH (mean age, 63 ± 12 years; men/women, 73/256) with a near-normal mean pulmonary artery pressure (≤30 mm Hg) at rest were enrolled. We assessed exercise capacity by peak oxygen consumption (peak VO2) using cardiopulmonary exercise testing with a right heart catheter. We also measured the 6-minute walk distance (6MWD) and quadriceps muscle strength. RESULTS The mean pulmonary artery pressure was 19 ± 4 mmHg and mean cardiac output was 4.8 ± 1.5 L/min at rest. The mean 6MWD was 444 ± 101 m, while the mean peak VO2 was 14.4 ± 3.9 mL/min/kg. A multivariate model that predicted 6MWD included quadriceps strength (β = 0.45, p < 0.001) and peak arterial venous oxygen difference (β = 0.29, p < 0.001). In contrast, the peak VO2 was best correlated with mPAP-CO slope (β = -0.30, p < 0.001), followed by quadriceps strength and peak arterial venous oxygen difference. CONCLUSIONS The 6MWD performance may be significantly influenced by peripheral oxygen use and muscular factors, while peak VO2 is influenced by hemodynamic and peripheral factors in CTEPH patients with near-normal hemodynamics.
Collapse
|
12
|
Sree Raman K, Shah R, Stokes M, Walls A, Woodman RJ, Ananthakrishna R, Walker JG, Proudman S, Steele PM, De Pasquale CG, Celermajer DS, Selvanayagam JB. Left ventricular ischemia in pre-capillary pulmonary hypertension: a cardiovascular magnetic resonance study. Cardiovasc Diagn Ther 2020; 10:1280-1292. [PMID: 33224752 DOI: 10.21037/cdt-20-698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Prognosis in pulmonary arterial hypertension (PAH) is largely dependent on right ventricular (RV) function. However, recent studies have suggested the presence of left ventricular (LV) dysfunction in PAH patients. The potential role of LV ischemia, as a contributor to progressive LV dysfunction, has not been systematically studied in PAH. We aim to assess the presence and extent of LV myocardial ischemia in patients with known PH and without obstructive coronary artery disease (CAD), using oxygen-sensitive (OS) cardiovascular magnetic resonance (CMR) and stress/rest CMR T1 mapping. Methods We prospectively recruited 28 patients with right heart catheter-proven PH and no significant CAD, 8 patients with known CAD and 11 normal age-matched controls (NC). OS-CMR images were acquired using a T2* sequence and T1 maps were acquired using Shortened Modified Look-Locker Inversion recovery (ShMOLLI) at rest and adenosine-induced stress vasodilatation; ΔOS-CMR signal intensity (SI) index (stress/rest SI) and ΔT1 reactivity (stress-rest/rest T1 mapping) were calculated. Results Global LV ΔOS SI index was significantly lower in PH patients compared with controls (11.1%±6.7% vs. 20.5%±10.5%, P=0.016), as was ΔT1 reactivity (5.2%±4.5% vs. 8.0%±2.9%, P=0.047). The ischemic segments of CAD patients had comparable ΔOS SI (10.3%±6.4% vs. 11.1%±6.7%, P=0.773) to PH patients, but lower ΔT1 reactivity (1.1%±4.2% vs. 5.2%±4.5%, P=0.036). Conclusions Decreased OS-CMR SI and T1 reactivity signify the presence of impaired myocardial oxygenation and vasodilatory response in PH patients. Given their unobstructed epicardial coronary arteries, this is likely secondary to coronary microvascular dysfunction (CMD).
Collapse
Affiliation(s)
- Karthigesh Sree Raman
- College of Medicine and Public Health, Flinders University, Flinders, Australia.,Flinders Medical Centre, Flinders, Australia.,Cardiac Imaging Research, South Australian Health & Medical Research Institute, Australia.,Whangarei Hospital, Northland District Health Board, Whangarei, New Zealand.,Department of Medicine (Northland Campus), Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ranjit Shah
- College of Medicine and Public Health, Flinders University, Flinders, Australia.,Flinders Medical Centre, Flinders, Australia.,Cardiac Imaging Research, South Australian Health & Medical Research Institute, Australia
| | - Michael Stokes
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Angela Walls
- Clinical Research and Imaging Centre, South Australian Health & Medical Research Institute, Adelaide, South Australia, Australia
| | - Richard J Woodman
- Flinders Centre of Epidemiology and Biostatistics, College of Medicine and Public Health, Flinders University, Flinders, Australia
| | - Rajiv Ananthakrishna
- College of Medicine and Public Health, Flinders University, Flinders, Australia.,Flinders Medical Centre, Flinders, Australia.,Cardiac Imaging Research, South Australian Health & Medical Research Institute, Australia
| | | | - Susanna Proudman
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Peter M Steele
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Carmine G De Pasquale
- College of Medicine and Public Health, Flinders University, Flinders, Australia.,Flinders Medical Centre, Flinders, Australia
| | - David S Celermajer
- Sydney Medical School, University of Sydney and Royal Prince Alfred Hospital, Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Joseph B Selvanayagam
- College of Medicine and Public Health, Flinders University, Flinders, Australia.,Flinders Medical Centre, Flinders, Australia.,Cardiac Imaging Research, South Australian Health & Medical Research Institute, Australia
| |
Collapse
|
13
|
Joseph P, Oliveira RKF, Eslam RB, Agarwal M, Waxman AB, Systrom DM. Fick principle and exercise pulmonary hemodynamic determinants of the six-minute walk distance in pulmonary hypertension. Pulm Circ 2020; 10:2045894020957576. [PMID: 32994925 PMCID: PMC7502687 DOI: 10.1177/2045894020957576] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 08/13/2020] [Indexed: 01/12/2023] Open
Abstract
The six-minute walk test is widely used to assess the severity and prognosis of
pulmonary hypertension. However, the pathophysiology underlying a compromised
six-minute walk distance is incompletely characterized. The purpose of this
study is to evaluate the Fick principle and pulmonary hemodynamic determinants
of the six-minute walk distance in patients with suspected pulmonary
hypertension. Twenty-nine patients were retrospectively studied and underwent a
right heart catheterization for the evaluation of suspected pulmonary
hypertension. With the pulmonary artery catheter in place, patients were moved
to a treadmill and completed a six-minute walk test. Fick cardiac output and
indices of right heart afterload were calculated using continuous measurements
of pulmonary vascular pressures, gas exchange, and mixed venous blood samples.
Fifteen subjects who walked ≤ 348 m were compared to 14 subjects who
walked > 348 m. Systemic oxygen delivery was impaired in six-minute walk
distance ≤ 348 m compared to six-minute walk distance > 348 m (15.2 ± 6.2 vs.
23.2 ± 6.8 mL/kg/min, p < 0.01). Impaired oxygen delivery
was due to a depressed cardiac index and decreased cardiac reserve demonstrated
by the change in the stroke volume index (3.0 ± 14 vs.
17 ± 15 mL/min/m2, p = 0.02). The six-minute
walk distance positively correlated with oxygen delivery
(r = 0.501, p = 0.006) and inversely
correlated with oxygen extraction (r = 0.369,
p = 0.049). A decreased six-minute walk distance was
associated with an increased total pulmonary resistance
(r = 0.502, p = 0.006) and pulmonary vascular
resistance (r = 0.530, p = 0.003). In patients
with suspected pulmonary hypertension, a decreased six-minute walk distance is
due to compromised oxygen delivery, decreased cardiac reserve, and increased
right ventricular afterload.
Collapse
Affiliation(s)
- Phillip Joseph
- Department of Medicine, Yale School of Medicine/Yale New Haven Hospital, New Haven, CT, USA
| | - Rudolf K F Oliveira
- Department of Medicine, Federal University of Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Roza B Eslam
- Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Manyoo Agarwal
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Aaron B Waxman
- Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - David M Systrom
- Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Rosenkranz S, Howard LS, Gomberg-Maitland M, Hoeper MM. Systemic Consequences of Pulmonary Hypertension and Right-Sided Heart Failure. Circulation 2020; 141:678-693. [PMID: 32091921 DOI: 10.1161/circulationaha.116.022362] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pulmonary hypertension (PH) is a feature of a variety of diseases and continues to harbor high morbidity and mortality. The main consequence of PH is right-sided heart failure which causes a complex clinical syndrome affecting multiple organ systems including left heart, brain, kidneys, liver, gastrointestinal tract, skeletal muscle, as well as the endocrine, immune, and autonomic systems. Interorgan crosstalk and interdependent mechanisms include hemodynamic consequences such as reduced organ perfusion and congestion as well as maladaptive neurohormonal activation, oxidative stress, hormonal imbalance, and abnormal immune cell signaling. These mechanisms, which may occur in acute, chronic, or acute-on-chronic settings, are common and precipitate adverse functional and structural changes in multiple organs which contribute to increased morbidity and mortality. While the systemic character of PH and right-sided heart failure is often neglected or underestimated, such consequences place additional burden on patients and may represent treatable traits in addition to targeted therapy of PH and underlying causes. Here, we highlight the current state-of-the-art understanding of the systemic consequences of PH and right-sided heart failure on multiple organ systems, focusing on self-perpetuating pathophysiological mechanisms, aspects of increased susceptibility of organ damage, and their reciprocal impact on the course of the disease.
Collapse
Affiliation(s)
- Stephan Rosenkranz
- Clinic III for Internal Medicine (Cardiology) and Cologne Cardiovascular Research Center (CCRC), Heart Center at the University of Cologne, Germany (S.R.).,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany (S.R.)
| | - Luke S Howard
- National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, London, United Kingdom (L.S.H.)
| | | | - Marius M Hoeper
- Department of Respiratory Medicine, Hannover Medical School, Germany (M.M.H.).,German Center for Lung Research (DZL), Hannover, Germany (M.M.H.)
| |
Collapse
|
15
|
Riou M, Pizzimenti M, Enache I, Charloux A, Canuet M, Andres E, Talha S, Meyer A, Geny B. Skeletal and Respiratory Muscle Dysfunctions in Pulmonary Arterial Hypertension. J Clin Med 2020; 9:jcm9020410. [PMID: 32028638 PMCID: PMC7073630 DOI: 10.3390/jcm9020410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease, which leads to the progressive loss and remodeling of the pulmonary vessels, right heart failure, and death. Different clinical presentations can be responsible for such a bad prognosis disease and the underlying mechanisms still need to be further examined. Importantly, skeletal and respiratory muscle abnormalities largely contribute to the decreased quality of life and exercise intolerance observed in patients with PAH. At the systemic level, impaired oxygen supply through reduced cardiac output and respiratory muscle dysfunctions, which potentially result in hypoxemia, is associated with altered muscles vascularization, inflammation, enhanced catabolic pathways, and impaired oxygen use through mitochondrial dysfunctions that are likely participate in PAH-related myopathy. Sharing new insights into the pathological mechanisms of PAH might help stimulate specific research areas, improving the treatment and quality of life of PAH patients. Indeed, many of these muscular impairments are reversible, strongly supporting the development of effective preventive and/or therapeutic approaches, including mitochondrial protection and exercise training.
Collapse
Affiliation(s)
- Marianne Riou
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
- Pulmonology Service, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Mégane Pizzimenti
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Irina Enache
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Anne Charloux
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Mathieu Canuet
- Pulmonology Service, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Emmanuel Andres
- Internal Medicine, Diabete and Metabolic Diseases Service, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg CEDEX, France;
| | - Samy Talha
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Alain Meyer
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Bernard Geny
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 11 rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
- Correspondence:
| |
Collapse
|
16
|
Diaphragm function does not independently predict exercise intolerance in patients with precapillary pulmonary hypertension after adjustment for right ventricular function. Biosci Rep 2019; 39:BSR20190392. [PMID: 31427479 PMCID: PMC6723707 DOI: 10.1042/bsr20190392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022] Open
Abstract
Background: Several determinants of exercise intolerance in patients with precapillary pulmonary hypertension (PH) due to pulmonary arterial hypertension and/or chronic thromboembolic PH (CTEPH) have been suggested, including diaphragm dysfunction. However, these have rarely been evaluated in a multimodal manner. Methods: Forty-three patients with PH (age 58 ± 17 years, 30% male) and 43 age- and gender-matched controls (age 54 ± 13 years, 30% male) underwent diaphragm function (excursion and thickening) assessment by ultrasound, standard spirometry, arterial blood gas analysis, echocardiographic assessment of pulmonary artery pressure (PAP), assay of amino-terminal pro-brain natriuretic peptide (NT-proBNP) levels, and cardiac magnetic resonance (CMR) imaging to evaluate right ventricular systolic ejection fraction (RVEF). Exercise capacity was determined using the 6-min walk distance (6MWD). Results: Excursion velocity during a sniff maneuver (SniffV, 4.5 ± 1.7 vs. 6.8 ± 2.3 cm/s, P<0.01) and diaphragm thickening ratio (DTR, 1.7 ± 0.5 vs. 2.8 ± 0.8, P<0.01) were significantly lower in PH patients versus controls. PH patients with worse exercise tolerance (6MWD <377 vs. ≥377 m) were characterized by worse SniffV, worse DTR, and higher NT-pro-BNP levels as well as by lower arterial carbon dioxide levels and RVEF, which were all univariate predictors of exercise limitation. On multivariate analysis, the only independent predictors of exercise limitation were RVEF (r = 0.47, P=0.001) and NT-proBNP (r = −0.27, P=0.047). Conclusion: Patients with PH showed diaphragm dysfunction, especially as exercise intolerance progressed. However, diaphragm dysfunction does not independently contribute to exercise intolerance, beyond what can be explained from right heart failure.
Collapse
|
17
|
Anti-inflammatory nutrition with high protein attenuates cardiac and skeletal muscle alterations in a pulmonary arterial hypertension model. Sci Rep 2019; 9:10160. [PMID: 31308383 PMCID: PMC6629640 DOI: 10.1038/s41598-019-46331-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/23/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by remodelling of the pulmonary arteries and right ventricle (RV), which leads to functional decline of cardiac and skeletal muscle. This study investigated the effects of a multi-targeted nutritional intervention with extra protein, leucine, fish oil and oligosaccharides on cardiac and skeletal muscle in PAH. PAH was induced in female C57BL/6 mice by weekly injections of monocrotaline (MCT) for 8 weeks. Control diet (sham and MCT group) and isocaloric nutritional intervention (MCT + NI) were administered. Compared to sham, MCT mice increased heart weight by 7%, RV thickness by 13% and fibrosis by 60% (all p < 0.05) and these were attenuated in MCT + NI mice. Microarray and qRT-PCR analysis of RV confirmed effects on fibrotic pathways. Skeletal muscle fiber atrophy was induced (P < 0.05) by 22% in MCT compared to sham mice, but prevented in MCT + NI group. Our findings show that a multi-targeted nutritional intervention attenuated detrimental alterations to both cardiac and skeletal muscle in a mouse model of PAH, which provides directions for future therapeutic strategies targeting functional decline of both tissues.
Collapse
|
18
|
Grünig E, Eichstaedt C, Barberà JA, Benjamin N, Blanco I, Bossone E, Cittadini A, Coghlan G, Corris P, D'Alto M, D'Andrea A, Delcroix M, de Man F, Gaine S, Ghio S, Gibbs S, Gumbiene L, Howard LS, Johnson M, Jurevičienė E, Kiely DG, Kovacs G, MacKenzie A, Marra AM, McCaffrey N, McCaughey P, Naeije R, Olschewski H, Pepke-Zaba J, Reis A, Santos M, Saxer S, Tulloh RM, Ulrich S, Vonk Noordegraaf A, Peacock AJ. ERS statement on exercise training and rehabilitation in patients with severe chronic pulmonary hypertension. Eur Respir J 2018; 53:13993003.00332-2018. [DOI: 10.1183/13993003.00332-2018] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 09/12/2018] [Indexed: 12/17/2022]
Abstract
Objectives of this European Respiratory Society task force were to summarise current studies, to develop strategies for future research and to increase availability and awareness of exercise training for pulmonary hypertension (PH) patients.An evidence-based approach with clinical expertise of the task force members, based on both literature search and face-to-face meetings was conducted. The statement summarises current knowledge and open questions regarding clinical effects of exercise training in PH, training modalities, implementation strategies and pathophysiological mechanisms.In studies (784 PH patients in total, including six randomised controlled trials, three controlled trials, 10 prospective cohort studies and four meta-analyses), exercise training has been shown to improve exercise capacity, muscular function, quality of life and possibly right ventricular function and pulmonary haemodynamics. Nevertheless, further studies are needed to confirm these data, to investigate the impact on risk profiles and to identify the most advantageous training methodology and underlying pathophysiological mechanisms.As exercise training appears to be effective, cost-efficient and safe, but is scarcely reimbursed, support from healthcare institutions, commissioners of healthcare and research funding institutions is greatly needed. There is a strong need to establish specialised rehabilitation programmes for PH patients to enhance patient access to this treatment intervention.
Collapse
|
19
|
Inspiratory muscle weakness contributes to exertional dyspnea in chronic thromboembolic pulmonary hypertension. PLoS One 2018; 13:e0204072. [PMID: 30260970 PMCID: PMC6160017 DOI: 10.1371/journal.pone.0204072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/31/2018] [Indexed: 11/20/2022] Open
Abstract
Determination of potentially-reversible factors contributing to exertional dyspnea remains an unmet clinical need in chronic thromboembolic pulmonary hypertension (CTEPH). Therefore, we aimed to evaluate the influence of inspiratory muscle weakness (IMW) on exercise capacity and dyspnea during effort in patients with CTEPH. We performed a prospective cross-sectional study that included thirty-nine consecutive patients with CTEPH (48 ± 15 yrs, 61% female) confirmed by right heart catheterization that underwent an incremental cardiopulmonary exercise test, 6-minute walk test and maximum inspiratory pressure (MIP) measurement. MIP < 70%pred was found in 46% of patients. On a multiple linear regression analysis, MIP was independently associated with 6MWD and V˙O2PEAK. Patients with MIP < 70% presented greater ΔV˙E/ΔV˙CO2 than those with MIP ≥ 70%. Additionally, they also presented stronger sensations of dyspnea throughout exercise, even when adjusted for ventilation. At rest and at different levels of exercise, mean inspiratory flow (VT/TI) was significantly higher in patients with MIP < 70%. In conclusion, IMW is associated with a rapid increase of dyspnea, higher inspiratory load and poor exercise capacity in patients with CTEPH.
Collapse
|
20
|
Oldham WM, Oliveira RKF, Wang RS, Opotowsky AR, Rubins DM, Hainer J, Wertheim BM, Alba GA, Choudhary G, Tornyos A, MacRae CA, Loscalzo J, Leopold JA, Waxman AB, Olschewski H, Kovacs G, Systrom DM, Maron BA. Network Analysis to Risk Stratify Patients With Exercise Intolerance. Circ Res 2018; 122:864-876. [PMID: 29437835 PMCID: PMC5924425 DOI: 10.1161/circresaha.117.312482] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 01/09/2023]
Abstract
RATIONALE Current methods assessing clinical risk because of exercise intolerance in patients with cardiopulmonary disease rely on a small subset of traditional variables. Alternative strategies incorporating the spectrum of factors underlying prognosis in at-risk patients may be useful clinically, but are lacking. OBJECTIVE Use unbiased analyses to identify variables that correspond to clinical risk in patients with exercise intolerance. METHODS AND RESULTS Data from 738 consecutive patients referred for invasive cardiopulmonary exercise testing at a single center (2011-2015) were analyzed retrospectively (derivation cohort). A correlation network of invasive cardiopulmonary exercise testing parameters was assembled using |r|>0.5. From an exercise network of 39 variables (ie, nodes) and 98 correlations (ie, edges) corresponding to P<9.5e-46 for each correlation, we focused on a subnetwork containing peak volume of oxygen consumption (pVo2) and 9 linked nodes. K-mean clustering based on these 10 variables identified 4 novel patient clusters characterized by significant differences in 44 of 45 exercise measurements (P<0.01). Compared with a probabilistic model, including 23 independent predictors of pVo2 and pVo2 itself, the network model was less redundant and identified clusters that were more distinct. Cluster assignment from the network model was predictive of subsequent clinical events. For example, a 4.3-fold (P<0.0001; 95% CI, 2.2-8.1) and 2.8-fold (P=0.0018; 95% CI, 1.5-5.2) increase in hazard for age- and pVo2-adjusted all-cause 3-year hospitalization, respectively, were observed between the highest versus lowest risk clusters. Using these data, we developed the first risk-stratification calculator for patients with exercise intolerance. When applying the risk calculator to patients in 2 independent invasive cardiopulmonary exercise testing cohorts (Boston and Graz, Austria), we observed a clinical risk profile that paralleled the derivation cohort. CONCLUSIONS Network analyses were used to identify novel exercise groups and develop a point-of-care risk calculator. These data expand the range of useful clinical variables beyond pVo2 that predict hospitalization in patients with exercise intolerance.
Collapse
Affiliation(s)
- William M Oldham
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Rudolf K F Oliveira
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Rui-Sheng Wang
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Alexander R Opotowsky
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - David M Rubins
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Jon Hainer
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Bradley M Wertheim
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - George A Alba
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Gaurav Choudhary
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Adrienn Tornyos
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Calum A MacRae
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Joseph Loscalzo
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Jane A Leopold
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Aaron B Waxman
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Horst Olschewski
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Gabor Kovacs
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - David M Systrom
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.)
| | - Bradley A Maron
- From the Department of Medicine (W.M.O., R.K.F.O., R.-S.W., D.M.R., B.M.W., C.A.M., J.L., A.B.W., D.M.S., J.A.L.), Division of Pulmonary and Critical Care Medicine (W.M.O., B.M.W., A.B.W., D.M.S.), Division of Cardiovascular Medicine (A.R.O., C.A.M., J.L., J.A.L., B.A.M.), and Department of Radiology (J.H.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Brazil (R.K.F.O.); Department of Cardiology, Boston Children's Hospital and Harvard Medical School, MA (A.R.O.); Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (G.A.A.); Division of Cardiology, Department of Medicine, Providence Veterans Affairs Medical Center and Alpert Medical School of Brown University, Providence, RI (G.C.); Department of Pulmonology, Medical University of Graz, Austria (A.T., H.O., G.K.); Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (A.T., H.O., G.K.); and Department of Cardiology, Boston VA Healthcare System, MA (B.A.M.).
| |
Collapse
|
21
|
Dumitrescu D, Sitbon O, Weatherald J, Howard LS. Exertional dyspnoea in pulmonary arterial hypertension. Eur Respir Rev 2017; 26:26/145/170039. [PMID: 28877974 PMCID: PMC9488798 DOI: 10.1183/16000617.0039-2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 06/03/2017] [Indexed: 01/01/2023] Open
Abstract
Dyspnoea is a principal presenting symptom in pulmonary arterial hypertension (PAH), and often the most distressing. The pathophysiology of PAH is relatively well understood, with the primary abnormality of pulmonary vascular disease resulting in a combination of impaired cardiac output on exercise and abnormal gas exchange, both contributing to increased ventilatory drive. However, increased ventilatory drive is not the sole explanation for the complex neurophysiological and neuropsychological symptom of dyspnoea, with other significant contributions from skeletal muscle reflexes, respiratory muscle function, and psychological and emotional status. In this review, we explore the physiological aspects of dyspnoea in PAH, both in terms of the central cardiopulmonary abnormalities of PAH and the wider, systemic impact of PAH, and how these interact with common comorbidities. Finally, we discuss its relationship with disease severity. Dyspnoea is a complex integration of all the cardiopulmonary and systemic abnormalities in PAHhttp://ow.ly/D13W30dMDwJ
Collapse
|
22
|
Tran DL, Lau EM, Celermajer DS, Davis GM, Cordina R. Pathophysiology of exercise intolerance in pulmonary arterial hypertension. Respirology 2017; 23:148-159. [DOI: 10.1111/resp.13141] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 05/15/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Derek L. Tran
- Faculty of Health Sciences; The University of Sydney; Sydney NSW Australia
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences; Macquarie University; Sydney NSW Australia
- Pulmonary Hypertension Service; Royal Prince Alfred Hospital; Sydney NSW Australia
| | - Edmund M.T. Lau
- Pulmonary Hypertension Service; Royal Prince Alfred Hospital; Sydney NSW Australia
- Sydney Medical School; The University of Sydney; Sydney NSW Australia
| | - David S. Celermajer
- Pulmonary Hypertension Service; Royal Prince Alfred Hospital; Sydney NSW Australia
- Sydney Medical School; The University of Sydney; Sydney NSW Australia
| | - Glen M. Davis
- Faculty of Health Sciences; The University of Sydney; Sydney NSW Australia
| | - Rachael Cordina
- Pulmonary Hypertension Service; Royal Prince Alfred Hospital; Sydney NSW Australia
- Sydney Medical School; The University of Sydney; Sydney NSW Australia
| |
Collapse
|
23
|
Hautefort A, Chesné J, Preussner J, Pullamsetti SS, Tost J, Looso M, Antigny F, Girerd B, Riou M, Eddahibi S, Deleuze JF, Seeger W, Fadel E, Simonneau G, Montani D, Humbert M, Perros F. Pulmonary endothelial cell DNA methylation signature in pulmonary arterial hypertension. Oncotarget 2017; 8:52995-53016. [PMID: 28881789 PMCID: PMC5581088 DOI: 10.18632/oncotarget.18031] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/09/2017] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe and incurable pulmonary vascular disease. One of the primary origins of PAH is pulmonary endothelial dysfunction leading to vasoconstriction, aberrant angiogenesis and smooth muscle cell proliferation, endothelial-to-mesenchymal transition, thrombosis and inflammation. Our objective was to study the epigenetic variations in pulmonary endothelial cells (PEC) through a specific pattern of DNA methylation. DNA was extracted from cultured PEC from idiopathic PAH (n = 11), heritable PAH (n = 10) and controls (n = 18). DNA methylation was assessed using the Illumina HumanMethylation450 Assay. After normalization, samples and probes were clustered according to their methylation profile. Differential clusters were functionally analyzed using bioinformatics tools. Unsupervised hierarchical clustering allowed the identification of two clusters of probes that discriminates controls and PAH patients. Among 147 differential methylated promoters, 46 promoters coding for proteins or miRNAs were related to lipid metabolism. Top 10 up and down-regulated genes were involved in lipid transport including ABCA1, ABCB4, ADIPOQ, miR-26A, BCL2L11. NextBio meta-analysis suggested a contribution of ABCA1 in PAH. We confirmed ABCA1 mRNA and protein downregulation specifically in PAH PEC by qPCR and immunohistochemistry and made the proof-of-concept in an experimental model of the disease that its targeting may offer novel therapeutic options. In conclusion, DNA methylation analysis identifies a set of genes mainly involved in lipid transport pathway which could be relevant to PAH pathophysiology.
Collapse
Affiliation(s)
- Aurélie Hautefort
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Julie Chesné
- UMR_S 1087 CNRS UMR_6291, Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - Jens Preussner
- Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Soni S Pullamsetti
- Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Jorg Tost
- Centre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Mario Looso
- Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Fabrice Antigny
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Barbara Girerd
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
- AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marianne Riou
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Saadia Eddahibi
- INSERM U1046, Centre Hospitalier Universitaire Arnaud de Villeneuve, Montpellier, France
| | | | - Werner Seeger
- Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Elie Fadel
- Hôpital Marie Lannelongue, Service de Chirurgie Thoracique et Vasculaire, Le Plessis Robinson, France
| | - Gerald Simonneau
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
- AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - David Montani
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
- AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
- AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Frédéric Perros
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Univ Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
24
|
Left Ventricular Myocardial Fibrosis, Atrophy, and Impaired Contractility in Patients With Pulmonary Arterial Hypertension and a Preserved Left Ventricular Function: A Cardiac Magnetic Resonance Study. J Thorac Imaging 2017; 32:36-42. [PMID: 27861208 DOI: 10.1097/rti.0000000000000248] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Using a cardiac magnetic resonance (CMR) approach we investigated left ventricular (LV) myocardial changes associated with pulmonary arterial hypertension (PAH) by strain analysis and mapping techniques. MATERIALS AND METHODS Seventeen patients with PAH (9 men; mean age, 64.2±13.6 y) and 20 controls (10 men, 63.2±10.5 y) were examined using CMR at 1.5 T. Native LV T1-relaxation times (T1) and extracellular volume fraction (ECV) were assessed using a MOLLI sequence, T2-relaxation times (T2) by means of a gradient spin-echo sequence, and LV longitudinal strain (LVS) and right ventricular (RV) longitudinal strain (RVS) by means of CMR feature tracking. The hematocrit and serum levels of pro-Brain Natriuretic Peptide were determined on the day of the CMR examination. Pulmonary arterial pressure and 6-minute walking distance were assessed as part of the clinical evaluation. RESULTS T1 and ECV were higher (1048.5±46.6 vs. 968.3±22.9 ms and 32.4%±5.7% vs. 28.4%±3.8%; P<0.05) and LVS was lower in patients with PAH (-18.0±5.6 vs. -23.0±2.9; P<0.01) compared with controls. LV mass and interventricular septal thickness were lower in PAH patients (65.7±18.0 vs. 86.7±26.9 g and 7.6±1.9 vs. 10±2.4 mm; P<0.05); there were no differences in LV ejection fraction (61.2%±6.9% vs. 61.9%±6.7%; P=0.86). T1-derived parameters correlated significantly with RVS, LVS, the 6-minute walking distance, RV ejection fraction, pro-Brain Natriuretic Peptide, and baseline mean pulmonary arterial pressure. There were no significant differences in T2. CONCLUSIONS In patients with PAH, changes in T1 and ECV support the hypothesis of LV myocardial fibrosis and atrophy with a consecutively impaired contractility despite a preserved LV function, possibly due to longstanding PAH-associated LV underfilling.
Collapse
|
25
|
Stewart Coats AJ, Shewan LG. A comparison of research into cachexia, wasting and related skeletal muscle syndromes in three chronic disease areas. Int J Cardiol 2017; 235:33-36. [PMID: 28291621 DOI: 10.1016/j.ijcard.2017.02.136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/27/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We compared the frequency of cancer, heart and lung related cachexia and cachexia-related research articles in the specialist journal, Journal of Cachexia, Sarcopenia and Muscle (JCSM) to those seen in a leading European journal in each specialist area during 2015 and 2016 to assess whether work on cachexia and related fields is relatively over or under represented in each specialist area. RESULTS In the dedicated journal, Journal of Cachexia, Sarcopenia and Muscle, there were 44 references related to cancer, 5 related to respiratory disease, 5 related to heart failure, and 21 related to more than one of these chronic diseases. Despite this cancer preponderance, in the European Journal of Cancer in the two publication years, there were only 5 relevant publications (0.67% of the journal output), compared to 16 (1.41%) in the European Respiratory Journal and 10 (2.19%) in the European Journal of Heart Failure. CONCLUSIONS There is considerable under-representation of cancer cachexia-related papers in the major European Cancer journal despite a high proportion in the dedicated cachexia journal. The under-representation is even more marked when expressed as a percentage, 0.67%, compared to 1.41% and 2.19% of the lung and heart journals respectively. These results are consistent with a worrying lack of interest in, or publication of, cachexia and related syndromes research in the cancer literature in Europe compared to its importance as a clinical syndrome. Greater interest is shown in lung and cardiology journals.
Collapse
Affiliation(s)
| | - Louise G Shewan
- Monash University, Australia; University of Warwick, Coventry, UK
| |
Collapse
|
26
|
Dimopoulos S, Tzanis G, Karabinis A, Nanas S. Dynamic near-infrared spectroscopy assessment as an important tool to explore pulmonary arterial hypertension pathophysiology. Eur Respir J 2017; 49:49/1/1601932. [PMID: 28052959 DOI: 10.1183/13993003.01932-2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/08/2016] [Indexed: 02/07/2023]
Affiliation(s)
- Stavros Dimopoulos
- Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, National and Kapodistrian University of Athens, Athens, Greece
- 1st Critical Care Medicine Dept, Evaggelismos Hospital, Athens, Greece
- Cardiothoracic Intensive Care Unit, Onassis Cardiac Surgery Center, Athens, Greece
| | - Giorgos Tzanis
- Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, National and Kapodistrian University of Athens, Athens, Greece
- 1st Critical Care Medicine Dept, Evaggelismos Hospital, Athens, Greece
| | - Andreas Karabinis
- Cardiothoracic Intensive Care Unit, Onassis Cardiac Surgery Center, Athens, Greece
| | - Serafim Nanas
- Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, National and Kapodistrian University of Athens, Athens, Greece
- 1st Critical Care Medicine Dept, Evaggelismos Hospital, Athens, Greece
| |
Collapse
|
27
|
Maron BA, Galiè N. Diagnosis, Treatment, and Clinical Management of Pulmonary Arterial Hypertension in the Contemporary Era: A Review. JAMA Cardiol 2016; 1:1056-1065. [PMID: 27851839 PMCID: PMC5177491 DOI: 10.1001/jamacardio.2016.4471] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
IMPORTANCE Pulmonary arterial hypertension (PAH) is characterized by severe remodeling of the distal pulmonary arteries, increased pulmonary vascular resistance, and right ventricular dysfunction that promotes heart failure. Once regarded as largely untreatable, evidence-based decision making now guides clinical management of PAH and improves outcomes. However, misconceptions regarding the approach to PAH in the modern era are common and associated with substandard clinical care. OBSERVATIONS The clinical profile of PAH has changed substantially since its original description. Patients are older at diagnosis than previously reported; disease severity appears greater in men compared with women; and patients with PAH in association with connective tissue disease are identified as a particularly high-risk subgroup. Risk stratification scales for PAH are now available at point of care, which inform treatment goals, including a 6-minute walk distance of greater than 440 m, peak volume of oxygen consumption of greater than 15 mL/min/kg, right atrial area of less than 18 cm2, cardiac index of greater than 2.5 L/min/m2, and absent or low symptom burden with routine physical activity. At present, 14 therapies targeting 6 PAH-specific molecular intermediaries are used clinically. Recent landmark trial data have demonstrated the critical importance of initial combination therapy in treatment-naive patients. These findings underscore a global shift in PAH that couples early disease detection with aggressive pharmacotherapy. Indeed, recent longitudinal data from patients receiving combination therapy show that the 3-year survival rate in PAH may be as high as 84% compared with 48% from the original National Institutes of Health registry on idiopathic PAH (1980-1985). Despite these gains, incomplete clinical evaluation and misdiagnosis by referring clinicians is common and associated with inappropriate therapy. CONCLUSIONS AND RELEVANCE Compared with the original clinical experience, PAH has evolved into a contemporary and treatable disease characterized by improved survival and a high standard for defining therapeutic success. However, underawareness among clinicians regarding the importance of early and accurate PAH diagnosis persists and is a potentially reversible cause of adverse outcome in this disease.
Collapse
Affiliation(s)
- Bradley A. Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA and Department of Cardiology, Boston VA Healthcare System, Boston, MA, USA
| | - Nazzareno Galiè
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, via Massarenti 9, Bologna 40138, Italy
| |
Collapse
|
28
|
Abstract
Over the past 20 years, there has been an explosion in the development of therapeutics to treat pulmonary arterial hypertension (PAH), a rare but life-threatening disorder associated with progressive elevation of pulmonary pressures and severe right heart failure. Recently, the field has seen the introduction of riociguat, a soluble guanylate cyclase stimulator, a new endothelin receptor antagonist (macitentan), and oral prostanoids (treprostinil and selexipag). Besides new drugs, there have been significant advances in defining the role of upfront combination therapy in treatment-naïve patients as well as proposed methods to deliver systemic prostanoids by use of implantable pumps. In this review, we will touch upon the most important developments in PAH therapeutics over the last three years and how these have changed the guidelines for the treatment of PAH. These exciting developments herald a new era in the treatment of PAH which will be punctuated by the use of more clinically relevant endpoints in clinical research trials and a novel treatment paradigm that may involve upfront double- or triple-combination therapy. We anticipate that the future will make use of these strategies to test the efficacy of upcoming new drugs that aspire to reduce disease progression and improve survival in patients afflicted with this devastating disease.
Collapse
Affiliation(s)
- Halley Tsai
- Division of Pulmonary/Critical Care, Stanford University School of Medicine, Stanford, CA, 94305-5236, USA
| | - Yon K Sung
- Division of Pulmonary/Critical Care, Stanford University School of Medicine, Stanford, CA, 94305-5236, USA.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA, 94305-5414, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary/Critical Care, Stanford University School of Medicine, Stanford, CA, 94305-5236, USA.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA, 94305-5414, USA
| |
Collapse
|
29
|
Alencar AK, Montes GC, Montagnoli T, Silva AM, Martinez ST, Fraga AG, Wang H, Groban L, Sudo RT, Zapata-Sudo G. Activation of GPER ameliorates experimental pulmonary hypertension in male rats. Eur J Pharm Sci 2016; 97:208-217. [PMID: 27836751 DOI: 10.1016/j.ejps.2016.11.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/29/2016] [Accepted: 11/07/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling that leads to pulmonary congestion, uncompensated right-ventricle (RV) failure, and premature death. Preclinical studies have demonstrated that the G protein-coupled estrogen receptor (GPER) is cardioprotective in male rats and that its activation elicits vascular relaxation in rats of either sex. OBJECTIVES To study the effects of GPER on the cardiopulmonary system by the administration of its selective agonist G1 in male rats with monocrotaline (MCT)-induced PH. METHODS Rats received a single intraperitoneal injection of MCT (60mg/kg) for PH induction. Experimental groups were as follows: control, MCT+vehicle, and MCT+G1 (400μg/kg/daysubcutaneous). Animals (n=5pergroup) were treated with vehicle or G1 for 14days after disease onset. MEASUREMENTS AND MAIN RESULTS Activation of GPER attenuated exercise intolerance and reduced RV overload in PH rats. Rats with PH exhibited echocardiographic alterations, such as reduced pulmonary flow, RV hypertrophy, and left-ventricle dysfunction, by the end of protocol. G1 treatment reversed these PH-related abnormalities of cardiopulmonary function and structure, in part by promoting pulmonary endothelial nitric oxide synthesis, Ca2+ handling regulation and reduction of inflammation in cardiomyocytes, and a decrease of collagen deposition by acting in pulmonary and cardiac fibroblasts. CONCLUSIONS G1 was effective to reverse PH-induced RV dysfunction and exercise intolerance in male rats, a finding that have important implications for ongoing clinical evaluation of new cardioprotective and vasodilator drugs for the treatment of the disease.
Collapse
Affiliation(s)
- Allan K Alencar
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Guilherme C Montes
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tadeu Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ananssa M Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Sabrina T Martinez
- Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Aline G Fraga
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hao Wang
- Department of Anesthesiology, Wake Forest University, Winston-Salem, NC, USA
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest University, Winston-Salem, NC, USA
| | - Roberto T Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
30
|
Manders E, Bonta PI, Kloek JJ, Symersky P, Bogaard HJ, Hooijman PE, Jasper JR, Malik FI, Stienen GJM, Vonk-Noordegraaf A, de Man FS, Ottenheijm CAC. Reduced force of diaphragm muscle fibers in patients with chronic thromboembolic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2016; 311:L20-8. [PMID: 27190061 DOI: 10.1152/ajplung.00113.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/17/2016] [Indexed: 11/22/2022] Open
Abstract
Patients with pulmonary hypertension (PH) suffer from inspiratory muscle weakness. However, the pathophysiology of inspiratory muscle dysfunction in PH is unknown. We hypothesized that weakness of the diaphragm, the main inspiratory muscle, is an important contributor to inspiratory muscle dysfunction in PH patients. Our objective was to combine ex vivo diaphragm muscle fiber contractility measurements with measures of in vivo inspiratory muscle function in chronic thromboembolic pulmonary hypertension (CTEPH) patients. To assess diaphragm muscle contractility, function was studied in vivo by maximum inspiratory pressure (MIP) and ex vivo in diaphragm biopsies of the same CTEPH patients (N = 13) obtained during pulmonary endarterectomy. Patients undergoing elective lung surgery served as controls (N = 15). Muscle fiber cross-sectional area (CSA) was determined in cryosections and contractility in permeabilized muscle fibers. Diaphragm muscle fiber CSA was not significantly different between control and CTEPH patients in both slow-twitch and fast-twitch fibers. Maximal force-generating capacity was significantly lower in slow-twitch muscle fibers of CTEPH patients, whereas no difference was observed in fast-twitch muscle fibers. The maximal force of diaphragm muscle fibers correlated significantly with MIP. The calcium sensitivity of force generation was significantly reduced in fast-twitch muscle fibers of CTEPH patients, resulting in a ∼40% reduction of submaximal force generation. The fast skeletal troponin activator CK-2066260 (5 μM) restored submaximal force generation to levels exceeding those observed in control subjects. In conclusion, diaphragm muscle fiber contractility is hampered in CTEPH patients and contributes to the reduced function of the inspiratory muscles in CTEPH patients.
Collapse
Affiliation(s)
- Emmy Manders
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, Amsterdam, The Netherlands; Department of Physiology, VU University Medical Center/Institute for Cardiovascular Research, The Netherlands
| | - Peter I Bonta
- Department of Respiratory Medicine, Amsterdam Medical Center, University of Amsterdam, The Netherlands
| | - Jaap J Kloek
- Department of Cardiothoracic Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Petr Symersky
- Department of Cardiothoracic Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Harm-Jan Bogaard
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, Amsterdam, The Netherlands
| | - Pleuni E Hooijman
- Department of Physiology, VU University Medical Center/Institute for Cardiovascular Research, The Netherlands
| | - Jeff R Jasper
- Research & Early Development, Cytokinetics Inc., South San Francisco, California
| | - Fady I Malik
- Research & Early Development, Cytokinetics Inc., South San Francisco, California
| | - Ger J M Stienen
- Department of Physiology, VU University Medical Center/Institute for Cardiovascular Research, The Netherlands; Faculty of Science, Department of Physics and Astronomy, VU University, Amsterdam, The Netherlands; and
| | - Anton Vonk-Noordegraaf
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, Amsterdam, The Netherlands
| | - Frances S de Man
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, Amsterdam, The Netherlands
| | - Coen A C Ottenheijm
- Department of Physiology, VU University Medical Center/Institute for Cardiovascular Research, The Netherlands; Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
31
|
Mendes-Ferreira P, Maia-Rocha C, Adão R, Mendes MJ, Santos-Ribeiro D, Alves BS, Cerqueira RJ, Castro-Chaves P, Lourenço AP, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Neuregulin-1 improves right ventricular function and attenuates experimental pulmonary arterial hypertension. Cardiovasc Res 2015; 109:44-54. [PMID: 26503987 DOI: 10.1093/cvr/cvv244] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/15/2015] [Indexed: 12/24/2022] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is a serious disease that affects both the pulmonary vasculature and the right ventricle (RV). Current treatment options are insufficient. The cardiac neuregulin (NRG)-1/ErbB system is deregulated during heart failure, and treatment with recombinant human NRG-1 (rhNRG-1) has been shown to be beneficial in animal models and in patients with left ventricular (LV) dysfunction. This study aimed to evaluate the effects of rhNRG-1 in RV function and pulmonary vasculature in monocrotaline (MCT)-induced PAH and RV hypertrophy (RVH). METHODS AND RESULTS Male wistar rats (7- to 8-weeks old, n = 78) were injected with MCT (60 mg/kg, s.c.) or saline and treated with rhNRG-1 (40 µg/kg/day) or vehicle for 1 week, starting 2 weeks after MCT administration. Another set of animals was submitted to pulmonary artery banding (PAB) or sham surgery, and followed the same protocol. MCT administration resulted in the development of PAH, pulmonary arterial and RV remodelling, and dysfunction, and increased RV markers of cardiac damage. Treatment with rhNRG-1 attenuated RVH, improved RV function, and decreased RV expression of disease markers. Moreover, rhNRG-1 decreased pulmonary vascular remodelling and attenuated MCT-induced endothelial dysfunction. The anti-remodelling effects of rhNRG-1 were confirmed in the PAB model, where rhNRG-1 treatment was able to attenuate PAB-induced RVH. CONCLUSION rhNRG-1 treatment attenuates pulmonary arterial and RV remodelling, and dysfunction in a rat model of MCT-induced PAH and has direct anti-remodelling effects on the pressure-overloaded RV.
Collapse
Affiliation(s)
- Pedro Mendes-Ferreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carolina Maia-Rocha
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Adão
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Maria José Mendes
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Diana Santos-Ribeiro
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Bárbara Silvana Alves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui João Cerqueira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paulo Castro-Chaves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - André Pedro Lourenço
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | | | - Adelino Ferreira Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carmen Brás-Silva
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| |
Collapse
|