• Open Access
    Review

    The role of pulmonary vascular endothelium in chronic obstructive pulmonary disease (COPD): Does endothelium play a role in the onset and progression of COPD?

    Silvia Siragusa 1
    Giulia Natali 2
    Antonella Maria Nogara 1
    Marcello Trevisani 2
    Costanza Anna Maria Lagrasta 1
    Silvia Pontis 2*

    Explor Med. 2023;4:1116–1134 DOI: https://doi.org/10.37349/emed.2023.00199

    Received: July 14, 2023 Accepted: September 18, 2023 Published: December 29, 2023

    Academic Editor: Bernhard Ryffel, University of Orleans, France

    This article belongs to the special issue Lung Fibrosis—Models and Mechanisms

    Abstract

    Chronic obstructive pulmonary disease (COPD) is an inflammatory lung pathology characterized by persistent airflow limitation and is the third leading cause of death globally. COPD pathophysiology includes both environmental and host risk factors and the presence of comorbidities contributes to its harmful outcome. Cardiovascular disease (CVD) is closely related to COPD and their coexistence is associated with worse outcomes than either condition alone. COPD impairs the cardiovascular system favoring mostly endothelial dysfunction that is a significant COPD prognostic factor at different stages of the disease. The mechanisms promoting endothelial dysfunction in the systemic and/or pulmonary circulation of COPD patients are different and include systemic inflammation, alteration of adhesion and pro-inflammatory molecules, oxidative stress, cellular senescence, and apoptosis. Nevertheless, the role of endothelium in the onset and progression of COPD and CVD is not yet fully understood. Hence, the purpose of this narrative review is to analyze the literature and provide evidence supporting the importance of endothelial dysfunction in COPD.

    The role of endothelium and its potential relationship in COPD and CVD. The dotted arrows in the figure indicate that the factors linking COPD and CVD are not yet fully understood and are likely to be interconnected through endothelial dysfunction. While the solid arrows represent the well-known risk factors for COPD and the mechanisms associated with endothelial dysfunction. Created with BioRender.com

    Keywords

    Endothelium, chronic obstructive pulmonary disease, pulmonary, vascular, cardiovascular disease

    Introduction

    Chronic obstructive pulmonary disease (COPD) is an incurable inflammatory pathology [1] characterized by persistent limitation of the airflow and lung tissue destruction [2]. Approved pharmacological treatments (e.g., long-acting muscarinic antagonists, long-acting beta-agonists, corticosteroids) display the limited ability to halt disease progression mainly because they do not significantly tackle the underlying inflammation. Considerable efforts have been made to develop efficient anti-inflammatory compounds however drugs that can reverse the patient’s pathological state are still lacking [3]. For all these reasons, COPD remains a major worldwide health problem and global burden of disease studies have estimated COPD as the third leading cause of death worldwide [4].

    The pathophysiology of COPD is complex and it includes both environmental and host risk factors. Long-term exposure to irritant gases or particles and more frequently, to cigarette smoke (CS) are among the first [5]. Whereas host factors include polymorphisms of genes like those encoding for metalloproteinase matrix 12 and glutathione S-transferase both associated with decreased lung function [6, 7] or increased risk of COPD onset [8]. Moreover, a small percentage of individuals (about 1–5%) develop COPD secondary to a genetic deficiency in alpha-1 antitrypsin (AAT). Beyond genetics, lung cell hyperresponsiveness to stimuli and poor lung growth during childhood play crucial roles in the development and severity of COPD [9, 10].

    Patients experience cough, mucus hypersecretion, wheezing, chest tightness, and breathing difficulty, especially during physical activities [11]. In addition to the daily symptom burden, COPD may be punctuated by periods of acute worsening of respiratory symptoms requiring additional therapeutic interventions [12] and resulting in a significant increase in both morbidity and mortality [13]. These events are called exacerbations and are mainly triggered by viral and bacterial respiratory infections. This complex and faceted clinical scenario is classified according to Global Initiative for Chronic Obstructive Lung Disease (GOLD) guidelines that rank the severity of airflow limitation according to specific spirometric cut-offs as it follows: GOLD 1 (mild), forced expiratory volume in 1 s (FEV1) ≥ 80% predicted; GOLD 2 (moderate), 50% ≤ FEV1 < 80% predicted; GOLD 3 (severe), 30% ≤ FEV1 < 50% predicted; GOLD 4 (very severe), FEV1 < 30% predicted [14]. Moreover, a post-bronchodilator FEV1/forced vital capacity (FVC) < 0.70 confirms the presence of persistent obstruction. Spirometry is the most reproducible and objective measurement of airflow limitation, representing the fundamental test used to diagnose COPD nowadays.

    Even if COPD is heterogenous in its manifestations, it usually includes two major pathologies: small airway disease with peribronchiolar fibrosis and emphysema with alveolar wall destruction [15]. In chronic bronchitis, the principal alteration is airflow obstruction caused mainly by airways remodeling. On one side, bronchial tube become inflamed and narrowed while on the other, lung produces more mucus because of submucosal glands hypertrophy and hyperplasia that can further affect the narrowed tubes [16]. In emphysema, the pre-eminent physio-pathological alteration is the loss of elastic return of the lung by parenchymal destruction. These conditions are not mutually exclusive but can occur together [17].

    A relevant aspect of the mechanism of both airway disease and emphysema is inflammation. Indeed, COPD is mainly an inflammatory disease and different inflammatory pathways can be activated [18]. The inflammatory response involves both innate and adaptive immunity and is most pronounced in the bronchial walls with neutrophilic infiltration, the commonest inflammatory phenotype of COPD. Exposure to CS, pollutants, and oxidants [19] causes the release of chemotactic factors such as interleukin (IL)-4, IL-5, IL-6, IL-8, IL-13, interferon-γ, monokine, and leukotriene B4 by the airway epithelium and macrophages, which are the key inflammatory cells coating the airway mucosa [20]. Excessive release of IL causes accumulation in the airways of neutrophils, CD8 T cells, and CD4 helper T cells [21]. This causes an extended immune response that causes elastin degradation mainly in alveoli, the consequence of which is a lack of elastic pulmonary recoil and emphysema. In addition, the accumulation and the prolonged activation of these factors result in hypertropia and hyperplasia of goblet cells, with an increased production of mucus. Along with chemotactic factors, macrophages, and epithelial cells secrete transforming growth factor-β (TGF-β), which causes fibroblast activation [22]. Fibroblasts produce a connective tissue growth factor, a chemokine that promotes the senescence of lung epithelial cells, decreasing cell rebuilding and leading to emphysema [23].

    It is known that the inflammatory phenotype most associated with COPD is the neutrophil phenotype, but the most recent searches show a 10–40% increase of eosinophilic inflammation in the sputum and blood of COPD patients [24]. Eosinophilic inflammation is related to a type 2 inflammatory profile that leads to the activation of IL-5, the main inflammatory cytokine responsible for the maturation, survival, and activation of eosinophils. Although increased IL-5 has been detected in patients’ sputum, anti-IL-5 therapies are not effective in preventing exacerbations, highlighting that the mechanism by which eosinophils contribute to COPD is not yet to be understood [25]. However, eosinophilic inflammation and an increase in the eosinophilic count can be found in lung tissue, sputum, and blood both during stable disease and during exacerbations, so for this reason, eosinophils could be considered as biomarker of disease [26].

    Another level of complexity that describes the heterogeneity of the COPD patient population is given by comorbidities which influences the prognosis of COPD. Comorbidities not primarily involving the vascular system such as diabetes, metabolic syndrome, and lung cancer can be distinguished from those including cardiovascular disease (CVD) [27]. CVD is often linked to COPD and their coexistence is associated with worse outcomes than either condition alone [28]. COPD impairs the cardiovascular system, especially the endothelium, favoring the development of atherosclerotic plaques. As a result, CVD mortality accounts for 25% of all causes of death in COPD patients [29]. Not by chance, COPD and CVD share common risk factors, pathophysiological processes, clinical signs, and symptoms [30]. Among them, chronic low-grade systemic inflammation emerged as a possible link between the two disorders [31].

    Based on these contentions, several questions remain open: Are COPD and CVD two different pathologies? How is one reflected in the other? And above all, is COPD a risk factor for CVD or the opposite?

    In the attempt to answer these questions, is relevant to highlight the importance of endothelium and its dysfunctions in both COPD and CVD as shown in Figure 1. Several studies suggest that endothelial dysfunction contributes to the progression of CVD and at the same time, it is linked to COPD severity [32]. In COPD, endothelial dysfunction occurs in the early stages of the disease and worsens with pulmonary obstruction severity and during acute exacerbations [33]. In this scenario, it becomes very attractive to study and explore the endothelium, through which inflammatory cells must transmigrate [transendothelial migration (TEM)] to reach the lung parenchyma and cause damage ultimately leading to COPD onset and progression. Understanding the mechanisms underlying the trafficking of leukocytes, neutrophils, cytokine, chemokine, and inflammatory molecules across the endothelium to the lung parenchyma, new treatments may be developed and the risk of COPD onset may be reduced.

    Exposure to harmful substances leads to the development of inflammatory mechanisms causing COPD and CVD. The two diseases are closely related and can lead to endothelial dysfunction. Figure was created with Canva

    Endothelium, from physiological function to COPD dysfunction

    Vascular endothelial cells (ECs) cover the internal surface of the vessels forming a continuous monolayer [34]. ECs rest on a basal membrane consisting of collagen and glycoproteins, while on the luminal part, there are glycocalyces, a structure of proteoglycans and glycoproteins that have different functions [35]. The basal membrane is also constituted by a non-continuous layer of pericytes that are mediators of different microvascular processes, such as angiogenesis and ECs proliferation [36]. Endothelium is involved in multiple and different mechanisms such as vascular permeability, angiogenesis, leukocyte trafficking, inflammation, and coagulation [37, 38]. In the respiratory system, it has another important role: it detoxifies xenobiotics incoming via the airways, CS, and air pollution [39]. The participation of pulmonary capillary ECs in the blood-air barrier makes the integrity of pulmonary endothelium critical for the maintenance of the lung physiological performance [40].

    The first description of vascular injury, including capillary loss, in emphysematous lungs of COPD patients comes from Liebow [41]. Thanks to histological studies, he concluded that, in centrilobular emphysema, a lower inflow of blood in the pre-capillaries contributes to alveolar septa damage [41]. What Liebow [41] discovered for years remained obscure, but later, significant advances were made in the study of endothelial dysfunction in lung diseases. Several years later, Dinh-Xuan et al. [42] described that endothelial dysfunction occurs in the pulmonary arteries of COPD patients with end-stage disease as evaluated by in vitro experiments measuring relaxation of pulmonary artery rings using increasing doses of exogenous acetylcholine and adenosine diphosphate (ADP). They suggested that endothelial dysfunction in end stage COPD might contribute to the development of pulmonary arterial hypertension in COPD. However, a subsequent study conducted by Peinado et al. [43] reported that endothelial dysfunction appears not only in the end stage but also as an early feature of COPD. This relevant clinical study was conducted on 41 patients, divided into non-smokers, smokers with normal lung function, and patients with COPD to check whether patients with mild COPD showed endothelial dysfunction of the pulmonary artery. Endothelial-dependent relaxation, mediated by nitric oxide (NO), was evaluated in vitro in pulmonary artery rings exposed to different concentrations of acetylcholine and ADP. Authors suggested that the intima of the small pulmonary arteries was thickened and level of relaxation reduced in both patients with mild COPD and smokers with regular lung activity suggesting that smoking may cause morpho-functional changes in COPD [43]. They also detected endothelial apoptosis and endothelial dysfunction in the lungs of COPD patients thanks to computed tomography that revealed reduced and reshaped peripheral vascularization [44].

    The mechanisms that promote endothelial dysfunction in the systemic and/or pulmonary circulation of COPD patients are different and include systemic inflammation, alteration of adhesion and pro-inflammatory molecules, oxidative stress, cellular senescence, and apoptosis as shown in Figure 2.

    Different mechanisms by which endothelial dysfunction is promoted in COPD, including systemic inflammation, oxidative stress, ECs senescence, and apoptosis. ROS: reactive oxygen species; ICAM-1: intracellular adhesion molecule-1; MAC-1: macrophage-1 antigen; ELAM-1: endothelial-leukocyte adhesion molecule; HIF-1α: hypoxia-inducible factor 1α; VEGF: vascular endothelial growth factors. Red arrows indicate an increase or decrease in the corresponding factors involved in the mechanisms. Created with BioRender.com

    Systemic inflammation and alteration of inflammatory mediators

    Systemic inflammation occurring in COPD patients may contribute to the development of both pulmonary and systemic endothelial dysfunction. Activation and alteration of adhesion molecules in ECs promote the inflammatory reactions occurring in pulmonary vessels. The surface activation of cellular adhesion molecules represents a common endothelial response to a variety of toxic and inflammatory stimuli and their shedding into the systemic circulation provides evidence of endothelial dysfunction.

    As described above, the endothelium produces and activates molecules and inflammatory factors, which exit the bloodstream and reach the target tissue through the endothelium. Trafficking of leukocytes out of the circulation into tissues is a highly regulated, complex, multistep process. Physiologically, inflammation is initiated by a relatively loose adhesion of leukocytes to ECs followed by a firmer adhesion and transmigration of leukocytes across pulmonary ECs [45]. Specifically, neutrophils are provided by two different mechanisms: The first one is called paracellular transmigration and consists of a connection with the apical domain of ECs via cell surface proteins (e.g., pseudopods) over one or between two ECs. The second mechanism is called TEM by which neutrophils can extravasate at junction regions between two ECs. This process consists of multiple steps of interaction between neutrophils and ECs and has a relevant role in the inflammatory response [46]. TEM is a mechanism by which endothelium may play a role in COPD and therefore may be important in the development of inflammation and in the pathogenesis of COPD. Patients with COPD have high levels of TEM and during this process, the MAC-1, an important human cell surface receptor found on B and T lymphocytes, is upregulated [47]. MAC-1 binds to ICAM-1 on the surface of ECs resulting in increased ICAM-1. This mechanism could be clinically relevant, as when ICAM-1 serum levels are high, lung function is reduced. Conversely, when ICAM-1 is turned off, pulmonary inflammation is reduced, supporting the hypothesis that ICAM-1 upregulation could relate to the increased inflammation seen in COPD patients [48]. Furthermore, another study by Johnston et al. [49] suggested that the upregulation of ICAM-1 could be reduced by an anti-ICAM-1 molecule, administered at the initial stage of the disease, thus preventing COPD exacerbations and its adverse outcomes [49, 50]. Another relevant adhesion molecule involved in TEM is ELAM-1. This has also been found upregulated in COPD patients’ serum and its levels are particularly high in patients with chronic bronchitis. Altogether, this evidence further supports the involvement of adhesion molecules in lung inflammation and COPD pathogenesis [51].

    In the last years, scientists improved the research about a variety of COPD inflammatory biomarkers [52]. This research field is interesting for the study of disease progression, since inflammatory biomarkers are diagnostic and prognostic factors of COPD patients with acute exacerbation of COPD (AECOPD). For example, are studied as markers for degradation and repair processes factors like neutrophil elastase (NE) or matrix metalloproteases (MMP), which are detectable in bronchoalveolar lavage fluid (BALF) or sputum [53]. Among the most significative systemic markers of COPD there are fibrinogen and acute C-reactive protein (CRP), which is one of the predictive factors. This project, called “Evaluation of COPD Longitudinally to Identify Predictive Surrogate End-points (ECLIPSE)”, is a three-year longitudinal study whose main objective was to identify factors that predict disease progression in individuals with different endotype of COPD [54]. Literature studies show that serum CRP is associated with mortality, morbidity, number of exacerbations and is inversely related to lung function. Increased serum concentrations of IL-6, tumor necrosis factor α (TNFα), and monocyte chemoattractant protein-1 (MCP-1) are also related to systemic inflammation in COPD patients [55].

    As described in the sections above, the inflammatory endotype in COPD can be mediated by neutrophils [T helper 1 (Th1)] and by eosinophils (Th2) [56]. Eosinophil recruitment to inflammation sites is mediated by eotaxins [eotaxin-1 (CCL11), eotaxin-2 (CCL24), eotaxin-3 (CCL26)]. These chemokines are considered as candidate biomarkers to monitor COPD development and progression since they are upregulated in COPD patients [57]. The main source of eotaxins are epithelial cells, motile cells present in BALF (macrophages, monocytes, basophils, eosinophils, lymphocytes), and structural cells like fibroblasts or smooth muscle cells [58]. Eotaxins can also be released by ECs but there are few studies supporting this statement. One of these showed that CCL11 can be released by both epithelial and ECs to favor angiogenesis. Unfortunately, none of these studies describe the release of eotaxins by ECs in the context of COPD [59]. Similar considerations can be made for neutrophils. Indeed, there is evidence about neutrophil chemoattractant cytokines [C-X-C motif chemokine ligand 1 (CXCL1) and CXCL5)] release by ECs but not in the context of COPD even if they are dysregulated in COPD patients [60, 61]. To study Th2 response is particularly appealing since it defines a specific COPD patient population as shown by the ECLIPSE study reporting that eosinophilic inflammation was present in 37.4% of patients [62]. In these patients, eosinophil count might be relevant to predict their disease progression and to choose the most appropriate pharmacological therapy. Indeed, peripheral eosinophil level from a complete blood count has been evaluated as a surrogate marker for corticosteroid responsiveness and eosinophilic bronchitis [63]. A higher blood eosinophil count appears to indicate a subgroup of COPD patients in which the use of inhaled corticosteroids (ICS) results in reduced exacerbation frequency and when ICS [64] is withdrawn significant increase in peripheral eosinophil count has been associated with a higher exacerbation rate [65]. To deepen these issues could help to improve the discovery of therapeutic targets and to identify patients who are likely to respond to classic or novel treatments.

    Neurogenic inflammation

    Neurogenic inflammation is confined to the central and peripheral nervous system and is caused by nerve activation inducing the release of neuropeptides and rapid extravasation. During airway inflammation, neuropeptides are secreted from the airways in response to a multitude of inflammatory mediators, such as exogenous irritants like CS or gas. According to this, neurogenic inflammation may participate in the development and progression of chronic inflammatory airway diseases such as allergic asthma or COPD. Among the large variety of neuropeptides stored in and secreted from airway sensory nerves the most studied are the tachykinins like substance P (SP), neurokinin A (NKA), and the calcitonin gene-related peptide (CGRP) [66]. Tachykinins can bind to three different receptors, called neurokinin receptors (NK). Specifically, SP acts via NK1, NKA acts via NK2 and neurokinin B acts via NK3. The first one is predominantly localized to airway epithelium, submucosal glands, and vessels, the second one is mainly expressed by airway smooth muscle, while NK3 is expressed by airway parasympathetic ganglia. Tachykinins exert various actions on airway functions. They have a direct effect on coronary arteries to decrease or increase tone. Stimulation of NK1 receptors on the endothelium causes vasodilatation mediated by NO. They can vasodilate or vasoconstrict the state of the endothelium depending on the situation; this state appears to be of considerable importance [67]. Tachykinins also enhance eosinophils chemotaxis through the activation of alveolar macrophages and monocytes that release inflammatory cytokines such as IL-6. CGRP is a 37 amino acid peptide that is abundantly expressed in the respiratory tract of several species and is co-stored and co-localized with tachykinins in sensory nerve fibres innervating both upper and lower airways. CGRP is a powerful vasodilator, with a long-lasting effect on bronchial vessels and this is evident in vitro and in vivo. Mapping of the receptors showed that CGRP receptors are predominantly situated in the bronchial vessels, in the epithelium of the human airways, and in the smooth muscles on which it has effects on tone by acting indirectly through the release of other constrictors, such as endothelin. Like tachykinins, CGRP is chemotactic for eosinophils [68]. The molecular mechanisms underlying the different events of neurogenic inflammation in the airways of mammals have been identified thanks to animal models. Based on data obtained from these studies, the existence of a neurogenic inflammatory component in human airway diseases such as asthma and COPD seems very likely. Nevertheless, further studies are required to assess the clinical relevance of this inflammatory process in the onset and progression of COPD [69].

    Oxidative stress

    Oxidative stress is another crucial mechanism underlying COPD development and may be a causal link in COPD comorbidities such as CVD [70]. Oxidative stress is caused by an alteration of the prooxidant/antioxidant balance that results in the production and accumulation of ROS. ROS refer to a variety of molecules, either containing unpaired electrons (such as superoxide, O2) or having a strong ability to oxidize other molecules (such as hydrogen peroxide, H2O2) [71], and are produced by both endogenous and exogenous factors. Inflammatory cells, infiltrated in pulmonary arteries, may be one of the major sources of endogenous ROS generated mainly by xanthine oxidase, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and mitochondria. On the other hand, genetic factors, CS, and air pollutants are the main producers of exogenous ROS [72]. More in detail, CS favors the activation of inflammatory cells through the recruitment of transcription factors nuclear factor-kappa B (NF-κB), p38, mitogen-activated protein kinase (MAPK), and post-translational modification of histone deacetylase (HDAC) in macrophages [73]. These processes stimulate the cells to release pro-inflammatory cytokines and activate the immune system and inflammation [74]. Experimental studies performed with cultured rat pulmonary microvascular ECs demonstrated that exposure to tobacco smoke condensate causes increased transcription and activity of the ROS generating enzyme xanthine dehydrogenase/oxidase [75]. Among the causes of oxidative stress, there is also a decrease in antioxidants. Antioxidants, such as superoxide dismutase (SOD), glutathione (GSH), and nuclear factor erythroid 2-related factor 2 (Nrf2) are thought to play very important roles in the lungs since they are continuously exposed to oxidant endogenous and exogenous risk factors. NO released from the vascular endothelium is also an antioxidative factor that neutralizes O2 [76] and impairment of NO synthesis marks the onset of endothelial dysfunction that is mainly mediated by the endothelial NO synthase (eNOS) uncoupling mechanism [77].

    Increased oxidative stress and ROS overproduction may cause not only endothelial dysfunction but also programmed ECs death such as ferroptosis, pyroptosis, or necroptosis that are involved in the pathophysiology of many diseases, like COPD [78]. Ferroptosis is an iron-dependent process and is characterized by lipid peroxidation, which is the reaction by which OH attacks the carbon-carbon double bonds of lipids. It is directed by the glutathione antioxidant system, which is formed by GSH, glutathione peroxidase (GPX), and glutaredoxin (GRX) but most importantly it can effectively prevent ROS overproduction [79]. In recent years, studies have shown that ferroptosis is closely related to ECs death. For example, Qin et al. [80] found that zinc oxide nanoparticles could induce iron and lipid peroxidation in ECs depending on the dose and time. Luo et al. [81] showed that ferroptosis is related to endothelial dysfunction, meanwhile, Sheng et al. [82] showed that lysophosphatidylcholine (LPC) can induce an increase in the amount of intracellular iron and lipid peroxide levels and mitochondrial atrophy in ECs. In a study conducted by Wang et al. [83] they demonstrated, with in vitro and in vivo experiments, that pulmonary microvascular endothelial barrier dysfunction plays an important role in the mechanism by which COPD affects the progression of atherosclerosis and that oxidative stress and ferroptosis are involved in this effect. Another mechanism of programmed cell death is pyroptosis. The molecular features of pyroptosis include the activation of inflammasome, membrane pore formation, and pro-inflammatory cytokine maturation and release. Pyroptosis may require activation of caspase-1, depending on whether it is activated or not, it may be involved in classical or non-classical inflammatory pathways. Among the classical inflammasome pathway, Nod-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome-mediated pyroptosis is the most extensively studied phenomenon and can be activated by different stimuli, first of all, ROS. The NLRP3 inflammasome in ECs can be caused by stimuli such as low-density lipoprotein oxidized, hyperglycemia, and nicotine, thus leading to the death of ECs [84, 85]. For example, Wu et al. [86] found that oxidized low-density lipoprotein (ox-LDL) induced, in a dose-dependent manner, the upregulation of NLRP3, caspase-1, and IL-1β in ECs. Hang et al. [87] found that ox-LDL stimulated in ECs NLRP3 inflammasome activation, increased IL-1β and IL-18 maturation and secretion, increased intracellular ROS and increased lactate dehydrogenase (LDH) release in ECs. Zhang et al. [88] found that nicotine could activate the NLRP3 inflammasome. The team also found that due to the activation of NLRP3 inflammation can promote the destruction of the proteins of the narrow junction between ECs, resulting in increased vascular permeability. Cau et al. [89] found that angiotensin II (Ang-II) induces endothelial dysfunction, vascular remodeling, and hypertension through NLRP3 inflammasome activation. The last cell death program we here describe is necroptosis, a genetically encoded mechanism of necrotic regulated cell death defined by rupture of the plasma membrane. Necroptosis causes the release of damage-associated molecular patterns (DAMPs) into the extracellular environment and for this reason is highly proinflammatory. Indeed, Pouwels et al. [90] discovered that CS-induced necroptosis and the release of DAMPs trigger neutrophilic airway inflammation in mice BAL and this effect was reduced by treatment with the necroptosis inhibitor necrostatin-1. Wang et al. [91] reported a novel regulatory mechanism of necroptosis-mediated inflammation. Endoplasmic reticulum chaperone glucose-regulating protein 78 (GRP78) promoted a CS extract (CSE)-induced inflammatory response and mucus hyperproduction in airway epithelial cells, likely through the upregulation of necroptosis and subsequent activation of the NF-κB and activator protein-1 pathways [91, 92]. Based on data present in literature, necroptosis is increased in the lungs of COPD patients and its inhibition attenuates CS-induced emphysema, airway inflammation, and remodeling making this process an attractive candidate for COPD patients. Nevertheless, there is not enough data about the correlation between necroptosis and ECs, this could be a starting point for new research.

    Cellular senescence

    Cellular senescence is now considered an important driving mechanism for chronic lung diseases contributing to the pathophysiology of COPD and CVD.

    Cellular senescence is due to telomere shortening (replicative senescence) and oxidative stress (stress-related senescence) with activation of p53 and p16 that lead to activation of p21 and cell cycle arrest, respectively [93, 94]. Patients with COPD showed, accumulated in the lungs, senescent cells, including alveolar epithelial and ECs [9597]. Exposure to CS is among the most relevant factors that cause oxidative stress and is also an important inducer of senescence in COPD, increases markers of senescence in the epithelial cells of the airways, resulting in endothelial damage. Chronic CS exposure causes pulmonary endothelial dysfunction in animal models, healthy young smokers, and chronic smokers with COPD [98, 99]. Accordingly in a study performed by Paschalaki et al. [100] using circulating endothelial progenitors named endothelial colony-forming cells, accelerated endothelial senescence in smokers and patients with COPD was shown compared with healthy controls. They also demonstrated that patients with COPD on ICS exhibit significantly reduced endothelial senescence [100]. Another very recent study by Zeng et al. [101], documented that CSE averages the ubiquitin-specific protease (USP7)/p300 dependent high expression of p53 and activates the expression of target p53 genes, especially p21. They also showed that the activation of p53, via p21, inhibits cell activity and leads to cell cycle arrest with premature senescence of ECs progenitors [101]. Finally, studies reported that senescent pulmonary ECs release more inflammatory markers and this boosts the inflammatory state in COPD patients [102].

    Apoptosis

    Another mechanism that plays an important role in the pathogenesis of COPD is apoptosis and interestingly, increased apoptotic events correlate with ECs damage [103]. Apoptosis is not a marginal event in COPD. Indeed, recent studies suggest that apoptosis of pulmonary vascular ECs plays a role in initiating and participating in the pathogenesis of COPD [104]. Studies show that the number of dead ECs in the lungs of patients with COPD is increased as assessed by specific nuclei fragmentation, which is a clear feature of apoptotic cells [105]. The importance of apoptosis in COPD, especially in the pathogenesis of emphysema, originates from in vitro, in vivo, and in human studies.

    In an in vitro study performed by Farid et al. [106], the authors studied the effect of CSE exposure on VEGF release by human lung fibroblasts demonstrating that the TGF-β receptor/mothers against decapentaplegic homolog 3 (Smad3) pathway mediates CSE-induced VEGF production. VEGF plays important roles by mediating angiogenesis and vascular permeability, inducing cell proliferation, and preventing ECs death. Following CS administration VEGF levels decrease and in parallel emphysematous degeneration increases [106]. In line with this observation, Taraseviciene-Stewart et al. [107] by inducing apoptosis of rat pulmonary vascular ECs, with an intraperitoneal injection of CSE, successfully established a rat emphysema model. To explain this result, they suggested that chronic blockade of VEGF induces alveolar cells apoptosis and emphysema. Along this line, another preclinical study suggests that it is possible to induce emphysema in rodents by deliberately causing endothelial apoptosis through VEGF blockade [108]. The authors treated rodents with a VEGF receptor inhibitor which resulted in EC apoptosis and emphysema. Conversely, when rodents were treated with a caspase inhibitor, apoptosis, and emphysema were reverted, suggesting that apoptosis of ECs may be key in emphysema development [109]. Evidence of increased apoptosis of ECs in humans has also been published. Segura-Valdez et al. [110] were the first to show increased apoptosis in the lungs of COPD patients. They showed that ECs and arterioles exhibited prominent intranuclear staining by in situ-end labeling of fragmented DNA [110]. Moreover, Kasahara et al. [109] described an increased apoptosis of alveolar epithelial and ECs in lung tissue from patients with emphysema. Different and contrasting studies suggest that VEGF and HIF-1α levels change with the severity of pathology. For example, in the study by Yasuo et al. [111], patients with emphysema displayed reduced levels of HIF-1α in lung ECs. On the other hand, a study by Lee et al. [112] showed increased levels of VEGF and HIF-1α in patients with chronic bronchitis. These contrasting findings suggest that the endothelium is variably involved and VEGF is not uniquely implicated in the mechanism of endothelial apoptosis in COPD patients. In this regard, other players could be the cystic fibrosis transmembrane regulator (CFTR) and AAT. CFTR, who participates in the apoptotic process, is necessary for stress-induced apoptosis of human pulmonary ECs as confirmed by the evidence its inhibition, by hydrogen peroxide, attenuates EC apoptosis [113]. AAT has an important role in preventing caspase-3 activation and therefore apoptosis in pulmonary ECs [114]. Overall, endothelial apoptosis may contribute to the destruction of alveoli and consequently, to the onset and progression of emphysema in COPD patients.

    Therefore, understanding the mechanisms by which apoptosis is induced in the pulmonary endothelium is important for the development of new therapies to prevent or treat lung diseases characterized by endothelial dysfunction.

    Measurement of endothelial dysfunction and new parameters to evaluate COPD

    Different methods have been developed over the years to assess endothelial damage in COPD patients and some are used in clinical practice. Today, the most widely used method to assess endothelial dysfunction is flow-mediated dilatation (FMD) that is typically performed on the brachial artery. FMD is a non-invasive diagnostic test that measures the ability of blood vessels to dilate in response to increased blood flow. To this aim, arterial diameter is recorded at baseline and at end-diastole and FMD is calculated as a percentage change from baseline [115]. Alterations of FMD, reflect the inability of the endothelium to produce and release substances that help to regulate blood vessels tone and blood flow. FMD is often used as an indicator of cardiovascular risk [116]. In line with this, Barr et al. [117] found that FMD was impaired in a group of former smokers with COPD, especially in those with low FEV1 and advanced emphysema. In two other studies, authors have shown that COPD patients had lower FMD when compared to healthy controls [118, 119]. Marchetti et al. [120], have conducted a study to evaluate endothelial dysfunction in a group of hospitalized patients with COPD using FMD. Patients were 40–80 years old and had more than 20 pack-year history of smoking. The authors demonstrated that patients hospitalized for exacerbation of COPD had severe vascular dysfunction as evidenced by markedly reduced FMD [120]. Since FMD is a valid test to evaluate possible changes in endothelial function in COPD patients, Clarenbach et al. [121] and his team conducted a clinical study using this diagnostic technique. They recruited patients between 45 and 75 years with very severe COPD. Thanks to this simple and inexpensive technique they found a relative annual 5.6% decrease in FMD in COPD patients indicating progressive vascular dysfunction. An annual reduction in lung function tends to be associated with reduced endothelial function over time [121].

    Another functional method to evaluate endothelial dysfunction is the venous occlusion plethysmography (VOP), which is a non-invasive diagnostic test that measures venous blood flow in body extremities, typically arm or leg. The test involves the use of a pneumatic cuff that is placed around the body’s extremity and inflated to temporarily stop blood flow. By measuring changes in the volume of the extremity before, during, and after occlusion, VOP can provide information about the ability of the veins to carry blood back to the heart. Thanks to VOP measures, Yang et al. [122] demonstrated that patients with COPD and overweight smokers have impaired endothelial function [122]. However, this technique has important limitations as shown in a study performed by Maclay et al. [123] where endothelium-dependent and endothelium-independent vasodilation, assessed with VOP, did not significantly differentiate COPD patients from healthy controls.

    An additional technique is peripheral arterial tonometry (PAT), a non-invasive diagnostic test that measures changes in blood volume in fingers or toes. It is used to assess arterial integrity and to determine CVD risk. Results of the PAT test are used to calculate the PAT index, which is a measure of arteries’ ability to dilate in response to increased blood flow. A low PAT index is associated with an increased risk of CVD. Also, in this case, the literature shows conflicting results about the utility of this test to diagnose endothelial dysfunction [124].

    More recent, noninvasive technologies are available to evaluate endothelial damage such as nailfold capillaroscopy, laser speckle contrast imaging, or near-infrared spectroscopy, and can be combined with several reactivity tests and exercises. Currently, these technologies are used in other pathological states and no data are reported about their use in COPD patients. Since endothelium participates in the development and progression of COPD, it would be relevant to expand the clinical utility of diagnostic techniques to improve diagnostic and therapeutic approaches.

    Conclusions

    CVD is one of the most common risks of morbidity and mortality in COPD. Different studies suggest that endothelial dysfunction is present in a remarkable number of COPD patients and COPD contributes to increasing the cardiovascular risk. The typical COPD patient is just as likely to die from both respiratory and cardiovascular causes [125]. Although these two pathologies undoubtedly recognize CS as the common and principal risk factor, today this statement is restrictive. Indeed, other shared risk factors include physical inactivity, air pollution exposure, diabetes, and hypertension. Those factors are highly prevalent in COPD patients, especially in those who present severe disease [126]. Inflammation, aging, oxidative stress, and hypoxia are the most important mechanisms that can explain the relationship between these pathologies. At the basis of each of them, can certainly found systemic inflammation, foam cell production, and increased production of cell adhesion molecules in ECs, which contribute to the progression of CVD. These events also induce pulmonary vascular remodeling, intimal thickening, and endothelial dysfunction of the pulmonary artery that could be aggravated by the increased presence of inflammatory cells [127]. Accordingly, a review by Ye and others [128] suggests that patients with COPD have increased levels of endothelial dysfunction as well as increased incidence of CVD documented by intima-medial carotid thickening (this finding is reported in 22 different studies). Based on scientific evidence COPD and CVD are closely related and the key shared mechanism is likely inflammation. Considering the clear relationship between COPD and CVD, future studies are needed to deepen our understanding of this aspect to improve the quality of life of patients affected by these diseases.

    Accumulated evidence from different works suggests that endothelial dysfunction occurs in COPD and worsens with it. This phenomenon is driven by multiple factors: CS, chemokines, cytokines, ROS, and neutrophils activation. All these factors converge in a common activation of systemic inflammation, which contributes to disease progression and to the onset of other pathologic states such as CVD. To date, there are several tests and biomarkers that allow the evaluation of endothelial dysfunction and damage. However, the mechanisms leading to endothelial damage remain to be clarified, prior to obtain promising new treatment strategies and to understand whether the endothelial injury is primarily involved in the pathology or reflects the consequences of COPD occurrence.

    Abbreviations

    AAT:

    alpha-1 antitrypsin

    CCL11:

    eotaxin-1

    CGRP:

    calcitonin gene-related peptide

    COPD:

    chronic obstructive pulmonary disease

    CS:

    cigarette smoke

    CSE:

    cigarette smoke extract

    CVD:

    cardiovascular disease

    ECs:

    endothelial cells

    FEV1:

    forced expiratory volume in 1 s

    FMD:

    flow-mediated dilatation

    GOLD:

    Global Initiative for Chronic Obstructive Lung Disease

    HIF-1α:

    hypoxia-inducible factor 1α

    ICAM-1:

    intracellular adhesion molecule-1

    ICS:

    inhaled corticosteroids

    IL:

    interleukin

    MAC-1:

    macrophage-1 antigen

    NK:

    neurokinin receptors

    NLRP3:

    Nod-like receptor pyrin domain-containing protein 3

    NO:

    nitric oxide

    PAT:

    peripheral arterial tonometry

    ROS:

    reactive oxygen species

    TEM:

    transendothelial migration

    VEGF:

    vascular endothelial growth factors

    VOP:

    venous occlusion plethysmography

    Declarations

    Author contributions

    SS: Conceptualization, Writing—original draft. GN: Writing—review & editing. AMN: Writing—review & editing. MT: Writing—review & editing. CAML: Writing—review & editing. SP: Conceptualization, Writing—review & editing.

    Conflicts of interest

    GN, SP, and MT are employees of Chiesi Farmaceutici S.p.A. All the remaining authors have not actual and perceived conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Petty TL. Definitions in chronic obstructive pulmonary disease. Clin Chest Med. 1990;11:36373. [PubMed]
    Tanner L, Single AB. Animal models reflecting chronic obstructive pulmonary disease and related respiratory disorders: translating pre-clinical data into clinical relevance. J Innate Immun. 2020;12:20325. [DOI] [PubMed] [PMC]
    Martinez FJ, Donohue JF, Rennard SI. The future of chronic obstructive pulmonary disease treatment—difficulties of and barriers to drug development. Lancet. 2011;378:102737. [DOI] [PubMed]
    GBD 2015 Mortality and Causes of Death Collaborators. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388:1459544. [DOI] [PubMed] [PMC]
    Vij N, Chandramani-Shivalingappa P, Van Westphal C, Hole R, Bodas M. Cigarette smoke-induced autophagy impairment accelerates lung aging, COPD-emphysema exacerbations and pathogenesis. Am J Physiol Cell Physiol. 2018;314:C7387. [DOI] [PubMed] [PMC]
    Stoller JK, Aboussouan LS. α1-antitrypsin deficiency. Lancet. 2005;365:222536. [DOI] [PubMed]
    Hunninghake GM, Cho MH, Tesfaigzi Y, Soto-Quiros ME, Avila L, Lasky-Su J, et al. MMP12, lung function, and COPD in high-risk populations. N Engl J Med. 2009;361:2599608. [DOI] [PubMed] [PMC]
    Ding Z, Wang K, Li J, Tan Q, Tan W, Guo G. Association between glutathione S-transferase gene M1 and T1 polymorphisms and chronic obstructive pulmonary disease risk: a meta-analysis. Clin Genet. 2019;95:5362. [DOI] [PubMed]
    Huang X, Mu X, Deng L, Fu A, Pu E, Tang T, et al. The etiologic origins for chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2019;14:113958. [DOI] [PubMed] [PMC]
    Moschino L, Bonadies L, Baraldi E. Lung growth and pulmonary function after prematurity and bronchopulmonary dysplasia. Pediatr Pulmonol. 2021;56:3499508. [DOI] [PubMed] [PMC]
    Sheikh K, Coxson HO, Parraga G. This is what COPD looks like. Respirology. 2016;21:22436. [DOI] [PubMed]
    Vogelmeier CF, Román-Rodríguez M, Singh D, Han MK, Rodríguez-Roisin R, Ferguson GT. Goals of COPD treatment: focus on symptoms and exacerbations. Respir Med. 2020;166:105938. [DOI] [PubMed]
    Soler-Cataluña JJ, Martínez-García MA, Román Sánchez P, Salcedo E, Navarro M, Ochando R. Severe acute exacerbations and mortality in patients with chronic obstructive pulmonary disease. Thorax. 2005;60:92531. [DOI] [PubMed] [PMC]
    Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease: the GOLD science committee report 2019. Eur Respir J. 2019;53:1900164. [DOI] [PubMed]
    Barnes PJ, Celli BR. Systemic manifestations and comorbidities of COPD. Eur Respir J. 2009;33:116585. [DOI] [PubMed] [PMC]
    Rabe KF, Hurst JR, Suissa S. Cardiovascular disease and COPD: dangerous liaisons? Eur Respir Rev. 2018;27:180057. [DOI] [PubMed] [PMC]
    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. [DOI] [PubMed] [PMC]
    Brightling C, Greening N. Airway inflammation in COPD: progress to precision medicine. Eur Respir J. 2019;54:1900651. [DOI] [PubMed]
    Strzelak A, Ratajczak A, Adamiec A, Feleszko W. Tobacco smoke induces and alters immune responses in the lung triggering inflammation, allergy, asthma and other lung diseases: a mechanistic review. Int J Environ Res Public Health. 2018;15:1033. [DOI] [PubMed] [PMC]
    Rouadi PW, Idriss SA, Naclerio RM, Peden DB, Ansotegui IJ, Canonica GW, et al. Immunopathological features of air pollution and its impact on inflammatory airway diseases (IAD). World Allergy Organ J. 2020;13:100467. [DOI] [PubMed] [PMC]
    Gelfand EW, Joetham A, Wang M, Takeda K, Schedel M. Spectrum of T-lymphocyte activities regulating allergic lung inflammation. Immunol Rev. 2017;278:6386. [DOI] [PubMed] [PMC]
    Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med. 2020;217:e20190103. [DOI] [PubMed] [PMC]
    Price DT, Loscalzo J. Cellular adhesion molecules and atherogenesis. Am J Med. 1999;107:8597.
    Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2016;138:1627. [DOI] [PubMed]
    David B, Bafadhel M, Koenderman L, De Soyza A. Eosinophilic inflammation in COPD: from an inflammatory marker to a treatable trait. Thorax. 2021;76:18895. [DOI] [PubMed] [PMC]
    Mycroft K, Krenke R, Górska K. Eosinophils in COPD-current concepts and clinical implications. J Allergy Clin Immunol Pract. 2020;8:256574. [DOI] [PubMed]
    Chatila WM, Thomashow BM, Minai OA, Criner GJ, Make BJ. Comorbidities in chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2008;5:54955. [DOI] [PubMed] [PMC]
    Westerik JA, Metting EI, van Boven JF, Tiersma W, Kocks JW, Schermer TR. Associations between chronic comorbidity and exacerbation risk in primary care patients with COPD. Respir Res. 2017;18:31. [DOI] [PubMed] [PMC]
    McGarvey LP, John M, Anderson JA, Zvarich M, Wise RA; TORCH Clinical Endpoint Committee. Ascertainment of cause-specific mortality in COPD: operations of the TORCH Clinical Endpoint Committee. Thorax. 2007;62:4115. [DOI] [PubMed] [PMC]
    André S, Conde B, Fragoso E, Boléo-Tomé JP, Areias V, Cardoso J; GI DPOC-Grupo de Interesse na Doença Pulmonar Obstrutiva Crónica. COPD and cardiovascular disease. Pulmonology. 2019;25:16876. [DOI] [PubMed]
    Austin V, Crack PJ, Bozinovski S, Miller AA, Vlahos R. COPD and stroke: are systemic inflammation and oxidative stress the missing links? Clin Sci (Lond). 2016;130:103950. [DOI] [PubMed] [PMC]
    Morgan AD, Zakeri R, Quint JK. Defining the relationship between COPD and CVD: What are the implications for clinical practice? Ther Adv Respir Dis. 2018;12:1753465817750524. [DOI] [PubMed] [PMC]
    Theodorakopoulou MP, Bakaloudi DR, Alexandrou ME, Papakosta D, Pataka A, Kioumis I, et al. Endothelial dysfunction during acute exacerbations of chronic obstructive pulmonary disease: a systematic review and meta-analysis. COPD. 2021;18:24653. [DOI] [PubMed]
    Goldenberg NM, Kuebler WM. Endothelial cell regulation of pulmonary vascular tone, inflammation, and coagulation. Compr Physiol. 2015;5:53159. [DOI] [PubMed]
    Green CE, Turner AM. The role of the endothelium in asthma and chronic obstructive pulmonary disease (COPD). Respir Res. 2017;18:20. [DOI] [PubMed] [PMC]
    Kutcher ME, Herman IM. The pericyte: cellular regulator of microvascular blood flow. Microvasc Res. 2009;77:23546. [DOI] [PubMed] [PMC]
    Szucs B, Szucs C, Petrekanits M, Varga JT. Molecular characteristics and treatment of endothelial dysfunction in patients with COPD: a review article. Int J Mol Sci. 2019;20:4329. [DOI] [PubMed] [PMC]
    Aird WC. Phenotypic heterogeneity of the endothelium: I. structure, function, and mechanisms. Circ Res. 2007;100:15873. [DOI] [PubMed]
    Laniado-Laborín R. Smoking and chronic obstructive pulmonary disease (COPD). Parallel epidemics of the 21st century. Int J Environ Res Public Health. 2009;6:20924. [DOI] [PubMed]
    Huertas A, Guignabert C, Barberà JA, Bärtsch P, Bhattacharya J, Bhattacharya S, et al. Pulmonary vascular endothelium: the orchestra conductor in respiratory diseases: highlights from basic research to therapy. Eur Respir J. 2018;51:1700745. [DOI] [PubMed]
    Liebow AA. Pulmonary emphysema with special reference to vascular changes. Am Rev Respir Dis. 1959;80:6793. [DOI] [PubMed]
    Dinh-Xuan AT, Pepke-Zaba J, Butt AY, Cremona G, Higenbottam TW. Impairment of pulmonary-artery endothelium-dependent relaxation in chronic obstructive lung disease is not due to dysfunction of endothelial cell membrane receptors nor to L-arginine deficiency. Br J Pharmacol. 1993;109:58791. [DOI] [PubMed] [PMC]
    Peinado VI, Barbera JA, Ramirez J, Gomez FP, Roca J, Jover L, et al. Endothelial dysfunction in pulmonary arteries of patients with mild COPD. Am J Physiol. 1998;274:L90813. [DOI] [PubMed]
    Washko GR. The role and potential of imaging in COPD. Med Clin North Am. 2012;96:72943. [DOI] [PubMed] [PMC]
    Gane J, Stockley R. Mechanisms of neutrophil transmigration across the vascular endothelium in COPD. Thorax. 2012;67:55361. [DOI] [PubMed]
    Giavazzi R, Nicoletti MI, Chirivi RG, Hemingway I, Bernasconi S, Allavena P, et al. Soluble intercellular adhesion molecule-1 (ICAM-1) is released into the serum and ascites of human ovarian carcinoma patients and in nude mice bearing tumour xenografts. Eur J Cancer. 1994;30A:186570. [DOI] [PubMed]
    Woolhouse IS, Bayley DL, Lalor P, Adams DH, Stockley RA. Endothelial interactions of neutrophils under flow in chronic obstructive pulmonary disease. Eur Respir J. 2005;25:6127. [DOI] [PubMed]
    Sorkness RL, Mehta H, Kaplan MR, Miyasaka M, Hefle SL, Lemanske RF Jr. Effect of ICAM-1 blockade on lung inflammation and physiology during acute viral bronchiolitis in rats. Pediatr Res. 2000;47:81924. [DOI] [PubMed]
    Johnston NW, Olsson M, Edsbäcker S, Gerhardsson de Verdier M, Gustafson P, McCrae C, et al. Colds as predictors of the onset and severity of COPD exacerbations. Int J Chron Obstruct Pulmon Dis. 2017;12:83948. [DOI] [PubMed] [PMC]
    Shukla SD, Hansbro PM, Walters EH. Blocking rhinoviral adhesion molecule (ICAM-1): potential to prevent COPD exacerbations. Int J Chron Obstruct Pulmon Dis. 2017;12:14134. [DOI] [PubMed] [PMC]
    Riise GC, Larsson S, Löfdahl CG, Andersson BA. Circulating cell adhesion molecules in bronchial lavage and serum in COPD patients with chronic bronchitis. Eur Respir J. 1994;7:16737. [DOI] [PubMed]
    Garcia-Rio F, Miravitlles M, Soriano JB, Muñoz L, Duran-Tauleria E, Sánchez G, et al.; EPI-SCAN Steering Committee. Systemic inflammation in chronic obstructive pulmonary disease: a population-based study. Respir Res. 2010;11:63. [DOI] [PubMed] [PMC]
    Faner R, Tal-Singer R, Riley JH, Celli B, Vestbo J, MacNee W, et al.; ECLIPSE Study Investigators. Lessons from ECLIPSE: a review of COPD biomarkers. Thorax. 2014;69:66672. [DOI] [PubMed]
    Wang Z, Locantore N, Haldar K, Ramsheh MY, Beech AS, Ma W, et al. Inflammatory endotype-associated airway microbiome in chronic obstructive pulmonary disease clinical stability and exacerbations: a multicohort longitudinal analysis. Am J Respir Crit Care Med. 2021;203:1488502. [DOI] [PubMed] [PMC]
    Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 Report. GOLD executive summary. Am J Respir Crit Care Med. 2017;195:55782. [DOI] [PubMed]
    Pantazopoulos I, Magounaki K, Kotsiou O, Rouka E, Perlikos F, Kakavas S, et al. Incorporating biomarkers in COPD management: the research keeps going. J Pers Med. 2022;12:379. [DOI] [PubMed] [PMC]
    Heiman AS, Abonyo BO, Darling-Reed SF, Alexander MS. Cytokine-stimulated human lung alveolar epithelial cells release eotaxin-2 (CCL24) and eotaxin-3 (CCL26). J Interferon Cytokine Res. 2005;25:8291. [DOI] [PubMed]
    Henrot P, Prevel R, Berger P, Dupin I. Chemokines in COPD: from implication to therapeutic use. Int J Mol Sci. 2019;20:2785. [DOI] [PubMed] [PMC]
    Ivanovska M, Abdi Z, Murdjeva M, Macedo D, Maes A, Maes M. CCL-11 or eotaxin-1: an immune marker for ageing and accelerated ageing in neuro-psychiatric disorders. Pharmaceuticals (Basel). 2020;13:230. [DOI] [PubMed] [PMC]
    Wang LY, Tu YF, Lin YC, Huang CC. CXCL5 signaling is a shared pathway of neuroinflammation and blood-brain barrier injury contributing to white matter injury in the immature brain. J Neuroinflammation. 2016;13:6. [DOI] [PubMed] [PMC]
    Miyake M, Goodison S, Urquidi V, Gomes Giacoia E, Rosser CJ. Expression of CXCL1 in human endothelial cells induces angiogenesis through the CXCR2 receptor and the ERK1/2 and EGF pathways. Lab Invest. 2013;93:76878. [DOI] [PubMed]
    Pavord ID, Lettis S, Locantore N, Pascoe S, Jones PW, Wedzicha JA, et al. Blood eosinophils and inhaled corticosteroid/long-acting β-2 agonist efficacy in COPD. Thorax. 2016;71:11825. [DOI] [PubMed] [PMC]
    Singh D, Kolsum U, Brightling CE, Locantore N, Agusti A, Tal-Singer R; ECLIPSE investigators. Eosinophilic inflammation in COPD: prevalence and clinical characteristics. Eur Respir J. 2014;44:1697700. [DOI] [PubMed]
    Siddiqui SH, Guasconi A, Vestbo J, Jones P, Agusti A, Paggiaro P, et al. Blood eosinophils: a biomarker of response to extrafine beclomethasone/formoterol in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2015;192:5235. [DOI] [PubMed] [PMC]
    Aaron SD, Vandemheen KL, Maltais F, Field SK, Sin DD, Bourbeau J, et al. TNFα antagonists for acute exacerbations of COPD: a randomised double-blind controlled trial. Thorax. 2013;68:1428. [DOI] [PubMed]
    Pisi G, Olivieri D, Chetta A. The airway neurogenic inflammation: clinical and pharmacological implications. Inflamm Allergy Drug Targets. 2009;8:17681. [DOI] [PubMed]
    Walsh DA, McWilliams DF. Tachykinins and the cardiovascular system. Curr Drug Targets. 2006;7:103142. [DOI] [PubMed]
    Barnes PJ. Neurogenic inflammation in the airways. Respir Physiol. 2001;125:14554. [DOI] [PubMed]
    Groneberg DA, Quarcoo D, Frossard N, Fischer A. Neurogenic mechanisms in bronchial inflammatory diseases. Allergy. 2004;59:113952. [DOI] [PubMed]
    Sin DD, Anthonisen NR, Soriano JB, Agusti AG. Mortality in COPD: role of comorbidities. Eur Respir J. 2006;28:124557. [DOI] [PubMed]
    Boukhenouna S, Wilson MA, Bahmed K, Kosmider B. Reactive oxygen species in chronic obstructive pulmonary disease. Oxid Med Cell Longev. 2018;2018:5730395. [DOI] [PubMed] [PMC]
    Pryor WA, Stone K. Oxidants in cigarette smoke radicals, hydrogen peroxide, peroxynitrate, and peroxynitrite. Ann N Y Acad Sci. 1993;686:1227. [DOI] [PubMed]
    Yang SR, Chida AS, Bauter MR, Shafiq N, Seweryniak K, Maggirwar SB, et al. Cigarette smoke induces proinflammatory cytokine release by activation of NF-kappaB and posttranslational modifications of histone deacetylase in macrophages. Am J Physiol Lung Cell Mol Physiol. 2006;291:L4657. [DOI] [PubMed]
    Zuo L, He F, Sergakis GG, Koozehchian MS, Stimpfl JN, Rong Y, et al. Interrelated role of cigarette smoking, oxidative stress, and immune response in COPD and corresponding treatments. Am J Physiol Lung Cell Mol Physiol. 2014;307:L20518. [DOI] [PubMed]
    Walter DH, Rittig K, Bahlmann FH, Kirchmair R, Silver M, Murayama T, et al. Statin therapy accelerates reendothelialization: a novel effect involving mobilization and incorporation of bone marrow-derived endothelial progenitor cells. Circulation. 2002;105:301724. [DOI] [PubMed]
    Barnes PJ. Oxidative stress-based therapeutics in COPD. Redox Biol. 2020;33:101544. [DOI] [PubMed] [PMC]
    Tejero J, Shiva S, Gladwin MT. Sources of vascular nitric oxide and reactive oxygen species and their regulation. Physiol Rev. 2019;99:31179. [DOI] [PubMed] [PMC]
    Zheng D, Liu J, Piao H, Zhu Z, Wei R, Liu K. ROS-triggered endothelial cell death mechanisms: focus on pyroptosis, parthanatos, and ferroptosis. Front Immunol. 2022;13:1039241. [DOI] [PubMed] [PMC]
    Lv H, Zhen C, Liu J, Yang P, Hu L, Shang P. Unraveling the potential role of glutathione in multiple forms of cell death in cancer therapy. Oxid Med Cell Longev. 2019;2019:3150145. [DOI] [PubMed] [PMC]
    Qin X, Zhang J, Wang B, Xu G, Yang X, Zou Z, et al. Ferritinophagy is involved in the zinc oxide nanoparticles-induced ferroptosis of vascular endothelial cells. Autophagy. 2021;17:426685. [DOI] [PubMed] [PMC]
    Luo EF, Li HX, Qin YH, Qiao Y, Yan GL, Yao YY, et al. Role of ferroptosis in the process of diabetes-induced endothelial dysfunction. World J Diabetes. 2021;12:12437. [DOI] [PubMed] [PMC]
    Sheng S, Xu J, Liang Q, Hong L, Zhang L. Astragaloside IV inhibits bleomycin-induced ferroptosis in human umbilical vein endothelial cells by mediating LPC. Oxid Med Cell Longev. 2021;2021:6241242. [DOI] [PubMed] [PMC]
    Wang Y, Kuang X, Yin Y, Han N, Chang L, Wang H, et al. Tongxinluo prevents chronic obstructive pulmonary disease complicated with atherosclerosis by inhibiting ferroptosis and protecting against pulmonary microvascular barrier dysfunction. Biomed Pharmacother. 2022;145:112367. [DOI] [PubMed]
    Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19:47789. [DOI] [PubMed] [PMC]
    Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Signal. 2015;22:111129. [DOI] [PMC]
    Wu Q, He X, Wu LM, Zhang RY, Li LM, Wu CM, et al. MLKL aggravates Ox-LDL-induced cell pyroptosis via activation of NLRP3 inflammasome in human umbilical vein endothelial cells. Inflammation. 2020;43:222231. [DOI] [PubMed]
    Hang L, Peng Y, Xiang R, Li X, Li Z. Ox-LDL causes endothelial cell injury through ASK1/NLRP3-mediated inflammasome activation via endoplasmic reticulum stress. Drug Des Devel Ther. 2020;14:73144. [DOI] [PubMed] [PMC]
    Zhang Y, Chen Y, Zhang Y, Li PL, Li X. Contribution of cathepsin B-dependent Nlrp3 inflammasome activation to nicotine-induced endothelial barrier dysfunction. Eur J Pharmacol. 2019;865:172795. [DOI] [PubMed] [PMC]
    Cau SB, Bruder-Nascimento A, Silva MB, Ramalho FNZ, Mestriner F, Alves-Lopes R, et al. Angiotensin-II activates vascular inflammasome and induces vascular damage. Vascul Pharmacol. 2021;139:106881. [DOI] [PubMed] [PMC]
    Pouwels SD, Zijlstra GJ, van der Toorn M, Hesse L, Gras R, Ten Hacken NH, et al. Cigarette smoke-induced necroptosis and DAMP release trigger neutrophilic airway inflammation in mice. Am J Physiol Lung Cell Mol Physiol. 2016;310:L37786. [DOI] [PubMed]
    Wang Y, Zhou JS, Xu XC, Li ZY, Chen HP, Ying SM, et al. Endoplasmic reticulum chaperone GRP78 mediates cigarette smoke-induced necroptosis and injury in bronchial epithelium. Int J Chron Obstruct Pulmon Dis. 2018;13:57181. [DOI] [PubMed] [PMC]
    Mizumura K, Maruoka S, Shimizu T, Gon Y. Autophagy, selective autophagy, and necroptosis in COPD. Int J Chron Obstruct Pulmon Dis. 2018;13:316572. [DOI] [PubMed] [PMC]
    Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123:96672. [DOI] [PubMed] [PMC]
    Amsellem V, Gary-Bobo G, Marcos E, Maitre B, Chaar V, Validire P, et al. Telomere dysfunction causes sustained inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2011;184:135866. [DOI] [PubMed]
    Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence in patients with pulmonary emphysema. Am J Respir Crit Care Med. 2006;174:88693. [DOI] [PubMed]
    Rutten EP, Gopal P, Wouters EF, Franssen FM, Hageman GJ, Vanfleteren LE, et al. Various mechanistic pathways representing the aging process are altered in COPD. Chest. 2016;149:5361. [DOI] [PubMed]
    Birch J, Anderson RK, Correia-Melo C, Jurk D, Hewitt G, Marques FM, et al. DNA damage response at telomeres contributes to lung aging and chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2015;309:L112437. [DOI] [PubMed] [PMC]
    Borgas D, Chambers E, Newton J, Ko J, Rivera S, Rounds S, et al. Cigarette smoke disrupted lung endothelial barrier integrity and increased susceptibility to acute lung injury via histone deacetylase 6. Am J Respir Cell Mol Biol. 2016;54:68396. [DOI] [PubMed] [PMC]
    Ferrer E, Peinado VI, Castañeda J, Prieto-Lloret J, Olea E, González-Martín MC, et al. Effects of cigarette smoke and hypoxia on pulmonary circulation in the guinea pig. Eur Respir J. 2011;38:61727. [DOI] [PubMed]
    Paschalaki K, Rossios C, Pericleous C, MacLeod M, Rothery S, Donaldson GC, et al. Inhaled corticosteroids reduce senescence in endothelial progenitor cells from patients with COPD. Thorax. 2022;77:61620. [DOI] [PubMed] [PMC]
    Zeng M, Zhang X, Xing W, Wang Q, Liang G, He Z. Cigarette smoke extract mediates cell premature senescence in chronic obstructive pulmonary disease patients by up-regulating USP7 to activate p300-p53/p21 pathway. Toxicol Lett. 2022;359:3145. [DOI] [PubMed]
    Rivas M, Gupta G, Costanzo L, Ahmed H, Wyman AE, Geraghty P. Senescence: pathogenic driver in chronic obstructive pulmonary disease. Medicina (Kaunas). 2022;58:817. [DOI] [PubMed] [PMC]
    Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF. Endothelial cell death and decreased expression of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 in emphysema. Am J Respir Crit Care Med. 2001;163:73744. [DOI] [PubMed]
    Zeng H, Kong X, Zhang H, Chen Y, Cai S, Luo H, et al. Inhibiting DNA methylation alleviates cigarette smoke extract-induced dysregulation of Bcl-2 and endothelial apoptosis. Tob Induc Dis. 2020;18:51. [DOI] [PubMed] [PMC]
    Henson PM, Vandivier RW, Douglas IS. Cell death, remodeling, and repair in chronic obstructive pulmonary disease? Proc Am Thorac Soc. 2006;3:7137. [DOI] [PubMed] [PMC]
    Farid M, Kanaji N, Nakanishi M, Gunji Y, Michalski J, Iwasawa S, et al. Smad3 mediates cigarette smoke extract (CSE) induction of VEGF release by human fetal lung fibroblasts. Toxicol Lett. 2013;220:12634. [DOI] [PubMed]
    Taraseviciene-Stewart L, Douglas IS, Nana-Sinkam PS, Lee JD, Tuder RM, Nicolls MR, et al. Is alveolar destruction and emphysema in chronic obstructive pulmonary disease an immune disease? Proc Am Thorac Soc. 2006;3:68790. [DOI] [PubMed]
    Zhang C, Cai S, Chen P, Chen JB, Wu J, Wu SJ. Inhibition of tumor necrosis factor-alpha reduces alveolar septal cell apoptosis in passive smoking rats. Chin Med J. 2008;121:597601. [PubMed]
    Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S, Hirth PK, et al. Inhibition of VEGF receptors causes lung cell apoptosis and emphysema. J Clin Invest. 2000;106:13119. [DOI] [PubMed] [PMC]
    Segura-Valdez L, Pardo A, Gaxiola M, Uhal BD, Becerril C, Selman M. Upregulation of gelatinases A and B, collagenases 1 and 2, and increased parenchymal cell death in COPD. Chest. 2000;117:68494. [DOI] [PubMed]
    Yasuo M, Mizuno S, Kraskauskas D, Bogaard HJ, Natarajan R, Cool CD, et al. Hypoxia inducible factor-1α in human emphysema lung tissue. Eur Respir J. 2011;37:77583. [DOI] [PubMed]
    Lee SH, Lee SH, Kim CH, Yang KS, Lee EJ, Min KH, et al. Increased expression of vascular endothelial growth factor and hypoxia inducible factor-1α in lung tissue of patients with chronic bronchitis. Clin Biochem. 2014;47:5529. [DOI] [PubMed]
    Noe J, Petrusca D, Rush N, Deng P, VanDemark M, Berdyshev E, et al. CFTR regulation of intracellular pH and ceramides is required for lung endothelial cell apoptosis. Am J Respir Cell Mol Biol. 2009;41:31423. [DOI] [PubMed] [PMC]
    Petrache I, Fijalkowska I, Medler TR, Skirball J, Cruz P, Zhen L, et al. α-1 antitrypsin inhibits caspase-3 activity, preventing lung endothelial cell apoptosis. Am J Pathol. 2006;169:115566. [DOI] [PubMed] [PMC]
    Rodriguez-Miguelez P, Seigler N, Bass L, Dillard TA, Harris RA. Assessments of endothelial function and arterial stiffness are reproducible in patients with COPD. Int J Chron Obstruct Pulmon Dis. 2015;10:197786. [DOI] [PubMed] [PMC]
    Flammer AJ, Anderson T, Celermajer DS, Creager MA, Deanfield J, Ganz P, et al. The assessment of endothelial function: from research into clinical practice. Circulation. 2012;126:75367. [DOI] [PubMed] [PMC]
    Barr RG, Mesia-Vela S, Austin JH, Basner RC, Keller BM, Reeves AP, et al. Impaired flow-mediated dilation is associated with low pulmonary function and emphysema in ex-smokers: the Emphysema and Cancer Action Project (EMCAP) Study. Am J Respir Crit Care Med. 2007;176:12007. [DOI] [PubMed] [PMC]
    Eickhoff P, Valipour A, Kiss D, Schreder M, Cekici L, Geyer K, et al. Determinants of systemic vascular function in patients with stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;178:12118. [DOI] [PubMed]
    Moro L, Pedone C, Scarlata S, Malafarina V, Fimognari F, Antonelli-Incalzi R. Endothelial dysfunction in chronic obstructive pulmonary disease. Angiology. 2008;59:35764. [DOI] [PubMed]
    Marchetti N, Ciccolella DE, Jacobs MR, Crookshank A, Gaughan JP, Kashem MA, et al. Hospitalized acute exacerbation of COPD impairs flow and nitroglycerin-mediated peripheral vascular dilation. COPD. 2011;8:605. [DOI] [PubMed]
    Clarenbach CF, Sievi NA, Kohler M. Annual progression of endothelial dysfunction in patients with COPD. Respir Med. 2017;132:1520. [DOI] [PubMed]
    Yang L, Mäki-Petäjä K, Cheriyan J, McEniery C, Wilkinson IB. The role of epoxyeicosatrienoic acids in the cardiovascular system. Br J Clin Pharmacol. 2015;80:2844. [DOI] [PubMed] [PMC]
    Maclay JD, McAllister DA, Mills NL, Paterson FP, Ludlam CA, Drost EM, et al. Vascular dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:51320. [DOI] [PubMed]
    Theodorakopoulou MP, Bakaloudi DR, Dipla K, Zafeiridis A, Boutou AK. Vascular endothelial damage in COPD: current functional assessment methods and future perspectives. Expert Rev Respir Med. 2021;15:112133. [DOI] [PubMed]
    Berry CE, Wise RA. Mortality in COPD: causes, risk factors, and prevention. COPD. 2010;7:37582. [DOI] [PubMed] [PMC]
    Mannino DM, Thorn D, Swensen A, Holguin F. Prevalence and outcomes of diabetes, hypertension and cardiovascular disease in COPD. Eur Respir J. 2008;32:9629. [DOI] [PubMed]
    Yang Q, Underwood MJ, Hsin MK, Liu XC, He GW. Dysfunction of pulmonary vascular endothelium in chronic obstructive pulmonary disease: basic considerations for future drug development. Curr Drug Metab. 2008;9:6617. [DOI] [PubMed]
    Ye C, Younus A, Malik R, Roberson L, Shaharyar S, Veledar E, et al. Subclinical cardiovascular disease in patients with chronic obstructive pulmonary disease: a systematic review. QJM. 2017;110:3419. [DOI] [PubMed]