1
|
Benavides-Córdoba V, Palacios M, Vonk-Noordegraaf A. Historical milestones and future horizons: exploring the diagnosis and treatment evolution of the pulmonary arterial hypertension in adults. Expert Opin Pharmacother 2025; 26:743-753. [PMID: 40091694 DOI: 10.1080/14656566.2025.2480764] [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: 11/28/2024] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION Pulmonary hypertension is a life-threatening condition characterized by elevated mean pulmonary arterial pressure and vascular resistance. Significant advances in diagnosis and treatment have been achieved over the 20th and 21st centuries, yet challenges remain in improving long-term outcomes. AREAS COVERED This review discusses the historical milestones in understanding and pharmacotherapy of the pulmonary arterial hypertension (PAH). A comprehensive literature search was conducted to explore the earliest reports of each approved medication for pulmonary hypertension, along with historical papers detailing the pathophysiological and diagnostic development. Additionally, the search aimed to identify novel therapeutic strategies, including repositioned drugs and emerging targets. EXPERT OPINION While current therapies, such as prostacyclin analogs and PDE5 inhibitors, improve functional capacity and hemodynamics, they face limitations, including costs, administration, and a predominantly vasodilatory approach. Additionally, the limitations of current clinical trial designs for rare diseases like pulmonary arterial hypertension hinder the evaluation of potentially effective drugs. These challenges underscore the urgent need for translational research to optimize trial methodologies, accelerating the development of new therapies. Innovative approaches, such as drug repositioning and the exploration of novel molecular targets, are critical to overcoming these barriers and ensuring timely, effective, and affordable treatment options for patients with PAH.
Collapse
Affiliation(s)
| | - Mauricio Palacios
- Department of Physiological Sciences, Pharmacology, Universidad del Valle, Cali, Colombia
| | - Anton Vonk-Noordegraaf
- Department of Pulmonary Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Nakamura K, Akagi S, Ejiri K, Taya S, Saito Y, Kuroda K, Takaya Y, Toh N, Nakayama R, Katanosaka Y, Yuasa S. Pathophysiology of Group 3 Pulmonary Hypertension Associated with Lung Diseases and/or Hypoxia. Int J Mol Sci 2025; 26:835. [PMID: 39859549 PMCID: PMC11765551 DOI: 10.3390/ijms26020835] [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: 11/18/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Pulmonary hypertension associated with lung diseases and/or hypoxia is classified as group 3 in the clinical classification of pulmonary hypertension. The efficacy of existing selective pulmonary vasodilators for group 3 pulmonary hypertension is still unknown, and it is currently associated with a poor prognosis. The mechanisms by which pulmonary hypertension occurs include hypoxic pulmonary vasoconstriction, pulmonary vascular remodeling, a decrease in pulmonary vascular beds, endothelial dysfunction, endothelial-to-mesenchymal transition, mitochondrial dysfunction, oxidative stress, hypoxia-inducible factors (HIFs), inflammation, microRNA, and genetic predisposition. Among these, hypoxic pulmonary vasoconstriction and subsequent pulmonary vascular remodeling are characteristic factors involving the pulmonary vasculature and are the focus of this review. Several factors have been reported to mediate vascular remodeling induced by hypoxic pulmonary vasoconstriction, such as HIF-1α and mechanosensors, including TRP channels. New therapies that target novel molecules, such as mechanoreceptors, to inhibit vascular remodeling are awaited.
Collapse
Affiliation(s)
- Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
- Center for Advanced Heart Failure, Okayama University Hospital, Okayama 700-8558, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Kentaro Ejiri
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Satoshi Taya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Kazuhiro Kuroda
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yoichi Takaya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Norihisa Toh
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Rie Nakayama
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yuki Katanosaka
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan;
- Graduate School of Pharmaceutical Sciences, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| |
Collapse
|
3
|
Kay D, Bernardo R, Elwing J. Pulmonary Vascular Disease in Chronic Obstructive Pulmonary Disease. Semin Respir Crit Care Med 2024; 45:574-581. [PMID: 39532092 DOI: 10.1055/s-0044-1791824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Pulmonary hypertension (PH) is a vascular disease characterized by pulmonary artery remodeling and right heart failure. PH related to COPD is a precapillary form of the disease, with hemodynamic measurements including a mean pulmonary artery pressure of greater than 20 mm Hg, a wedge pressure of less than 15 mm Hg, and a pulmonary vascular resistance of greater than 3 WU (Woods units), categorized under the World Health Organization classification as group 3. The presence of PH in COPD has been known to increase morbidity and mortality. Limited studies have evaluated treatment options for PH related to COPD.
Collapse
Affiliation(s)
- Dana Kay
- College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Roberto Bernardo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Jean Elwing
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
4
|
Blanco I, Torres-Castro R, Barberà JA. Pulmonary vascular disease in chronic lung diseases: cause or comorbidity? Curr Opin Pulm Med 2024; 30:437-443. [PMID: 38958570 DOI: 10.1097/mcp.0000000000001091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW To provide timely and relevant insights into the complex relationship between pulmonary vascular disease (PVD) and chronic lung disease (CLD), focusing on the causative and consequential dynamics between these conditions. RECENT FINDINGS There are shared pathogenic mechanisms between pulmonary arterial hypertension (PAH) and group 3 pulmonary hypertension, including altered expression of mediators and growth factors implicated in both conditions. Factors such as hypoxia, hypoxemia, and hypercapnia also contribute to pulmonary vascular remodelling and endothelial dysfunction. However, the role of hypoxia as the sole driver of pulmonary hypertension in CLD is being reconsidered, particularly in chronic obstructive pulmonary disease (COPD), with evidence suggesting a potential role for cigarette smoke products in initiating pulmonary vascular impairment. On the other hand, interstitial lung disease (ILD) encompasses a group of heterogeneous lung disorders characterized by inflammation and fibrosis of the interstitium, leading to impaired gas exchange and progressive respiratory decline, which could also play a role as a cause of pulmonary hypertension. SUMMARY Understanding the intricate interplay between the pulmonary vascular compartment and the parenchymal and airway compartments in respiratory disease is crucial for developing effective diagnostic and therapeutic strategies for patients with PVD and CLD, with implications for both clinical practice and research.
Collapse
Affiliation(s)
- Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES); Madrid, Spain
| | - Rodrigo Torres-Castro
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona
- Department of Physical Therapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES); Madrid, Spain
| |
Collapse
|
5
|
Mora-Cuesta VM, Martínez-Meñaca A, Iturbe-Fernández D, Tello-Mena S, Izquierdo-Cuervo S, García-Camarero T, Gil-Ongay A, Sánchez-Moreno L, Alonso-Lecue P, Naranjo-Gozalo S, Cifrián-Martínez JM. Impact of the New Definition of Pulmonary Hypertension on the Prevalence of Primary Graft Dysfunction in Lung Transplant Recipients. Heart Lung Circ 2024; 33:524-532. [PMID: 38429191 DOI: 10.1016/j.hlc.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 11/08/2023] [Accepted: 12/22/2023] [Indexed: 03/03/2024]
Abstract
BACKGROUND & AIM Pulmonary hypertension (PH) secondary to lung disease (Group-3 PH) is the second leading cause of PH. The role of PH as a risk factor for primary graft dysfunction (PGD) following lung transplant (LT) is controversial. OBJECTIVE To assess the impact that the new definition of PH had on the prevalence of PH in patients with advanced lung disease-candidate for LT, and its association with the occurrence of PGD. METHOD A retrospective study was performed in all patients undergoing cardiac catheterisation referred for consideration as candidates to LT in a centre between 1 January 2017 and 31 December 2022. The baseline and haemodynamic characteristics of patients were analysed, along with the occurrence of PGD and post-transplant course in those who ultimately underwent transplantation. RESULTS A total of 396 patients were included. Based on the new 2022 European Society of Cardiology/European Respiratory Society definitions, as many as 70.7% of patients met PH criteria. Since the introduction of the 2022 definition, a significant reduction was observed in the frequency of severe Group-3 PH (41.1% vs 10.3%; p<0.001), with respect to the 2015 definition. As many as 236 patients underwent transplantation. None of the variables associated with PH was identified as a risk factor for PGD. CONCLUSION The new classification did not have any impact on the prevalence of PGD after transplantation. These results exclude that any significant differences exist in the baseline characteristics or post-transplant course of patients with Group-3 PH vs unclassified PH.
Collapse
Affiliation(s)
- Víctor M Mora-Cuesta
- Respiratory Department, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain; Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain.
| | - Amaya Martínez-Meñaca
- Respiratory Department, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain; Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | - David Iturbe-Fernández
- Respiratory Department, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain; Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Sandra Tello-Mena
- Respiratory Department, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain; Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Sheila Izquierdo-Cuervo
- Respiratory Department, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain; Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | | | - Aritz Gil-Ongay
- Cardiology Department, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Laura Sánchez-Moreno
- Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Pilar Alonso-Lecue
- Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, Santander, Spain
| | - Sara Naranjo-Gozalo
- Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | - José M Cifrián-Martínez
- Respiratory Department, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain; Department of Thoracic Surgery, Lung Transplant Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| |
Collapse
|
6
|
Blanco I, Hernández-González F, García A, Torres-Castro R, Barberà JA. Management of Pulmonary Hypertension Associated with Chronic Lung Disease. Semin Respir Crit Care Med 2023; 44:826-839. [PMID: 37487524 DOI: 10.1055/s-0043-1770121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Pulmonary hypertension (PH) is a common complication of chronic lung diseases, particularly in chronic obstructive pulmonary disease (COPD) and interstitial lung diseases (ILD) and especially in advanced disease. It is associated with greater mortality and worse clinical course. Given the high prevalence of some respiratory disorders and because lung parenchymal abnormalities might be present in other PH groups, the appropriate diagnosis of PH associated with respiratory disease represents a clinical challenge. Patients with chronic lung disease presenting symptoms that exceed those expected by the pulmonary disease should be further evaluated by echocardiography. Confirmatory right heart catheterization is indicated in candidates to surgical treatments, suspected severe PH potentially amenable with targeted therapy, and, in general, in those conditions where the result of the hemodynamic assessment will determine treatment options. The treatment of choice for these patients who are hypoxemic is long-term oxygen therapy and pulmonary rehabilitation to improve symptoms. Lung transplant is the only curative therapy and can be considered in appropriate cases. Conventional vasodilators or drugs approved for pulmonary arterial hypertension (PAH) are not recommended in patients with mild-to-moderate PH because they may impair gas exchange and their lack of efficacy shown in randomized controlled trials. Patients with severe PH (as defined by pulmonary vascular resistance >5 Wood units) should be referred to a center with expertise in PH and lung diseases and ideally included in randomized controlled trials. Targeted PAH therapy might be considered in this subset of patients, with careful monitoring of gas exchange. In patients with ILD, inhaled treprostinil has been shown to improve functional ability and to delay clinical worsening.
Collapse
Affiliation(s)
- Isabel Blanco
- Department of Pulmonary Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic-University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- European Reference Network on Rare Pulmonary Diseases (ERN-LUNG), Spain
| | - Fernanda Hernández-González
- Department of Pulmonary Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic-University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- European Reference Network on Rare Pulmonary Diseases (ERN-LUNG), Spain
| | - Agustín García
- Department of Pulmonary Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic-University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- European Reference Network on Rare Pulmonary Diseases (ERN-LUNG), Spain
| | - Rodrigo Torres-Castro
- Department of Pulmonary Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic-University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- European Reference Network on Rare Pulmonary Diseases (ERN-LUNG), Spain
| | - Joan A Barberà
- Department of Pulmonary Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic-University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- European Reference Network on Rare Pulmonary Diseases (ERN-LUNG), Spain
| |
Collapse
|
7
|
Alqarni AA, Aldhahir AM, Alghamdi SA, Alqahtani JS, Siraj RA, Alwafi H, AlGarni AA, Majrshi MS, Alshehri SM, Pang L. Role of prostanoids, nitric oxide and endothelin pathways in pulmonary hypertension due to COPD. Front Med (Lausanne) 2023; 10:1275684. [PMID: 37881627 PMCID: PMC10597708 DOI: 10.3389/fmed.2023.1275684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Pulmonary hypertension (PH) due to chronic obstructive pulmonary disease (COPD) is classified as Group 3 PH, with no current proven targeted therapies. Studies suggest that cigarette smoke, the most risk factor for COPD can cause vascular remodelling and eventually PH as a result of dysfunction and proliferation of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs). In addition, hypoxia is a known driver of pulmonary vascular remodelling in COPD, and it is also thought that the presence of hypoxia in patients with COPD may further exaggerate cigarette smoke-induced vascular remodelling; however, the underlying cause is not fully understood. Three main pathways (prostanoids, nitric oxide and endothelin) are currently used as a therapeutic target for the treatment of patients with different groups of PH. However, drugs targeting these three pathways are not approved for patients with COPD-associated PH due to lack of evidence. Thus, this review aims to shed light on the role of impaired prostanoids, nitric oxide and endothelin pathways in cigarette smoke- and hypoxia-induced pulmonary vascular remodelling and also discusses the potential of using these pathways as therapeutic target for patients with PH secondary to COPD.
Collapse
Affiliation(s)
- Abdullah A. Alqarni
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Respiratory Therapy Unit, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Abdulelah M. Aldhahir
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Sara A. Alghamdi
- Respiratory Care Department, Al Murjan Hospital, Jeddah, Saudi Arabia
| | - Jaber S. Alqahtani
- Department of Respiratory Care, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Rayan A. Siraj
- Department of Respiratory Care, College of Applied Medical Sciences, King Faisal University, Al Ahsa, Saudi Arabia
| | - Hassan Alwafi
- Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Abdulkareem A. AlGarni
- King Abdulaziz Hospital, The Ministry of National Guard Health Affairs, Al Ahsa, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, College of Applied Medical Sciences, Al Ahsa, Saudi Arabia
| | - Mansour S. Majrshi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory Medicine, Royal Brompton Hospital, London, United Kingdom
| | - Saad M. Alshehri
- Department of Respiratory Therapy, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Linhua Pang
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, Nottingham, United Kingdom
| |
Collapse
|
8
|
Hung YC, Lee PF, Lin CF, Su YJ, Hsieh JW, Lin YJ, Ho CC, Chen YT. Associations between Smoking Status and Health-Related Physical Fitness and Balance Ability among Older Males in Taiwan. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1350. [PMID: 37512161 PMCID: PMC10386566 DOI: 10.3390/medicina59071350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/01/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
The primary aim of this study was to examine the relationships between smoking status and health-related physical fitness and balance ability in older males residing in Taiwan. This investigation adopted a cross-sectional design, utilizing data from 7688 older males who took part in the 2014-2015 wave of the National Physical Fitness Survey of Taiwan. Various data sources, including a standardized structured questionnaire, anthropometric measurements, health-related physical fitness assessments, and balance ability tests, were analyzed. The participants were divided into three categories based on their smoking habits: never smokers, former smokers, and current smokers. Multiple regression analyses were performed to evaluate the linear association between cigarette smoking status and health-related physical fitness and balance ability performance. Health-related physical fitness and balance performance were significantly greater (p < 0.05) in the never smoker group than in the current smoker group. Current smoking status was significantly negatively (p < 0.05) associated with cardiopulmonary function, muscular endurance, flexibility, and balance performance. A history of smoking (former smoker) was significantly negatively (p < 0.05) associated with the 2-min step test, 30-s arm curl and chair stand, as well as the 8-foot up-and-go test; however, the association was not significant for the back scratch, chair sit-and-reach, and one-leg stance with eyes open performance. These results suggest that current cigarette smoking is detrimental to health-related physical fitness and balance performance in older males. Quitting smoking may reverse the effects of smoking on overall body flexibility and static balance performance in Taiwanese older adults, thereby reducing the risk of falls and incapacity.
Collapse
Affiliation(s)
- Yi-Chuan Hung
- Department of Sport Management, National Taiwan University of Sport, Taichung City 404, Taiwan
- Sports Administration, Ministry of Education, Taipei City 104, Taiwan
| | - Po-Fu Lee
- Department of Leisure Industry and Health Promotion, National Ilan University, Yilan County 260, Taiwan
| | - Chi-Fang Lin
- Department of Physical Education and Sport Sciences, National Taiwan Normal University, Taipei City 106, Taiwan
| | - Yan-Jhu Su
- Department of Gerontology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Jenn-Woei Hsieh
- Department of Physical Education, Fu Jen Catholic University, New Taipei City 24205, Taiwan
- Office of Physical Education, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Yu-Ju Lin
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Chien-Chang Ho
- Department of Physical Education, Fu Jen Catholic University, New Taipei City 24205, Taiwan
- Research and Development Center for Physical Education, Health and Information Technology, College of Education, Fu Jen Catholic University, New Taipei City 24205, Taiwan
- Sports Medicine Center, Fu Jen Catholic Hospital, New Taipei City 243, Taiwan
| | - Yun-Tsung Chen
- Department of Health and Leisure Management, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| |
Collapse
|
9
|
Liu CW, Le HHT, Denaro P, Dai Z, Shao NY, Ong SG, Lee WH. E-cigarettes Induce Dysregulation of Autophagy Leading to Endothelial Dysfunction in Pulmonary Arterial Hypertension. Stem Cells 2023; 41:328-340. [PMID: 36640125 PMCID: PMC10128958 DOI: 10.1093/stmcls/sxad004] [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: 06/20/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Given the increasing popularity of electronic cigarettes (e-cigs), it is imperative to evaluate the potential health risks of e-cigs, especially in users with preexisting health concerns such as pulmonary arterial hypertension (PAH). The aim of the present study was to investigate whether differential susceptibility exists between healthy and patients with PAH to e-cig exposure and the molecular mechanisms contributing to it. Patient-specific induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) from healthy individuals and patients with PAH were used to investigate whether e-cig contributes to the pathophysiology of PAH and affects EC homeostasis in PAH. Our results showed that PAH iPSC-ECs showed a greater amount of damage than healthy iPSC-ECs upon e-cig exposure. Transcriptomic analyses revealed that differential expression of Akt3 may be responsible for increased autophagic flux impairment in PAH iPSC-ECs, which underlies increased susceptibility upon e-cig exposure. Moreover, knockdown of Akt3 in healthy iPSC-ECs significantly induced autophagic flux impairment and endothelial dysfunction, which further increased with e-cig treatment, thus mimicking the PAH cell phenotype after e-cig exposure. In addition, functional disruption of mTORC2 by knocking down Rictor in PAH iPSC-ECs caused autophagic flux impairment, which was mediated by downregulation of Akt3. Finally, pharmacological induction of autophagy via direct inhibition of mTORC1 and indirect activation of mTORC2 with rapamycin reverses e-cig-induced decreased Akt3 expression, endothelial dysfunction, autophagic flux impairment, and decreased cell viability, and migration in PAH iPSC-ECs. Taken together, these data suggest a potential link between autophagy and Akt3-mediated increased susceptibility to e-cig in PAH.
Collapse
Affiliation(s)
- Chen-wei Liu
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Hoai Huong Thi Le
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Philip Denaro
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Zhiyu Dai
- Translational Cardiovascular Research Center, University of Arizona College of Medicine, Phoenix, AZ, USA
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Ning-Yi Shao
- Health Sciences, University of Macau, Macau, People’s Republic of China
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, University of Arizona College of Medicine, Phoenix, AZ, USA
| |
Collapse
|
10
|
Hahad O, Kuntic M, Kuntic I, Daiber A, Münzel T. Tobacco smoking and vascular biology and function: evidence from human studies. Pflugers Arch 2023:10.1007/s00424-023-02805-z. [PMID: 36961561 DOI: 10.1007/s00424-023-02805-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/25/2023]
Abstract
Tobacco cigarette smoking is among the most complex and least understood health risk factors. A deeper insight into the pathophysiological actions of smoking exposure is of special importance as smoking is a major cause of chronic non-communicable diseases, in particular of cardiovascular disease as well as risk factors such as atherosclerosis and arterial hypertension. It is well known that smoking exerts its negative effects on cardiovascular health through various interdependent pathophysiological actions including hemodynamic and autonomic alterations, oxidative stress, inflammation, endothelial dysfunction, thrombosis, and hyperlipidemia. Importantly, impaired vascular endothelial function is acknowledged as an early key event in the initiation and progression of smoking-induced atherosclerosis. Increasing evidence from human studies indicates that cigarette smoke exposure associates with a pathological state of the vascular endothelium mainly characterized by reduced vascular nitric oxide bioavailability due to increased vascular superoxide production. In the present overview, we provide compact evidence on the effects of tobacco cigarette smoke exposure on vascular biology and function in humans centered on main drivers of adverse cardiovascular effects including endothelial dysfunction, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Omar Hahad
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany.
| | - Marin Kuntic
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Ivana Kuntic
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| |
Collapse
|
11
|
Gonzales J, Fraidenburg DR. Pharmacology and Emerging Therapies for Group 3 Pulmonary Hypertension Due to Chronic Lung Disease. Pharmaceuticals (Basel) 2023; 16:418. [PMID: 36986517 PMCID: PMC10058846 DOI: 10.3390/ph16030418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Pulmonary hypertension (PH) frequently complicates chronic lung disease and is associated with high morbidity and poor outcomes. Individuals with interstitial lung disease and chronic obstructive pulmonary disease develop PH due to structural changes associated with the destruction of lung parenchyma and vasculature with concurrent vasoconstriction and pulmonary vascular remodeling similar to what is observed in idiopathic pulmonary arterial hypertension (PAH). Treatment for PH due to chronic lung disease is largely supportive and therapies specific to PAH have had minimal success in this population with exception of the recently FDA-approved inhaled prostacyclin analogue treprostinil. Given the significant disease burden of PH due to chronic lung diseases and its associated mortality, a great need exists for improved understanding of molecular mechanisms leading to vascular remodeling in this population. This review will discuss the current understanding of pathophysiology and emerging therapeutic targets and potential pharmaceuticals.
Collapse
|
12
|
Gu S, Goel K, Forbes LM, Kheyfets VO, Yu YRA, Tuder RM, Stenmark KR. Tensions in Taxonomies: Current Understanding and Future Directions in the Pathobiologic Basis and Treatment of Group 1 and Group 3 Pulmonary Hypertension. Compr Physiol 2023; 13:4295-4319. [PMID: 36715285 PMCID: PMC10392122 DOI: 10.1002/cphy.c220010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In the over 100 years since the recognition of pulmonary hypertension (PH), immense progress and significant achievements have been made with regard to understanding the pathophysiology of the disease and its treatment. These advances have been mostly in idiopathic pulmonary arterial hypertension (IPAH), which was classified as Group 1 Pulmonary Hypertension (PH) at the Second World Symposia on PH in 1998. However, the pathobiology of PH due to chronic lung disease, classified as Group 3 PH, remains poorly understood and its treatments thus remain limited. We review the history of the classification of the five groups of PH and aim to provide a state-of-the-art review of the understanding of the pathogenesis of Group 1 PH and Group 3 PH including insights gained from novel high-throughput omics technologies that have revealed heterogeneities within these categories as well as similarities between them. Leveraging the substantial gains made in understanding the genomics, epigenomics, proteomics, and metabolomics of PAH to understand the full spectrum of the complex, heterogeneous disease of PH is needed. Multimodal omics data as well as supervised and unbiased machine learning approaches after careful consideration of the powerful advantages as well as of the limitations and pitfalls of these technologies could lead to earlier diagnosis, more precise risk stratification, better predictions of disease response, new sub-phenotype groupings within types of PH, and identification of shared pathways between PAH and other types of PH that could lead to new treatment targets. © 2023 American Physiological Society. Compr Physiol 13:4295-4319, 2023.
Collapse
Affiliation(s)
- Sue Gu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
- National Jewish Health, Denver, Colorodo, USA
| | - Khushboo Goel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- National Jewish Health, Denver, Colorodo, USA
| | - Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Vitaly O. Kheyfets
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
| | - Yen-rei A. Yu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
| | - Rubin M. Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
- Department of Pediatrics Section of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| |
Collapse
|
13
|
Swisher JW, Weaver E. The Evolving Management and Treatment Options for Patients with Pulmonary Hypertension: Current Evidence and Challenges. Vasc Health Risk Manag 2023; 19:103-126. [PMID: 36895278 PMCID: PMC9990521 DOI: 10.2147/vhrm.s321025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/01/2023] [Indexed: 03/06/2023] Open
Abstract
Pulmonary hypertension may develop as a disease process specific to pulmonary arteries with no identifiable cause or may occur in relation to other cardiopulmonary and systemic illnesses. The World Health Organization (WHO) classifies pulmonary hypertensive diseases on the basis of primary mechanisms causing increased pulmonary vascular resistance. Effective management of pulmonary hypertension begins with accurately diagnosing and classifying the disease in order to determine appropriate treatment. Pulmonary arterial hypertension (PAH) is a particularly challenging form of pulmonary hypertension as it involves a progressive, hyperproliferative arterial process that leads to right heart failure and death if untreated. Over the last two decades, our understanding of the pathobiology and genetics behind PAH has evolved and led to the development of several targeted disease modifiers that ameliorate hemodynamics and quality of life. Effective risk management strategies and more aggressive treatment protocols have also allowed better outcomes for patients with PAH. For those patients who experience progressive PAH with medical therapy, lung transplantation remains a life-saving option. More recent work has been directed at developing effective treatment strategies for other forms of pulmonary hypertension, such as chronic thromboembolic pulmonary hypertension (CTEPH) and pulmonary hypertension due to other lung or heart diseases. The discovery of new disease pathways and modifiers affecting the pulmonary circulation is an ongoing area of intense investigation.
Collapse
Affiliation(s)
- John W Swisher
- East Tennessee Pulmonary Hypertension Center, StatCare Pulmonary Consultants, Knoxville, TN, USA
| | - Eric Weaver
- East Tennessee Pulmonary Hypertension Center, StatCare Pulmonary Consultants, Knoxville, TN, USA
| |
Collapse
|
14
|
Qin X, Gao A, Hou X, Xu X, Chen L, Sun L, Hao Y, Shi Y. Connexins may play a critical role in cigarette smoke-induced pulmonary hypertension. Arch Toxicol 2022; 96:1609-1621. [PMID: 35344070 DOI: 10.1007/s00204-022-03274-6] [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: 02/17/2022] [Accepted: 03/02/2022] [Indexed: 11/02/2022]
Abstract
Pulmonary hypertension (PH) is a chronic progressive disease characterized by pulmonary vasoconstriction and remodeling. It causes a gradual increase in pulmonary vascular resistance leading to right-sided heart failure, and may be fatal. Chronic exposure to cigarette smoke (CS) is an essential risk factor for PH group 3; however, smoking continues to be prevalent and smoking cessation is reported to be difficult. A majority of smokers exhibit PH, which leads to a concomitant increase in the risk of mortality. The current treatments for PH group 3 focus on vasodilation and long-term oxygen supplementation, and fail to stop or reverse PH-associated continuous vascular remodeling. Recent studies have suggested that pulmonary vascular endothelial dysfunction induced by CS exposure may be an initial event in the natural history of PH, which in turn may be associated with abnormal alterations in connexin (Cx) expression. The relationship between Cx and CS-induced PH development has not yet been directly investigated. Therefore, this review will describe the roles of CS and Cx in the development of PH and discuss the related downstream pathways. We also discuss the possible role of Cx in CS-induced PH. It is hoped that this review may provide new perspectives for early intervention.
Collapse
Affiliation(s)
- Xiaojiang Qin
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
- China Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
| | - Anqi Gao
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xiaomin Hou
- Department of Pharmacology, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- China Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xinrong Xu
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Liangjin Chen
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Lin Sun
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Yuxuan Hao
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Yiwei Shi
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
15
|
Abstract
Pulmonary hypertension (PH) because of chronic lung disease is categorized as Group 3 PH in the most recent classification system. Prevalence of these diseases is increasing over time, creating a growing need for effective therapeutic options. Recent approval of the first pulmonary arterial hypertension therapy for the treatment of Group 3 PH related to interstitial lung disease represents an encouraging advancement. This review focuses on molecular mechanisms contributing to pulmonary vasculopathy in chronic hypoxia, the pathology and epidemiology of Group 3 PH, the right ventricular dysfunction observed in this population and clinical trial data that inform the use of pulmonary vasodilators in Group 3 PH.
Collapse
Affiliation(s)
- Navneet Singh
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI
| | - Peter Dorfmüller
- Department of Pathology, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Germany (P.D.).,German Center for Lung Research (DZL), Giessen, Germany (P.D.)
| | - Oksana A Shlobin
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA (O.A.S.)
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI.,Department of Health Services, Policy and Practice (C.E.V.), Brown University, Providence, RI
| |
Collapse
|
16
|
Smoking cessation for less than 10 years remains a risk factor of anastomotic leakage in mid-to-low rectal cancer patients undergoing sphincter-preserving surgery. Langenbecks Arch Surg 2022; 407:1131-1138. [DOI: 10.1007/s00423-021-02381-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
|
17
|
Czubaj-Kowal M, Nowicki GJ, Kurzawa R, Polak M, Ślusarska B. Factors Influencing the Concentration of Exhaled Nitric Oxide (FeNO) in School Children Aged 8–9-Years-Old in Krakow, with High FeNO Values ≥ 20 ppb. Medicina (B Aires) 2022; 58:medicina58020146. [PMID: 35208470 PMCID: PMC8877257 DOI: 10.3390/medicina58020146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Background and Objectives: Measurement of fractional exhaled nitric oxide (FeNO) concentration is currently used as a non-invasive biomarker to assess airway inflammation. Many factors can influence the FeNO level. However, there have been no reports concerning factors attributed to FeNO levels in different age groups of children, especially those with high FeNO values. Therefore, this study aimed to assess the influence of selected factors on nitric oxide concentration in exhaled air in children aged 8–9 attending class 3 of public primary schools in Krakow with high FeNO values ≥ 20 ppb. Materials and Methods: The population-based study covered all third-grade pupils attending primary schools in the city of Krakow. Five thousand, four hundred and sixty children participated in the first screening stage, conducted from October 2017 to January 2018. Then, 792 participants with an FeNO level ≥ 20 ppb were selected. Finally, those selected pupils were invited to participate in the second stage, diagnostic, in April 2018. Four hundred and fifty-four children completed the diagnostic stage of the study, and their data was included in the presented analysis. Results and Conclusions: Significantly higher FeNO levels were observed in children diagnosed with the following diseases: asthma, allergic rhinitis, atopic dermatitis, and allergy (p < 0.05). In addition, it was observed that a higher FeNO concentration characterised children taking antihistamines compared to children not taking those medications (p = 0.008). In multivariate models, we observed that regardless of sex, age, BMI value, home smoking, and whether they were taking medications, children who had allergic rhinitis, or atopic dermatitis, or allergies had significantly higher FeNO levels. The strongest relationship was noted with allergic diseases. The results of our study may be of importance to clinicians when interpreting FeNO results, for example, when making a therapeutic decision.
Collapse
Affiliation(s)
- Marta Czubaj-Kowal
- Department of Paediatrics, Stefan Zeromski Specialist Hospital in Krakow, Na Skarpie 66 Str., 31-913 Krakow, Poland
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzińskiego 1 Str., 30-705 Krakow, Poland
- Correspondence: ; Tel.: +48-604-433-42
| | - Grzegorz Józef Nowicki
- Department of Family and Geriatric Nursing, Medical University of Lublin, Staszica 6 Str., 20-081 Lublin, Poland; (G.J.N.); (B.Ś.)
| | - Ryszard Kurzawa
- Department of Alergology and Pneumonology, Institute of Tuberculosis and Lung Disorders, Prof. Jana Rudnika 3B Str., 34-700 Rabka-Zdrój, Poland;
| | - Maciej Polak
- Department of Epidemiology and Population Studies, Jagiellonian University Medical College, Grzegórzecka 20 Str., 31-531 Krakow, Poland;
| | - Barbara Ślusarska
- Department of Family and Geriatric Nursing, Medical University of Lublin, Staszica 6 Str., 20-081 Lublin, Poland; (G.J.N.); (B.Ś.)
| |
Collapse
|
18
|
Merianos AL, Jandarov RA, Cataletto M, Mahabee-Gittens EM. Tobacco smoke exposure and fractional exhaled nitric oxide levels among U.S. adolescents. Nitric Oxide 2021; 117:53-59. [PMID: 34688860 DOI: 10.1016/j.niox.2021.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/14/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Fractional exhaled nitric oxide (FeNO) can objectively guide clinical practice in the assessment, diagnosis, and treatment of eosinophilic airway inflammation. FeNO values may be affected by current smoking, but the role of tobacco smoke exposure (TSE) is understudied. OBJECTIVE This study investigated the associations between biochemically validated and self-reported TSE and FeNO levels among U.S. nonsmoking adolescents without asthma. METHODS National Health and Nutrition Examination Survey 2007-2012 data were used. TSE was assessed via serum cotinine and self-reported measures. We assessed FeNO continuously and using cutpoints of >35 ppb and >50 ppb to indicate likely eosinophilic inflammation in children and adults, respectively. We conducted linear and logistic regression adjusting for potential covariates. RESULTS Overall, 34.0% of adolescents had low cotinine (0.05-2.99 ng/ml), 6.2% had high cotinine (≥3.00 ng/ml), and 11.9% had home TSE. Compared to adolescents with no/minimal cotinine, adolescents with high cotinine were at reduced odds to have FeNO >35 ppb (adjusted odds ratio [aOR] = 0.54, 95%CI = 0.43,0.69). Adolescents with low cotinine had lower FeNO values (β = -2.05, 95%CI = -3.61,-0.49), and were also at decreased odds to have FeNO >35 ppb (aOR = 0.74, 95%CI = 0.66,0.83) and FeNO >50 ppb (aOR = 0.62, 95%CI = 0.53,0.72). Adolescents with home TSE were at reduced odds to have FeNO >50 ppb (aOR = 0.72, 95%CI = 0.57,0.91) than adolescents without home TSE. Adolescents with a higher number of cigarettes/day smoked inside their home were at reduced odds to have FeNO >35 ppb (OR = 0.98, 95%CI = 0.97,0.99) and FeNO >50 ppb (OR = 0.98, 95%CI = 0.96,0.99). CONCLUSIONS TSE was associated with decreased FeNO levels. The addition of TSE may be clinically important when interpreting thresholds for FeNO.
Collapse
Affiliation(s)
- Ashley L Merianos
- School of Human Services, University of Cincinnati, Cincinnati, OH, USA.
| | - Roman A Jandarov
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, 162 Kettering Lab Building, 160 Panzeca Way, Cincinnati, OH, 45267-0056, USA.
| | - Mary Cataletto
- Department of Pediatrics, NYU Long Island School of Medicine, 222 Station Plaza North, Mineola, NY, 11501, USA.
| | - E Melinda Mahabee-Gittens
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, 3333 Burnet Avenue, MLC 2008, Cincinnati, OH, 45229, USA.
| |
Collapse
|
19
|
Liao K, Lv DY, Yu HL, Chen H, Luo SX. iNOS regulates activation of the NLRP3 inflammasome through the sGC/cGMP/PKG/TACE/TNF-α axis in response to cigarette smoke resulting in aortic endothelial pyroptosis and vascular dysfunction. Int Immunopharmacol 2021; 101:108334. [PMID: 34768128 DOI: 10.1016/j.intimp.2021.108334] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cigarette smoke (CS) is associated with vascular injury and dysfunction, which may be mediated by iNOS and NLRP3. However, the exact mechanism is unknown. METHODS iNOS-knockout and NLRP3-knockout C57BL/6 mice were exposed to air or CS. The vascular structure was examined by hematoxylin-eosin staining. The vascular tension was measured by a vascular reactivity assay. The expression of iNOS, NLRP3, caspase-1p20, IL-1β and eNOS were measured by western blotting. Human aortic endothelial cells (HAECs) were exposed to L-NIL (iNOS inhibitor), MCC950 (NLRP3 inhibitor), ODQ (sGC inhibitor), KT5823 (PKG inhibitor) or TAPI-1 (TACE/ADAM17 inhibitor) for 1 h prior to cigarette smoke extract (CSE) treatment. The cell viability and lactate dehydrogenase activity were assessed and pyroptosis was determined by scanning electron microscopy. The mRNA expression of TNF-α, and protein expression of iNOS, active-TACE, NLRP3, caspase-1p20, IL-1β, and eNOS were measured. RESULTS CS resulted in shrinkage of endothelial cells, impaired aorta relaxation, reduced eNOS expression, and induced expression of iNOS, NLRP3, caspase-1p20 and IL-1β, which could be prevented by knockdown of iNOS and NLRP3. CSE reduced cell viability, induced LDH release and pyroptosis, and promoted iNOS, NLRP3, caspase-1p20, and IL-1β expression and reduced eNOS reduction, which could be reversed by inhibition of iNOS or NLRP3 in HAECs. Altogether, activation of the NLRP3 inflammasome by iNOS in CS-exposed HAECs may be mediated by the sGC/cGMP/PKG/TACE/TNF- α pathway. CONCLUSION These results link iNOS to NLRP3 in CSE-stimulated HAECs through the sGC/cGMP/PKG/TACE/TNF-α pathway. The findings identify a mechanism through which iNOS and NLRP3 contribute to the pathogenesis of CS-induced pyroptosis and impaired aorta relaxation in HAECs.
Collapse
Affiliation(s)
- Ke Liao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Ding-Yi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hui-Lin Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hong Chen
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| | - Su-Xin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| |
Collapse
|
20
|
Alvino VV, Thomas AC, Ghorbel MT, Rapetto F, Narayan SA, Kilcooley M, Iacobazzi D, Carrabba M, Fagnano M, Cathery W, Avolio E, Caputo M, Madeddu P. Reconstruction of the Swine Pulmonary Artery Using a Graft Engineered With Syngeneic Cardiac Pericytes. Front Bioeng Biotechnol 2021; 9:715717. [PMID: 34568300 PMCID: PMC8459923 DOI: 10.3389/fbioe.2021.715717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
The neonatal heart represents an attractive source of regenerative cells. Here, we report the results of a randomized, controlled, investigator-blinded preclinical study, which assessed the safety and effectiveness of a matrix graft cellularized with cardiac pericytes (CPs) in a piglet model of pulmonary artery (PA) reconstruction. Within each of five trios formed by 4-week-old female littermate piglets, one element (the donor) was sacrificed to provide a source of CPs, while the other two elements (the graft recipients) were allowed to reach the age of 10 weeks. During this time interval, culture-expanded donor CPs were seeded onto swine small intestinal submucosa (SIS) grafts, which were then shaped into conduits and conditioned in a flow bioreactor. Control unseeded SIS conduits were subjected to the same procedure. Then, recipient piglets were randomized to surgical reconstruction of the left PA (LPA) with unseeded or CP-seeded SIS conduits. Doppler echocardiography and cardiac magnetic resonance imaging (CMRI) were performed at baseline and 4-months post-implantation. Vascular explants were examined using histology and immunohistochemistry. All animals completed the scheduled follow-up. No group difference was observed in baseline imaging data. The final Doppler assessment showed that the LPA’s blood flow velocity was similar in the treatment groups. CMRI revealed a mismatch in the average growth of the grafted LPA and contralateral branch in both treatment groups. Histology of explanted arteries demonstrated that the CP-seeded grafts had a thicker luminal cell layer, more intraparietal arterioles, and a higher expression of endothelial nitric oxide synthase (eNOS) compared with unseeded grafts. Moreover, the LPA stump adjacent to the seeded graft contained more elastin and less collagen than the unseeded control. Syngeneic CP engineering did not accomplish the primary goal of supporting the graft’s growth but was able to improve secondary outcomes, such as the luminal cellularization and intraparietal vascularization of the graft, and elastic remodeling of the recipient artery. The beneficial properties of neonatal CPs may be considered in future bioengineering applications aiming to reproduce the cellular composition of native arteries.
Collapse
Affiliation(s)
- Valeria Vincenza Alvino
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Anita C Thomas
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Mohamed T Ghorbel
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Filippo Rapetto
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Srinivas A Narayan
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Michael Kilcooley
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Dominga Iacobazzi
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Michele Carrabba
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Marco Fagnano
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - William Cathery
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
21
|
Klinger JR. Treatment of Pulmonary Hypertension Associated With COPD: Time for Another Look? Chest 2021; 160:409-410. [PMID: 34366028 DOI: 10.1016/j.chest.2021.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- James R Klinger
- Divisions of Pulmonary, Sleep and Critical Care Medicine, Department of Medicine, and Hematology/Oncology, Department of Medicine, Rhode Island Hospital, and The Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
22
|
Al-Saeedi FJ, Al-Waheeb SK, Rajendran P, Khan KM, Sadan M. Early initiation of insulin attenuates histological and functional changes in the liver of streptozotocin-induced diabetic rats using 99mTc-sulfur colloid functional imaging. J Recept Signal Transduct Res 2021; 42:261-267. [PMID: 33853491 DOI: 10.1080/10799893.2021.1912097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This study aimed to investigate the effect of insulin on the reticuloendothelial system (RES) in the liver and spleen in diabetic rats. Sprague Dawley rats were divided into control, diabetic rats (DM) and diabetic rats treated with insulin (IDM) for 2 weeks. Rats were imaged with technetium-99m-sulfur colloid (99mTc-SC) tracer to determine regional distributions of the tracer for all groups by drawing regions of interest and then obtained the ratios as the cumulative counts of heart, liver, and spleen to the whole body (WB). Liver tissue from sacrificed rats from each group was examined by light and electron microscopy. 99mTc-SC uptake ratios showed a lower liver to WB uptake ratio in the DM rats compared to both controls and IDM rats. Electron microscopy showed severe vacuolization of the hepatocytes of DM rats. The IDM rats show complete resolution of the vacuolization. The early administration of insulin for 2 weeks to diabetic rats could significantly resolve the phagocytic RES function and histological changes in the liver.
Collapse
Affiliation(s)
- Fatma J Al-Saeedi
- Department of Nuclear Medicine, Faculty of Medicine, Kuwait University, Kuwait city, Kuwait
| | - Salah Kh Al-Waheeb
- Department of Pathology, Faculty of Medicine, Kuwait University, Kuwait city, Kuwait
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait city, Kuwait
| | - Moudhi Sadan
- Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
23
|
Karnati S, Seimetz M, Kleefeldt F, Sonawane A, Madhusudhan T, Bachhuka A, Kosanovic D, Weissmann N, Krüger K, Ergün S. Chronic Obstructive Pulmonary Disease and the Cardiovascular System: Vascular Repair and Regeneration as a Therapeutic Target. Front Cardiovasc Med 2021; 8:649512. [PMID: 33912600 PMCID: PMC8072123 DOI: 10.3389/fcvm.2021.649512] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality worldwide and encompasses chronic bronchitis and emphysema. It has been shown that vascular wall remodeling and pulmonary hypertension (PH) can occur not only in patients with COPD but also in smokers with normal lung function, suggesting a causal role for vascular alterations in the development of emphysema. Mechanistically, abnormalities in the vasculature, such as inflammation, endothelial dysfunction, imbalances in cellular apoptosis/proliferation, and increased oxidative/nitrosative stress promote development of PH, cor pulmonale, and most probably pulmonary emphysema. Hypoxemia in the pulmonary chamber modulates the activation of key transcription factors and signaling cascades, which propagates inflammation and infiltration of neutrophils, resulting in vascular remodeling. Endothelial progenitor cells have angiogenesis capabilities, resulting in transdifferentiation of the smooth muscle cells via aberrant activation of several cytokines, growth factors, and chemokines. The vascular endothelium influences the balance between vaso-constriction and -dilation in the heart. Targeting key players affecting the vasculature might help in the development of new treatment strategies for both PH and COPD. The present review aims to summarize current knowledge about vascular alterations and production of reactive oxygen species in COPD. The present review emphasizes on the importance of the vasculature for the usually parenchyma-focused view of the pathobiology of COPD.
Collapse
Affiliation(s)
- Srikanth Karnati
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Thati Madhusudhan
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Akash Bachhuka
- UniSA Science, Technology, Engineering and Mathematics, University of South Australia, Mawson Lakes Campus, Adelaide, SA, Australia
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, University of Giessen, Giessen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Sadaka AS, Faisal A, Khalil YM, Mourad SM, Zidan MH, Polkey MI, Hopkinson NS. Reduced skeletal muscle endurance and ventilatory efficiency during exercise in adult smokers without airflow obstruction. J Appl Physiol (1985) 2021; 130:976-986. [DOI: 10.1152/japplphysiol.00762.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In adult smokers without airflow obstruction, the contributions of pulmonary and skeletal muscle functions to reduced exercise capacity are unclear. We found that non-COPD smokers had decreased exercise capacity and muscle endurance although strength was preserved compared with never-smoking controls. Exercise endurance was associated with quadriceps endurance and CO transfer factor. Despite similar physical activity levels, smokers developed leg fatigue, breathlessness, and displayed increased ventilation with reduced ventilatory efficiency at lower workloads, without exhibiting ventilatory constraint.
Collapse
Affiliation(s)
- Ahmed S. Sadaka
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Azmy Faisal
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Faculty of Physical Education for Men, Alexandria University, Alexandria, Egypt
| | - Yehia M. Khalil
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Sahar M. Mourad
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed H. Zidan
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Michael I. Polkey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory Medicine Department, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | | |
Collapse
|
25
|
Lázár Z, Mészáros M, Bikov A. The Nitric Oxide Pathway in Pulmonary Arterial Hypertension: Pathomechanism, Biomarkers and Drug Targets. Curr Med Chem 2021; 27:7168-7188. [PMID: 32442078 DOI: 10.2174/0929867327666200522215047] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/03/2020] [Accepted: 02/20/2020] [Indexed: 11/22/2022]
Abstract
The altered Nitric Oxide (NO) pathway in the pulmonary endothelium leads to increased vascular smooth muscle tone and vascular remodelling, and thus contributes to the development and progression of pulmonary arterial hypertension (PAH). The pulmonary NO signalling is abrogated by the decreased expression and dysfunction of the endothelial NO synthase (eNOS) and the accumulation of factors blocking eNOS functionality. The NO deficiency of the pulmonary vasculature can be assessed by detecting nitric oxide in the exhaled breath or measuring the degradation products of NO (nitrite, nitrate, S-nitrosothiol) in blood or urine. These non-invasive biomarkers might show the potential to correlate with changes in pulmonary haemodynamics and predict response to therapies. Current pharmacological therapies aim to stimulate pulmonary NO signalling by suppressing the degradation of NO (phosphodiesterase- 5 inhibitors) or increasing the formation of the endothelial cyclic guanosine monophosphate, which mediates the downstream effects of the pathway (soluble guanylate cyclase sensitizers). Recent data support that nitrite compounds and dietary supplements rich in nitrate might increase pulmonary NO availability and lessen vascular resistance. This review summarizes current knowledge on the involvement of the NO pathway in the pathomechanism of PAH, explores novel and easy-to-detect biomarkers of the pulmonary NO.
Collapse
Affiliation(s)
- Zsófia Lázár
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Martina Mészáros
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Andras Bikov
- Department of Pulmonology, Semmelweis University, Budapest, Hungary,Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
26
|
Kurakula K, Smolders VFED, Tura-Ceide O, Jukema JW, Quax PHA, Goumans MJ. Endothelial Dysfunction in Pulmonary Hypertension: Cause or Consequence? Biomedicines 2021; 9:biomedicines9010057. [PMID: 33435311 PMCID: PMC7827874 DOI: 10.3390/biomedicines9010057] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, complex, and progressive disease that is characterized by the abnormal remodeling of the pulmonary arteries that leads to right ventricular failure and death. Although our understanding of the causes for abnormal vascular remodeling in PAH is limited, accumulating evidence indicates that endothelial cell (EC) dysfunction is one of the first triggers initiating this process. EC dysfunction leads to the activation of several cellular signalling pathways in the endothelium, resulting in the uncontrolled proliferation of ECs, pulmonary artery smooth muscle cells, and fibroblasts, and eventually leads to vascular remodelling and the occlusion of the pulmonary blood vessels. Other factors that are related to EC dysfunction in PAH are an increase in endothelial to mesenchymal transition, inflammation, apoptosis, and thrombus formation. In this review, we outline the latest advances on the role of EC dysfunction in PAH and other forms of pulmonary hypertension. We also elaborate on the molecular signals that orchestrate EC dysfunction in PAH. Understanding the role and mechanisms of EC dysfunction will unravel the therapeutic potential of targeting this process in PAH.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Cell and Chemical Biology, Laboratory for CardioVascular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Valérie F. E. D. Smolders
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.F.E.D.S.); (P.H.A.Q.)
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain;
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institut (IDIBGI), 17190 Girona, Catalonia, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Paul H. A. Quax
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.F.E.D.S.); (P.H.A.Q.)
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Laboratory for CardioVascular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Correspondence:
| |
Collapse
|
27
|
Ren Z, Li J, Shen J, Yu H, Mei X, Zhao P, Xiao Z, Wu W. Therapeutic sildenafil inhibits pulmonary damage induced by cigarette smoke exposure and bacterial inhalation in rats. PHARMACEUTICAL BIOLOGY 2020; 58:116-123. [PMID: 31967915 PMCID: PMC7006811 DOI: 10.1080/13880209.2019.1711135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 12/19/2019] [Accepted: 12/28/2019] [Indexed: 06/10/2023]
Abstract
Context: Clinical reports showed sildenafil beneficial therapy on severe chronic obstructive pulmonary disease (COPD) with pulmonary hypertension (PH) patients.Objective: The study investigated therapeutic effects of silenafil on pulmonary damage induced by cigarette smoke exposure and bacterial inhalation in rats.Materials and methods: Female Sprague-Dawley rats (200-250 g) were divided into control group (no exposure, n = 10) and exposure group (n = 50) suffered from cigarette smoke exposure and Klebsiella pneumonia inhalation for 8 weeks. Then rats were orally given normal saline (control group or model group), 2.0, 3.0, or 4.5 mg/kg sildenafil for 4 weeks, respectively. Pulmonary pressure, RVHI and morphological analysis of pulmonary vascular remodeling, respiratory functions assay, morphological analysis of pulmonary alveoli, and expression of PCNA and caspase-3 of epithelial cells in bronchioles wall were examined.Results: Compared to model rats, 2.0, 3.0, and 4.5 mg/kg sildenafil increased VT by -0.6 to 9.58%, PEF by 3.12 to 6.49%, EF50 by 0.81 to 6.50%, decreased mPAP by 4.43 to 25.58%, RVHI by 6.54 to 26.41%, showing a dose-dependent improvement. Furthermore, 4.5 mg/kg sildenafil significantly increased MAN by 39.70%, LA/CSA by 37.07%, decreased muscular pulmonary arteries by 48.00%, WT by 12.83%, MT by 22.89%, caspase-3 expression by 17.71%, and showed improvement on abnormality in lung interstitial and bronchioles by microscopy.Discussion and conclusion: Our results demonstrated that sildenafil decreased pathological changes in alveoli, bronchioles, interstitial tissue, and arterioles of rats with COPD and PH.
Collapse
Affiliation(s)
- Zhouxin Ren
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P. R. China, Zhengzhou, China
| | - Jiansheng Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P. R. China, Zhengzhou, China
| | - Junling Shen
- First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Haibin Yu
- First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaofeng Mei
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P. R. China, Zhengzhou, China
| | - Zhenya Xiao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wanliu Wu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
28
|
Gopallawa I, Lee RJ. Targeting the phosphoinositide-3-kinase/protein kinase B pathway in airway innate immunity. World J Biol Chem 2020; 11:30-51. [PMID: 33024516 PMCID: PMC7520643 DOI: 10.4331/wjbc.v11.i2.30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The airway innate immune system maintains the first line of defense against respiratory infections. The airway epithelium and associated immune cells protect the respiratory system from inhaled foreign organisms. These cells sense pathogens via activation of receptors like toll-like receptors and taste family 2 receptors (T2Rs) and respond by producing antimicrobials, inflammatory cytokines, and chemokines. Coordinated regulation of fluid secretion and ciliary beating facilitates clearance of pathogens via mucociliary transport. Airway cells also secrete antimicrobial peptides and radicals to directly kill microorganisms and inactivate viruses. The phosphoinositide-3-kinase/protein kinase B (Akt) kinase pathway regulates multiple cellular targets that modulate cell survival and proliferation. Akt also regulates proteins involved in innate immune pathways. Akt phosphorylates endothelial nitric oxide synthase (eNOS) enzymes expressed in airway epithelial cells. Activation of eNOS can have anti-inflammatory, anti-bacterial, and anti-viral roles. Moreover, Akt can increase the activity of the transcription factor nuclear factor erythroid 2 related factor-2 that protects cells from oxidative stress and may limit inflammation. In this review, we summarize the recent findings of non-cancerous functions of Akt signaling in airway innate host defense mechanisms, including an overview of several known downstream targets of Akt involved in innate immunity.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Robert J Lee
- Department of Otorhinolaryngology and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
29
|
Efficacy and safety of Sildenafil treatment in pulmonary hypertension caused by chronic obstructive pulmonary disease: A meta-analysis. Life Sci 2020; 257:118001. [PMID: 32634428 DOI: 10.1016/j.lfs.2020.118001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/29/2022]
Abstract
AIMS Pulmonary hypertension (PH) is a severe and prevalent complication of chronic obstructive pulmonary disease (COPD), with low quality of life and poor prognosis. This study was designed to evaluate the efficacy and safety of Sildenafil in the treatment of PH caused by COPD (COPD-PH) and provide reference for clinical treatment. MATERIALS AND METHODS We systematically searched PubMed, EMBASE, Cochrane Library, Clinical Trials.gov databases, Wanfang Data and CNKI for comprehensive literature reporting Sildenafil for randomized controlled trials (RCT) of COPD-PH. Quality assessment, data analysis used the modified Jadad scale and RevMan5.3 software. KEY FINDINGS A total of 9 RCTs involving 579 patients were included in our study. The primary outcome measure was Six minutes walking distance (6MWD). Secondary observations were Pulmonary artery systolic pressure (PASP), Borg dyspnea index, and Survey scale (SF-36). Our data demonstrate that Sildenafil can improve 6WMD [29.64, 95% CI (13.78, 45.50), P < 0.00001] and PASP [-7.86, 95% CI (-11.26, -4.46) P < 0.00001] of COPD-PH, compared with the control group. However, SF-36 [2.64, 95% CI (-6.85, 12.14) P = 0.59] and Borg dyspnea index [-0.28, 95% CI (-1.08, 0.52) P = 0.49] have no significant difference between those two groups. Adverse reactions in the Sildenafil treatment group were tolerated headaches and digestive symptoms, which were relatively safe. SIGNIFICANCE Available clinical evidence indicates that Sildenafil seems to be safe and effective for COPD-PH and can improve the patients' 6WMD. However, large-sample, high-quality multicenter RCTs are still needed to provide stronger evidence-based medical evidence.
Collapse
|
30
|
Costa ED, Silva JF, Garcia DC, Wainstein AJ, Rezende BA, Tostes RC, Teixeira MM, Cortes SF, Lemos VS. Decreased expression of neuronal nitric oxide synthase contributes to the endothelial dysfunction associated with cigarette smoking in human. Nitric Oxide 2020; 98:20-28. [DOI: 10.1016/j.niox.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 12/27/2022]
|
31
|
McGettrick M, Peacock A. Group 3 pulmonary hypertension: Challenges and opportunities. Glob Cardiol Sci Pract 2020; 2020:e202006. [PMID: 33150151 PMCID: PMC7590933 DOI: 10.21542/gcsp.2020.6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Michael McGettrick
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, UK
| | - Andrew Peacock
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, UK
| |
Collapse
|
32
|
Peacock AJ, Ling Y, Johnson MK, Kiely DG, Condliffe R, Elliot CA, Gibbs JSR, Howard LS, Pepke-Zaba J, Sheares KKK, Corris PA, Fisher AJ, Lordan JL, Gaine S, Coghlan JG, Wort SJ, Gatzoulis MA. Idiopathic pulmonary arterial hypertension and co-existing lung disease: is this a new phenotype? Pulm Circ 2020; 10:2045894020914851. [PMID: 32284847 PMCID: PMC7132795 DOI: 10.1177/2045894020914851] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/23/2020] [Indexed: 11/26/2022] Open
Abstract
Patients classified as idiopathic pulmonary arterial hypertension (defined as Group 1 on European Respiratory Society (ERS)/European Cardiac Society (ESC) criteria) may have evidence of minor co-existing lung disease on thoracic computed tomography. We hypothesised that these idiopathic pulmonary arterial hypertension patients (IPAH lung disease) are a separate subgroup of idiopathic pulmonary arterial hypertension with different phenotype and outcome compared with idiopathic pulmonary arterial hypertension patients without co-existing lung disease (IPAH no lung disease). Patients with ‘IPAH lung disease’ have been eligible for all clinical trials of Group 1 patients because they have normal clinical examination and normal spirometry but we wondered whether they responded to treatment and had similar survival to patients with ‘IPAH no lung disease’. We described the outcome of the cohort of patients with ‘IPAH no lung disease’ in a previous paper. Here, we have compared incident ‘IPAH lung disease’ patients with ‘IPAH no lung disease’ patients diagnosed concurrently in all eight Pulmonary Hypertension centres in the UK and Ireland between 2001–2009. Compared with ‘IPAH no lung disease’ (n = 355), ‘IPAH lung disease’ patients (n = 137) were older, less obese, predominantly male, more likely to be current/ex-smokers and had lower six-minute walk distance, lower % predicted diffusion capacity for carbon monoxide, lower mean pulmonary arterial pressure and lower pulmonary vascular resistance index. After three months of pulmonary hypertension-targeted treatment, six-minute walk distance improved equally in ‘IPAH lung disease’ and ‘IPAH no lung disease’. However, survival of ‘IPAH lung disease’ was lower than ‘IPAH no lung disease’ (one year survival: 72% compared with 93%). This survival was significantly worse in ‘IPAH lung disease’ even after adjusting for age, gender, smoking history, comorbidities and haemodynamics. ‘IPAH lung disease’ patients had similar short-term improvement in six-minute walk distance with anti-pulmonary arterial hypertension therapy but worse survival compared with ‘IPAH no lung disease’ patients. This suggests that ‘IPAH lung disease’ are a separate phenotype and should not be lumped with ‘IPAH no lung disease’ in clinical trials of Group 1 pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Andrew J Peacock
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, UK
| | - Yi Ling
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, UK
| | - Martin K Johnson
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Glasgow, UK
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Charlie A Elliot
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - J Simon R Gibbs
- National Heart and Lung Institute, Imperial College London and Hammersmith Hospital, London, UK
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London and Hammersmith Hospital, London, UK
| | | | | | - Paul A Corris
- Northern Pulmonary Vascular Unit, Freeman Hospital, Newcastle, UK
| | - Andrew J Fisher
- Northern Pulmonary Vascular Unit, Freeman Hospital, Newcastle, UK
| | - James L Lordan
- Northern Pulmonary Vascular Unit, Freeman Hospital, Newcastle, UK
| | - Sean Gaine
- National Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | - J Gerry Coghlan
- Pulmonary Hypertension Unit, Royal Free Hospital, London, UK
| | - S John Wort
- Royal Brompton Pulmonary Hypertension and Adult Congenital Heart Centre, London, UK
| | - Michael A Gatzoulis
- Royal Brompton Pulmonary Hypertension and Adult Congenital Heart Centre, London, UK
| |
Collapse
|
33
|
Gawrys J, Gajecki D, Szahidewicz-Krupska E, Doroszko A. Intraplatelet L-Arginine-Nitric Oxide Metabolic Pathway: From Discovery to Clinical Implications in Prevention and Treatment of Cardiovascular Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1015908. [PMID: 32215167 PMCID: PMC7073508 DOI: 10.1155/2020/1015908] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/12/2020] [Indexed: 12/31/2022]
Abstract
Despite the development of new drugs and other therapeutic strategies, cardiovascular disease (CVD) remains still the major cause of morbidity and mortality in the world population. A lot of research, performed mostly in the last three decades, revealed an important correlation between "classical" demographic and biochemical risk factors for CVD, (i.e., hypercholesterolemia, hyperhomocysteinemia, smoking, renal failure, aging, diabetes, and hypertension) with endothelial dysfunction associated directly with the nitric oxide deficiency. The discovery of nitric oxide and its recognition as an endothelial-derived relaxing factor was a breakthrough in understanding the pathophysiology and development of cardiovascular system disorders. The nitric oxide synthesis pathway and its regulation and association with cardiovascular risk factors were a common subject for research during the last decades. As nitric oxide synthase, especially its endothelial isoform, which plays a crucial role in the regulation of NO bioavailability, inhibiting its function results in the increase in the cardiovascular risk pattern. Among agents altering the production of nitric oxide, asymmetric dimethylarginine-the competitive inhibitor of NOS-appears to be the most important. In this review paper, we summarize the role of L-arginine-nitric oxide pathway in cardiovascular disorders with the focus on intraplatelet metabolism.
Collapse
Affiliation(s)
- Jakub Gawrys
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| | - Damian Gajecki
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| | - Ewa Szahidewicz-Krupska
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| | - Adrian Doroszko
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| |
Collapse
|
34
|
Impact de l’exposition à la fumée de cigarette sur la réponse hémodynamique à l’exercice de patients sous traitement spécifique de l’HTAP. Rev Mal Respir 2020; 37:222-234. [DOI: 10.1016/j.rmr.2019.11.651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/29/2019] [Indexed: 11/23/2022]
|
35
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a common and progressive disorder
which is characterised by pathological abnormalities driven by chronic airway inflammation. The
assessment of airway inflammation in routine clinical practice in COPD is limited to surrogate blood
markers. Fractional exhaled nitric oxide (FENO) is a marker of eosinophilic airway inflammation in
asthma, and it can predict steroid responsiveness and help tailor corticosteroid treatment. The clinical
value of FENO in COPD is less evident, but some studies suggest that it may be a marker of the
eosinophilic endotype. More importantly, mathematical methods allow investigation of the
alveolar/small airway production of NO which potentially better reflects inflammatory changes in
anatomical sites, most affected by COPD. This review summarises the pathophysiological role of
nitric oxide in COPD, explains the methodology of its measurement in exhaled air and discusses
clinical findings of FENO in COPD.
Collapse
Affiliation(s)
- Andras Bikov
- NIHR Manchester Clinical Research Facility, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Martina Meszaros
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Zsofia Lazar
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
36
|
Kovacs G, Agusti A, Barberà JA, Celli B, Criner G, Humbert M, Sin DD, Voelkel N, Olschewski H. Pulmonary Vascular Involvement in Chronic Obstructive Pulmonary Disease. Is There a Pulmonary Vascular Phenotype? Am J Respir Crit Care Med 2019; 198:1000-1011. [PMID: 29746142 DOI: 10.1164/rccm.201801-0095pp] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Gabor Kovacs
- 1 Medical University of Graz, Graz, Austria.,2 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Alvar Agusti
- 3 Respiratory Institute, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain.,4 Centro Investigacion Biomedica en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Joan Albert Barberà
- 3 Respiratory Institute, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain.,4 Centro Investigacion Biomedica en Red de Enfermedades Respiratorias, Madrid, Spain
| | | | - Gerard Criner
- 6 Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Marc Humbert
- 7 Université Paris-Sud, Université Paris-Saclay; Inserm U999; Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Don D Sin
- 8 Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada.,9 Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia; Canada; and
| | - Norbert Voelkel
- 10 Department of Pulmonary Medicine, Frije University, Medical Center, Amsterdam, the Netherlands
| | - Horst Olschewski
- 1 Medical University of Graz, Graz, Austria.,2 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| |
Collapse
|
37
|
Coste F, Benlala I, Dournes G, Girodet PO, Laurent F, Berger P. Assessing pulmonary hypertension in COPD. Is there a role for computed tomography? Int J Chron Obstruct Pulmon Dis 2019; 14:2065-2079. [PMID: 31564854 PMCID: PMC6732516 DOI: 10.2147/copd.s207363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/10/2019] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a common complication of chronic obstructive pulmonary disease (COPD) and is associated with increased morbidity and mortality. Reference standard method to diagnose PH is right heart catheterization. Several non-invasive imaging techniques have been employed in the detection of PH. Among them, computed tomography (CT) is the most commonly used for phenotyping and detecting complications of COPD. Several CT findings have also been described in patients with severe PH. Nevertheless, CT analysis is currently based on visual findings which can lead to reproducibility failure. Therefore, there is a need for quantification in order to assess objective criteria. In this review, progresses in automated analyses of CT parameters and their values in predicting PH and COPD outcomes are presented.
Collapse
Affiliation(s)
- Florence Coste
- University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000 France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC1401, Bordeaux, F-33000 France
| | - Ilyes Benlala
- University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000 France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC1401, Bordeaux, F-33000 France.,CHU de Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, CIC1401, Service d'Explorations Fonctionnelles Respiratoires, Pessac, F-33600 France
| | - Gaël Dournes
- University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000 France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC1401, Bordeaux, F-33000 France.,CHU de Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, CIC1401, Service d'Explorations Fonctionnelles Respiratoires, Pessac, F-33600 France
| | - Pierre-Olivier Girodet
- University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000 France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC1401, Bordeaux, F-33000 France.,CHU de Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, CIC1401, Service d'Explorations Fonctionnelles Respiratoires, Pessac, F-33600 France
| | - François Laurent
- University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000 France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC1401, Bordeaux, F-33000 France.,CHU de Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, CIC1401, Service d'Explorations Fonctionnelles Respiratoires, Pessac, F-33600 France
| | - Patrick Berger
- University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000 France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC1401, Bordeaux, F-33000 France.,CHU de Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, CIC1401, Service d'Explorations Fonctionnelles Respiratoires, Pessac, F-33600 France
| |
Collapse
|
38
|
Chen S, Wu H, Yang T, Li B, Hu Y, Sun H. Does Early Graft Patency Benefit from Perioperative Statin Therapy? A Propensity Score-Matched Study of Patients Undergoing Off-Pump Coronary Artery Bypass Surgery. Cardiovasc Ther 2019; 2019:1582183. [PMID: 31772605 PMCID: PMC6739783 DOI: 10.1155/2019/1582183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/01/2019] [Accepted: 07/21/2019] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Decreased graft patency after off-pump coronary artery bypass grafting (OPCAB) leads to substantial increases in cardiac events. However, there is paucity of data on efficacy and safety of perioperative statin therapy for OPCAB populations. METHODS 582 patients undergoing OPCAB in a single-institution database (October 1, 2009-September 30, 2012) were stratified by perioperative continuation of statin therapy (CS group, n=398) or not (DS group, n=184). Inverse probability weighted propensity adjustment was used to account for treatment assignment bias, resulting in a well-matched cohort. Primary outcomes were graft patency at an average of five days after operation and in-hospital mortality. Secondary outcomes included intraoperative blood loss, liver, and renal functions. RESULTS No in-hospital death occurred in this study. Early graft patency rates after OPCAB were 98.4% (1255 of 1275 grafts) in the CS group and 98.0% (583 of 595 grafts, P=0.486) in the DS group. Secondary outcomes showed a reduction in blood loss during operation (438.53 mL versus 480.47 mL, P=0.01). Continuation of statin therapy is associated with alanine transaminase (ALT) elevation (49.67 U/L versus 34.52 U/L, P<0.001), as well as aspartate transaminase (33.54 U/L versus 28.10 U/L, P<0.001). Abnormal ALT elevation was observed in 8.9% of the CS group and 3.1% in DS (odds ratio 3.06, 95% confidence interval, 1.77 to 5.29, P<0.001). There was no significant difference in estimated glomerular filtration rate (76.28 mL/min/1.73m2 versus 76.13 mL/min/1.73m2, P=0.90). Subgroup analyses suggested that graft occlusion was less common in CS than in DS group among smoking patients (odds ratio 0.41, 95% confidence interval, 0.20 to 0.86, P=0.026). CONCLUSIONS Perioperative continuation of statin therapy did not improve early graft patency in OPCAB patients. A lower risk of graft occlusion was observed among smoking patients. Continuous statin use correlated with liver function elevation (Clinical Trials.gov number, NCT01268917).
Collapse
Affiliation(s)
- Shanglin Chen
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hengchao Wu
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Tao Yang
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Baotong Li
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yuanyu Hu
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hansong Sun
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
39
|
Paul T, Blanco I, Aguilar D, Tura-Ceide O, Bonjoch C, Smolders VF, Peinado VI, Barberà JA. Therapeutic effects of soluble guanylate cyclase stimulation on pulmonary hemodynamics and emphysema development in guinea pigs chronically exposed to cigarette smoke. Am J Physiol Lung Cell Mol Physiol 2019; 317:L222-L234. [PMID: 31166128 DOI: 10.1152/ajplung.00399.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have analyzed the effect of the soluble guanylate cyclase (sGC) stimulator BAY 41-2272 in a therapeutic intervention in guinea pigs chronically exposed to cigarette smoke (CS). The effects of sGC stimulation on respiratory function, pulmonary hemodynamics, airspace size, vessel remodeling, and inflammatory cell recruitment to the lungs were evaluated in animals that had been exposed to CS for 3 mo. CS exposure was continued for an additional 3 mo in half of the animals and withdrawn in the other half. Animals that stopped CS exposure had slightly lower pulmonary artery pressure (PAP) and right ventricle (RV) hypertrophy than those who continued CS exposure, but they did not recover from the emphysema and the inflammatory cell infiltrate. Conversely, oral BAY 41-2272 administration stopped progression or even reversed the CS-induced emphysema in both current and former smokers, respectively. Furthermore, BAY 41-2272 produced a reduction in the RV hypertrophy, which correlated with a decrease in the PAP values. By contrast, the degree of vessel remodeling induced by CS remained unchanged in the treated animals. Functional network analysis suggested perforin/granzyme pathway downregulation as an action mechanism capable of stopping the progression of emphysema after sGC stimulation. The pathway analysis also showed normalization of the expression of cGMP-dependent serine/kinases. In conclusion, in guinea pigs chronically exposed to CS, sGC stimulation exerts beneficial effects on the lung parenchyma and the pulmonary vasculature, suggesting that sGC stimulators might be a potential alternative for chronic obstructive pulmonary disease treatment that deserves further evaluation.
Collapse
Affiliation(s)
- Tanja Paul
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Madrid, Spain
| | - Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Madrid, Spain
| | - Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases, Barcelona, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Madrid, Spain
| | - Cristina Bonjoch
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Valérie F Smolders
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- Department of Biochemistry and Molecular Biology and Institute of Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Victor I Peinado
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Madrid, Spain
| | - Joan A Barberà
- Department of Pulmonary Medicine, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Madrid, Spain
| |
Collapse
|
40
|
Abstract
Therapeutics for arachidonic acid pathways began with the development of non-steroidal anti-inflammatory drugs that inhibit cyclooxygenase (COX). The enzymatic pathways and arachidonic acid metabolites and respective receptors have been successfully targeted and therapeutics developed for pain, inflammation, pulmonary and cardiovascular diseases. These drugs target the COX and lipoxygenase pathways but not the third branch for arachidonic acid metabolism, the cytochrome P450 (CYP) pathway. Small molecule compounds targeting enzymes and CYP epoxy-fatty acid metabolites have evolved rapidly over the last two decades. These therapeutics have primarily focused on inhibiting soluble epoxide hydrolase (sEH) or agonist mimetics for epoxyeicosatrienoic acids (EET). Based on preclinical animal model studies and human studies, major therapeutic indications for these sEH inhibitors and EET mimics/analogs are renal and cardiovascular diseases. Novel small molecules that inhibit sEH have advanced to human clinical trials and demonstrate promise for cardiovascular diseases. Challenges remain for sEH inhibitor and EET analog drug development; however, there is a high likelihood that a drug that acts on this third branch of arachidonic acid metabolism will be utilized to treat a cardiovascular or kidney disease in the next decade.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
41
|
Zhang B, Paffett ML, Naik JS, Jernigan NL, Walker BR, Resta TC. Cholesterol Regulation of Pulmonary Endothelial Calcium Homeostasis. CURRENT TOPICS IN MEMBRANES 2018; 82:53-91. [PMID: 30360783 DOI: 10.1016/bs.ctm.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cholesterol is a key structural component and regulator of lipid raft signaling platforms critical for cell function. Such regulation may involve changes in the biophysical properties of lipid microdomains or direct protein-sterol interactions that alter the function of ion channels, receptors, enzymes, and membrane structural proteins. Recent studies have implicated abnormal membrane cholesterol levels in mediating endothelial dysfunction that is characteristic of pulmonary hypertensive disorders, including that resulting from long-term exposure to hypoxia. Endothelial dysfunction in this setting is characterized by impaired pulmonary endothelial calcium entry and an associated imbalance that favors production vasoconstrictor and mitogenic factors that contribute to pulmonary hypertension. Here we review current knowledge of cholesterol regulation of pulmonary endothelial Ca2+ homeostasis, focusing on the role of membrane cholesterol in mediating agonist-induced Ca2+ entry and its components in the normal and hypertensive pulmonary circulation.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Michael L Paffett
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States.
| |
Collapse
|
42
|
García-Lucio J, Peinado VI, de Jover L, del Pozo R, Blanco I, Bonjoch C, Coll-Bonfill N, Paul T, Tura-Ceide O, Barberà JA. Imbalance between endothelial damage and repair capacity in chronic obstructive pulmonary disease. PLoS One 2018; 13:e0195724. [PMID: 29672621 PMCID: PMC5908268 DOI: 10.1371/journal.pone.0195724] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/28/2018] [Indexed: 02/07/2023] Open
Abstract
Background Circulating endothelial microparticles (EMPs) and progenitor cells (PCs) are biological markers of endothelial function and endogenous repair capacity. The study was aimed to investigate whether COPD patients have an imbalance between EMPs to PCs compared to controls and to evaluate the effect of cigarette smoke on these circulating markers. Methods Circulating EMPs and PCs were determined by flow cytometry in 27 nonsmokers, 20 smokers and 61 COPD patients with moderate to severe airflow obstruction. We compared total EMPs (CD31+CD42b-), apoptotic if they co-expressed Annexin-V+ or activated if they co-expressed CD62E+, circulating PCs (CD34+CD133+CD45+) and the EMPs/PCs ratio between groups. Results COPD patients presented increased levels of total and apoptotic circulating EMPs, and an increased EMPs/PCs ratio, compared with nonsmokers. Women had less circulating PCs than men through all groups and those with COPD showed lower levels of PCs than both control groups. In smokers, circulating EMPs and PCs did not differ from nonsmokers, being the EMPs/PCs ratio in an intermediate position between COPD and nonsmokers. Conclusions We conclude that COPD patients present an imbalance between endothelial damage and repair capacity that might explain the frequent concurrence of cardiovascular disorders. Factors related to the disease itself and gender, rather than cigarette smoking, may account for this imbalance.
Collapse
Affiliation(s)
- Jéssica García-Lucio
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
| | - Victor I. Peinado
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES); Madrid, Spain
| | - Lluís de Jover
- Biostatistics Unit, Department of Public Health, School of Medicine, University of Barcelona; Barcelona, Spain
| | - Roberto del Pozo
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
| | - Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES); Madrid, Spain
| | - Cristina Bonjoch
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
| | - Núria Coll-Bonfill
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
| | - Tanja Paul
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES); Madrid, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES); Madrid, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona; Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES); Madrid, Spain
- * E-mail:
| |
Collapse
|
43
|
Nydegger C, Martinelli C, Di Marco F, Bulfamante G, von Segesser L, Tozzi P, Samaja M, Milano G. Phosphodiesterase-5 Inhibition Alleviates Pulmonary Hypertension and Basal Lamina Thickening in Rats Challenged by Chronic Hypoxia. Front Physiol 2018; 9:289. [PMID: 29636700 PMCID: PMC5880920 DOI: 10.3389/fphys.2018.00289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/12/2018] [Indexed: 11/30/2022] Open
Abstract
Background: Hypoxia represents both an outcome of cardiopulmonary diseases and a trigger for severe pulmonary complications as pulmonary hypertension. Because nitric oxide (NO) is a critical mediator in the development of pulmonary hypertension, the modulators of its downstream function may become target of pharmacological interventions aimed at alleviating the impact of this condition. Here, we investigate the effects of an early administration of phosphodiesterase-5 inhibitor in rats where pulmonary artery hypertension was induced by chronic exposure to hypoxia. Methods: Rats were divided into three groups: normoxic control, hypoxic with no treatments (2 weeks breathing an atmosphere containing 10% oxygen), and hypoxic treated with sildenafil (1.4 mg/Kg per day in 0.3 mL i.p.). After sacrifice, hearts and lungs were removed and harvested for analyses. Results: Sildenafil reduced hypoxia-induced right ventricle hypertrophy without effects in lung hypertrophy, and blunted the increase in right ventricle pressure without effects on left ventricle pressure. Furthermore, the NO-producing systems (i.e., the phosphorylation of the endothelial isoforms of NO synthase that was measured in both myocardial and lung tissues), and the blood NO stores (i.e., the plasma level of nitrates and nitrites) were up-regulated by sildenafil. We did not find significant effects of sildenafil on weight and hemoglobin concentration. Morphological analysis in lung biopsies revealed that 2-week hypoxia increased the frequency of small pulmonary vessels leaving large vessels unaffected. Finally, ultrastructural analysis showed that sildenafil down-regulated the hypoxia-induced increase in the thickness of the pulmonary basal lamina. Conclusions: In this model of pulmonary hypertension, sildenafil contrasts the negative effects of hypoxia on pulmonary vascular and right ventricle remodeling. This action does not only encompass the canonical vasomodulatory effect, but involves several biochemical pathways. Although the human pathological model is certainly more complex than that described here (for example, the inflammatory issue), the potential role of phosphodiesterase-5 for long-term treatment, and perhaps prevention, of pulmonary hypertension is worthy of investigation.
Collapse
Affiliation(s)
- Coline Nydegger
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - Fabiano Di Marco
- Department of Health Science, University of Milan, Milan, Italy.,ASST Santi Paolo e Carlo, Milan, Italy
| | - Gaetano Bulfamante
- Department of Health Science, University of Milan, Milan, Italy.,ASST Santi Paolo e Carlo, Milan, Italy
| | - Ludwig von Segesser
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Piergiorgio Tozzi
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Michele Samaja
- Department of Health Science, University of Milan, Milan, Italy
| | - Giuseppina Milano
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
44
|
Huertas A, Guignabert C, Barberà JA, Bärtsch P, Bhattacharya J, Bhattacharya S, Bonsignore MR, Dewachter L, Dinh-Xuan AT, Dorfmüller P, Gladwin MT, Humbert M, Kotsimbos T, Vassilakopoulos T, Sanchez O, Savale L, Testa U, Wilkins MR. Pulmonary vascular endothelium: the orchestra conductor in respiratory diseases. Eur Respir J 2018; 51:13993003.00745-2017. [DOI: 10.1183/13993003.00745-2017] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 02/03/2018] [Indexed: 12/15/2022]
Abstract
The European Respiratory Society (ERS) Research Seminar entitled “Pulmonary vascular endothelium: orchestra conductor in respiratory diseases - highlights from basic research to therapy” brought together international experts in dysfunctional pulmonary endothelium, from basic science to translational medicine, to discuss several important aspects in acute and chronic lung diseases. This review will briefly sum up the different topics of discussion from this meeting which was held in Paris, France on October 27–28, 2016. It is important to consider that this paper does not address all aspects of endothelial dysfunction but focuses on specific themes such as: 1) the complex role of the pulmonary endothelium in orchestrating the host response in both health and disease (acute lung injury, chronic obstructive pulmonary disease, high-altitude pulmonary oedema and pulmonary hypertension); and 2) the potential value of dysfunctional pulmonary endothelium as a target for innovative therapies.
Collapse
|
45
|
Lu Q, Gottlieb E, Rounds S. Effects of cigarette smoke on pulmonary endothelial cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L743-L756. [PMID: 29351435 DOI: 10.1152/ajplung.00373.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cigarette smoking is the leading cause of preventable disease and death in the United States. Cardiovascular comorbidities associated with both active and secondhand cigarette smoking indicate the vascular toxicity of smoke exposure. Growing evidence supports the injurious effect of cigarette smoke on pulmonary endothelial cells and the roles of endothelial cell injury in development of acute respiratory distress syndrome (ARDS), emphysema, and pulmonary hypertension. This review summarizes results from studies of humans, preclinical animal models, and cultured endothelial cells that document toxicities of cigarette smoke exposure on pulmonary endothelial cell functions, including barrier dysfunction, endothelial activation and inflammation, apoptosis, and vasoactive mediator production. The discussion is focused on effects of cigarette smoke-induced endothelial injury in the development of ARDS, emphysema, and vascular remodeling in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Eric Gottlieb
- Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| |
Collapse
|
46
|
Polverino F, Celli BR, Owen CA. COPD as an endothelial disorder: endothelial injury linking lesions in the lungs and other organs? (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018758528. [PMID: 29468936 PMCID: PMC5826015 DOI: 10.1177/2045894018758528] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/21/2018] [Indexed: 12/27/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic expiratory airflow obstruction that is not fully reversible. COPD patients develop varying degrees of emphysema, small and large airway disease, and various co-morbidities. It has not been clear whether these co-morbidities share common underlying pathogenic processes with the pulmonary lesions. Early research into the pathogenesis of COPD focused on the contributions of injury to the extracellular matrix and pulmonary epithelial cells. More recently, cigarette smoke-induced endothelial dysfunction/injury have been linked to the pulmonary lesions in COPD (especially emphysema) and systemic co-morbidities including atherosclerosis, pulmonary hypertension, and chronic renal injury. Herein, we review the evidence linking endothelial injury to COPD, and the pathways underlying endothelial injury and the "vascular COPD phenotype" including: (1) direct toxic effects of cigarette smoke on endothelial cells; (2) generation of auto-antibodies directed against endothelial cells; (3) vascular inflammation; (4) increased oxidative stress levels in vessels inducing increases in lipid peroxidation and increased activation of the receptor for advanced glycation end-products (RAGE); (5) reduced activation of the anti-oxidant pathways in endothelial cells; (6) increased endothelial cell release of mediators with vasoconstrictor, pro-inflammatory, and remodeling activities (endothelin-1) and reduced endothelial cell expression of mediators that promote vasodilation and homeostasis of endothelial cells (nitric oxide synthase and prostacyclin); and (7) increased endoplasmic reticular stress and the unfolded protein response in endothelial cells. We also review the literature on studies of drugs that inhibit RAGE signaling in other diseases (angiotensin-converting enzyme inhibitors and angiotensin receptor blockers), or vasodilators developed for idiopathic pulmonary arterial hypertension that have been tested on cell culture systems, animal models of COPD, and/or smokers and COPD patients.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Bartolome R. Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| |
Collapse
|
47
|
Singh N, Singh H, Jagavelu K, Wahajuddin M, Hanif K. Fatty acid synthase modulates proliferation, metabolic functions and angiogenesis in hypoxic pulmonary artery endothelial cells. Eur J Pharmacol 2017; 815:462-469. [DOI: 10.1016/j.ejphar.2017.09.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 01/06/2023]
|
48
|
Dulaney DT, Dokus KM, McIntosh S, Al-Judaibi B, Ramaraju GA, Tomiyama K, Levstik M, Hernandez-Alejandro R, Orloff MS, Kashyap R. Tobacco Use is a Modifiable Risk Factor for Post-Transplant Biliary Complications. J Gastrointest Surg 2017; 21:1643-1649. [PMID: 28785937 DOI: 10.1007/s11605-017-3519-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/24/2017] [Indexed: 01/31/2023]
Abstract
PURPOSE Biliary complications following liver transplantation are a significant source of morbidity, potentially leading to graft failure necessitating retransplantation. We sought to evaluate smoking as an independent risk factor for post-transplant biliary complications. METHODS The clinical course of all adult primary deceased donor liver transplants at our center from 1992 to 2012 was reviewed. Eligible patients were assigned to cohorts based on their lifetime tobacco exposure: never smokers indicating 0 pack-year exposure and all others were ever smokers. Biliary complications were defined as strictures, leaks, or bilomas requiring intervention. Complication rates were analyzed using univariate regression models correlated with donor and recipient characteristics. Associations found during univariate analysis were included in the final multivariate Cox model. RESULTS Eight hundred sixty-five subjects were followed for a median of 65 months; 482 (55.7%) of patients had a positive smoking history at the time of transplant. In univariate analysis, positive tobacco smoking history (HR = 1.36; p = 0.037) and increased time from quit date to transplantation (HR = 0.998; p = 0.011) were positive and negative predictors of biliary complication, respectively. Lifetime tobacco exposure remained a significant predictor of biliary complication on multivariate analysis (HR = 1.408; p = 0.023). CONCLUSIONS Smoking status is an independent predictor of post-transplant biliary complications, and the data presented reinforces the importance of early smoking cessation in the pre-transplantation period.
Collapse
Affiliation(s)
- David T Dulaney
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Katherine M Dokus
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Scott McIntosh
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Bandar Al-Judaibi
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Gopal A Ramaraju
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Koji Tomiyama
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Mark Levstik
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Roberto Hernandez-Alejandro
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Mark S Orloff
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA
| | - Randeep Kashyap
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Box SURG - TRANSPLANT, Rochester, NY, 14642, USA.
| |
Collapse
|
49
|
Malerba M, Nardin M, Radaeli A, Montuschi P, Carpagnano GE, Clini E. The potential role of endothelial dysfunction and platelet activation in the development of thrombotic risk in COPD patients. Expert Rev Hematol 2017; 10:821-832. [PMID: 28693343 DOI: 10.1080/17474086.2017.1353416] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Despite lack of knowledge in the field, several studies have underlined the role of endothelium dysfunction and platelet activation as significant players in the development and progression of chronic obstructive pulmonary disease (COPD). Indeed, endothelium plays a crucial role in vascular homeostasis and impairment, due to the inflammation process enhanced by smoking. Chronic inflammation and endothelial dysfunction have been proved to drive platelet activity. Consequently, thrombotic risk is enhanced in COPD, and might explain the higher percentage of cardiovascular death in such patients. Areas covered: This review aims to clarify the role of endothelium function and platelet hyper-activity as the pathophysiological mechanisms of the increased thrombotic risk in COPD. Expert commentary: In COPD patients, chronic inflammation does not impact only on lung parenchyma, but potentially involves all systems, including the endothelium of blood vessels. Impaired endothelium has several consequences, such as reduced vasodilatation capacity, enhanced blood coagulation, and increased platelet activation resulting in higher risk of thrombosis in COPD patients. Endothelium dysfunction and platelet activation are potential targets of therapy in patients with COPD aiming to reduce their risk of cardiovascular events.
Collapse
Affiliation(s)
- Mario Malerba
- a Department of Internal Medicine , University of Brescia and ASST Spedali Civili , Brescia , Italy
| | - Matteo Nardin
- a Department of Internal Medicine , University of Brescia and ASST Spedali Civili , Brescia , Italy
| | | | - Paolo Montuschi
- c Department of Pharmacology, Faculty of Medicine , University Hospital Agostino Gemelli Catholic University of the Sacred Heart, Pharmacology , Rome , Italy
| | - Giovanna E Carpagnano
- d Department of Medical and Surgical Sciences , Institute of Respiratory Diseases, University of Foggia , Foggia , Italy
| | - Enrico Clini
- e Department of Medical and Surgical Sciences , University of Modena-Reggio Emilia , Modena , Italy
| |
Collapse
|
50
|
Ursoniu S, Mikhailidis DP, Serban MC, Penson P, Toth PP, Ridker PM, Ray KK, Kees Hovingh G, Kastelein JJ, Hernandez AV, Manson JE, Rysz J, Banach M. The effect of statins on cardiovascular outcomes by smoking status: A systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 2017; 122:105-117. [PMID: 28602797 DOI: 10.1016/j.phrs.2017.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 01/03/2023]
Abstract
Smoking is an important risk factor for cardiovascular disease (CVD) morbidity and mortality. The impact of statin therapy on CVD risk by smoking status has not been fully investigated. Therefore we assessed the impact of statin therapy on CVD outcomes by smoking status through a systematic review of the literature and meta-analysis of available randomized controlled trials (RCTs). The literature search included EMBASE, ProQuest, CINAHL and PUBMED databases to 30 January 2016 to identify RCTs that investigated the effect of statin therapy on cumulative incidence of major CVD endpoints (e.g. non-fatal myocardial infarction, revascularization, unstable angina, and stroke). Relative risks (RR) ratios were calculated from the number of events in different treatment groups for both smokers and non-smokers. Finally 11 trials with 89,604 individuals were included. The number of smokers and non-smokers in the statin groups of the analyzed studies was 8826 and 36,090, respectively. The RR for major CV events was 0.73 (95% confidence interval [CI]: 0.67-0.81; p<0.001) in nonsmokers and 0.72 (95%CI: 0.64-0.81; p<0.001) in smokers. Moderate to high heterogeneity was observed both in non-smokers (I2=77.1%, p<0.001) and in smokers (I2=51.6%, p=0.024) groups. Smokers seemed to benefit slightly more from statins than non-smokers according to the number needed to treat (NNT) analysis (23.5 vs 26.8) based on RRs applied to the control event rates. The number of avoided events per 1000 individuals was 42.5 (95%CI: 28.9-54.6) in smokers and 37.3 (95%CI: 27.2-46.4) in non-smokers. In conclusion, this meta-analysis suggests that the effect of statins on CVD is similar for smokers and non-smokers, but in terms of NNTs and number of avoided events, smokers seem to benefit more although non-significantly.
Collapse
Affiliation(s)
- Sorin Ursoniu
- Department of Functional Sciences, Discipline of Public Health, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, UK
| | - Maria-Corina Serban
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Functional Sciences, Discipline of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Peter Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Peter P Toth
- Preventive Cardiology, CGH Medical Center, Sterling, Illinois, USA; The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kausik K Ray
- Department of Primary Care and Public Health, School of Public Health, Imperial College London, UK
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - John J Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Adrian V Hernandez
- Health Outcomes and Clinical Epidemiology Section, Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; School of Medicine, Universidad Peruana de Ciencias Aplicadas (UPC), Lima, Peru
| | - JoAnn E Manson
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jacek Rysz
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland; Polish Mother's Memorial Hospital Research Institute, Lodz, Poland.
| | | |
Collapse
|