Open access peer-reviewed chapter

Endothelial Dysfunction and Disruption in Pulmonary Hypertension

Written By

Rajamma Mathew

Submitted: 02 May 2019 Reviewed: 18 March 2020 Published: 04 May 2020

DOI: 10.5772/intechopen.92177

From the Edited Volume

Cardiovascular Risk Factors in Pathology

Edited by Alaeddin Abukabda, Maria Suciu and Minodora Andor

Chapter metrics overview

873 Chapter Downloads

View Full Metrics

Abstract

A number of systemic diseases lead to pulmonary hypertension (PH), a serious disorder with a high morbidity and mortality rate. Irrespective of the underlying disease, endothelial dysfunction or disruption plays a key role in the initiation and progression of PH. Endothelial dysfunction and disruption result in impaired vascular relaxation response, activation of proliferative pathways leading to medial hypertrophy and PH. Endothelial cells (EC) play a crucial role in regulating vascular tone and maintaining homeostasis. Caveolin-1, a 21-22 kD membrane protein, interacts with a number of transducing factors and maintains them in a negative conformation. Disruption of EC results in endothelial caveolin-1 loss and reciprocal activation of proliferative pathways leading to PH, and the accompanying loss of PECAM1 and vascular endothelial cadherin results in barrier dysfunction. These changes lead to the irreversibility of PH. Hypoxia-induced PH is not accompanied by endothelial disruption or caveolin-1 loss but is associated with caveolin-1 dysfunction and the activation of proliferative pathways. Removal of hypoxic exposure results in the reversal of the disease. Thus, EC integrity is an important factor that determines irreversibility vs. reversibility of PH. This chapter will discuss normal EC function and the differences encountered in PH following EC disruption and EC dysfunction.

Keywords

  • caveolin-1
  • endothelial cells
  • membrane integrity
  • smooth muscle cells
  • pulmonary hypertension

1. Pulmonary hypertension

A number of systemic diseases such as cardiopulmonary, infectious, inflammatory and autoimmune diseases, hematological disorders, drug toxicity and several genetic mutations lead to pulmonary hypertension (PH), a devastating disease with a high morbidity and mortality rate. Based on clinical diagnosis, PH has been classified into five major groups that were updated in 2013 [1]. Group 1, labeled as pulmonary arterial hypertension (PAH), includes idiopathic and heritable PAH, PAH associated with congenital heart defect (CHD), connective tissue diseases, portal hypertension, HIV, schistosomiasis and drug-/toxin-induced PAH. In addition, mutation of several genes such as BMPRII (bone morphogenetic protein receptor type 2), CAV1 (caveolin-1), ENG (endoglin), SMAD9 (SMAD family member 9), ACVRL1 (activin A receptor like type 1) and KCNK3 (potassium two pore domain channel subfamily K member 3) are among the well-documented causes of PAH. Pulmonary veno-occlusive disease (PVOD)/pulmonary capillary hemangioma and persistent pulmonary hypertension of the newborn (PPHN) are included in Group 1 as subcategories 1′ and 1″, respectively. Recently, mutation of EIF2AK4 (eukaryotic translation initiation factor 2a kinase 4) has been shown to be associated with PVOD and capillary hemangioma [2]. Included in Group 2 are PH associated with congenital and acquired left heart diseases; Group 3 comprises PH due to lung diseases and/or hypoxia. Group 4 includes chronic thromboembolic pulmonary hypertension (CTEPH). PH associated with hematological disorders, myeloproliferative diseases, splenectomy and a number of miscellaneous systemic and metabolic disorders is included in Group 5. Up until recently, the diagnosis of PAH was considered when the mean pulmonary artery pressure (PAP) of ≥25 mmHg, pulmonary capillary wedge pressure of ≤ 15 mmHg, and a pulmonary vascular resistance (PVR) of > 3 Wood units were observed at rest. During the 6th World Symposium on PH, the mean PAP of threshold was lowered to >20 mmHg and PVR was maintained as >3 Wood units [3]. These changes are based on the evaluation of 47 studies from 13 countries, which showed that independent of age the normal mean PAP rarely exceeded 20 mmHg [4]. The worldwide prevalence of PH is estimated to be 1%, increasing to 10% in patients older than 65 years of age. Globally, left heart diseases (Gr 2) and lung diseases (Gr 3) are considered the most common causes of PH. About 80% of patients are from developing countries; the common causes of PH in these patients are CHD, rheumatic heart disease and infection such as HIV and schistosomiasis. These patients tend to be younger than 65 years [5]. With modern therapy, the survival in patients with idiopathic and heritable PAH and PAH associated with anorexigen drugs has improved to 92%, 75% and 66% at 1, 3 and 5 years, respectively [6]. However, the underlying vascular changes remain progressive [7]. There is still a significant delay between the onset of symptoms and the final diagnosis. A recent retrospective study revealed a delay of 3.9 years between the onset of symptoms and the diagnosis of idiopathic PAH [8]. Thus, by the time the diagnosis is made, patients often have significant pulmonary vascular disease, which is a serious challenge to therapy. Interestingly, in an animal model of PH, significant disruption of endothelial cells and the activation of pro-proliferative pathways have been shown to occur before the onset of PH, indicating that the vascular pathology is already present by the time the symptoms appear [9].

The major causes of PH in children are CHD, PPHN, PH associated with disruption of normal pulmonary vascular and alveolar development in preterm infants, and congenital defects associated with hypoplasia of the lungs [10, 11]. A significant number of pediatric patients (>80%) have transient PH. These include resolution of PPHN and the majority of CHD cases that become free of PAH after the surgical correction of the defect [12]. However, preterm birth itself has an increased risk of developing PH even after adjusting for known factors such as heart and lung diseases, congenital diaphragmatic hernia and chromosomal abnormalities [13]. Furthermore, poor outcome has been reported in children with BMPRII mutation associated with idiopathic or heritable PAH [14]. Mutation of TBX4 (T-box transcription factor 4 gene) is associated with skeletal, cardiac and neurologic defects. It also leads to a form of developmental lung disease that has been shown to be associated with severe PH during infancy and childhood [15]. It is worth noting that infants over the age of 2 years who had CHD exhibited increased pulmonary artery pressure and PVR even after surgical correction of the defect [16]. Pulmonary vascular lesions found in PAH associated with CHD are reported to be similar to what is found in idiopathic PAH [17]. However, the plexiform lesions in idiopathic PAH have monoclonal cell population, whereas Eisenmenger disease (PAH associated with CHD) displays polyclonal cells [18], indicating a distinct difference between two forms of PAH.

Endothelial cells (EC) play a key role in maintaining vascular homeostasis in response to various stimuli and regulate vascular tone, permeability, coagulation, inflammation through mediators such as nitric oxide (NO), endothelium-derived hyperpolarization factor (EDHF), endothelin-1 (ET1), cell adhesion molecules, cytokines and chemokines. Regardless of the underlying disease, endothelial dysfunction/disruption plays a key role in the pathogenesis of PH. The genetic and environmental factors act as an initial trigger leading to endothelial cell injury and impaired regeneration resulting in vascular remodeling and loss of small pulmonary arteries [19]. Endothelial dysfunction, impaired vascular dilatation, alterations in the expression of NO, ET1 and serotonin, increased expression of inflammatory cytokines and chemokines, loss of endothelial caveolin-1 and disordered proteolysis of extracellular matrix contribute to the pathogenesis of PAH [20, 21]. Increased expression of chemokines such as CX(3)C (fractalkine) and RANTES (CCL5) has been reported in PAH; importantly, both these chemokines are produced in EC [22, 23]. In sugen + hypoxia model (mice), the deletion of CCL5 resulted in reduction in PH via caveolin-1–dependent amplification of BMPR2 signaling. It stabilized surface caveolin-1, restored BMPR2 signaling and enhanced BMPR2 and caveolin-1 interaction [24]. This observation further supports the role for inflammation in PH. In addition, perivascular infiltration with inflammatory cells (T and B cells) is present in plexiform lesions [25, 26]. Increased expression of interleukin-1 (IL-1) and IL-6 occurs in human PAH and monocrotaline (MCT)-induced PH, and inhibition of IL-6 expression and bioactivity as a preventive measure results in the abrogation of MCT-induced PH [27, 28].

In addition to the imbalance of vasoactive mediators and vascular remodeling, abnormality in ion channels (Ca2+, K+) and growth factors such as VEGF, EGF, TGF beta, MMPs, BMPR2 and Notch1 has been implicated in pathophysiology of PAH, leading to vasoconstriction, abnormal remodeling and plexiform lesions [29]. Proliferative EC reveals increased expression of angiogenesis and survival-related molecules such as VEGF, VEGFR2, Hif-1 α, and 1β and reduced expression of p27/kip1. Signal transducer and activator of transcription (STAT3) is essential for cell proliferation and survival, and antiapoptotic function [30]. In the MCT model of PH, the loss of endothelial caveolin-1 was shown to be associated with reciprocal activation of STAT3 (PY-STAT3) and increased proliferating cell nuclear antigen (PCNA) [31]. Furthermore, EC in plexiform and concentric lesions exhibits increased expression of PY-STAT3 [32]. Importantly, the inhibition of STAT3 prevents neointima formation by inhibiting cell proliferation and promoting the apoptosis of neointimal SMC [33]. BMPRII mutations linked to PAH are associated with the activation of STAT3. Furthermore, BMPR2 deficiency promotes inflammatory response resulting in increased IL-6 levels and PY-STAT3 activation [34]. BMPR2, a cell surface receptor, is essential for differentiation and proliferation of EC and SMC. Without altering the BMPRII mRNA levels, miR-17/92 modulates BMPR2 protein levels. Importantly, IL-6 regulates the expression of miR-17/92 in human pulmonary arterial EC via STAT3 signaling. Persistent activation of STAT3 results in the upregulation of miR-20, which leads to the reduction in the expression of BMPR2 protein [35]. BMPR2 expression is decreased also in patients with heritable and idiopathic PAH, without associated mutation [36]. Importantly, levels of SMAD-specific E3 ubiquitin protein ligase 1 (Smurf1), a key negative regulator of BMPPR2, has been shown to be increased in hypoxia and MCT models of PH in rats [37]. Increased Smurf1 immunoreactivity has also been reported in EC and SMC in the explanted lungs from patients with PAH. Furthermore, Smurf1 deletion protects mice from sugen + hypoxia-induced PH [38]. Interestingly, elafin reverses obliterative changes in pulmonary arteries via elastase inhibition and caveolin-1–dependent amplification of BMPR2. In addition, elafin promotes angiogenesis via increasing interaction of BMPR2 and caveolin-1 via mediating stabilization of endothelial surface caveolin-1 [39].

Recent studies have shown the involvement of Notch1 signaling in PAH. Increased expression of Notch1 has been reported in the lungs of patients with IPAH and in rats with sugen + hypoxia-induced PH. Notch1 positively regulates EC proliferation by downregulating p21 and negatively regulating apoptosis via Bcl2 and survivin. In-vitro studies with human pulmonary arterial EC revealed increased expression of Notch1 during hypoxia exposure, and Notch1 downregulation decreased cell proliferation [40]. Furthermore, Notch1 under hypoxia contributes to increased proliferation, migration and survival in cancer cells [41]. Notch1 is essential for VEGF-induced proliferation, migration and survival of EC [42]. Thus, Notch1 plays a significant role in the pathogenesis of PH. However, Notch1 also plays a key role in vascular morphogenesis, EC quiescence, junction stability and vascular homeostasis. Reduction in Notch1 activity destabilizes cellular junction and triggers EC proliferation and results in the loss of arterial identity and incorporation of these cells into veins. Notch1 is sensitive to shear stress and it requires VEGFA and VEGFR2 for growth [43, 44]. Interestingly, Notch-mediated inhibition of proliferation requires phosphatase-tensin homolog (PTEN), a dual lipid/protein phosphatase. PTEN localization is cell cycle dependent, negatively regulates cell cycle progression and has a restrictive role on angiogenesis [45]. Recent studies have shown significant loss of PTEN concomitant with caveolin-1 dysfunction in hypoxia-induced PH [46]. Fibroblasts from idiopathic pulmonary fibrosis lungs exhibit low membrane PTEN associated with low membrane caveolin-1 levels, and overexpression of caveolin-1 restores membrane PTEN levels. PTEN contains a caveolin-1–binding motif and, in part, colocalizes in caveolae [47]. Thus, caveolin-1 expression determines the membrane PTEN levels through its binding sequence. Furthermore, PTEN has also been shown to negatively regulate STAT3 and its activation, and importantly, membrane localization of PTEN is considered responsible for the inactivation of STAT3 [48].

Advertisement

2. Endothelial cell function

EC forms a monolayer in contact with blood flow and mechanical forces and underlying SMC. It is a non-thrombogenic and a selective barrier to circulating macromolecules. Juxtaposition of EC and SMC facilitates cross talk, and EC maintains SMC in quiescent state. Myoendothelial gap junction plays an important role in Ca2+ exchange between EC and SMC. EC is crucial for delivery of O2 and nutrients to underlying organs. EC maintains a balance between vasodilatation and vasoconstriction, apoptosis and cell proliferation, participate in immune and metabolic function, and maintain anticoagulant state [21, 49]. In addition, EC converts mechanical information into biological responses through mechanotransduction processes. EC adapts to mechanical inputs while maintaining crucial vascular barrier function. Failure of EC to adapt to changes has effects on vascular permeability, an important cause of vascular diseases [50].

2.1 Caveolae, caveolin-1 and cavin-1

Caveolae, a subset of specialized lipid rafts (50–100 nm), first described in 1950s by Palade [51] and Yamada [52], is found on plasma membranes of a variety of cell types including EC, SMC, fibroblasts and adipocytes. Caveolae are non-clathrin–coated plasma membrane vesicles (50–100 nm) enriched in glycosphingolipids, cholesterol, sphingomyelin and lipid-anchored membrane proteins. They form an important signaling platform that compartmentalizes and integrates a number of signaling molecules and allows cross talk between different signaling pathways, and mediates and integrates signaling events at the cell surface. EC contains 5000–10,000 caveolae per cell [53]. In addition, caveolae act as plasma membrane sensors and respond to stress. Caveolae flatten in response to membrane stretch. The flattening is a protective mechanism; it buffers the membrane and prevents its rupture [54, 55]. Caveolin-1 is a major protein (21–22 kDa) constituent of caveolae that maintains the shape of caveolae; EC has the highest levels of caveolin-1 [56]. Caveolin-1 is involved in multiple cellular processes such as molecular transport, cell proliferation, adhesion, migration and signal transduction. Caveolin-1 has an integral role in endocytosis. However, overexpression of caveolin-1 inhibits endocytosis [57, 58]. Caveolin-1 is synthesized in endoplasmic reticulum and then transported to Golgi complex. During its biosynthesis, it is associated with lipid rafts and become detergent resistant. From a structural standpoint, caveolin-1 contains a hairpin loop structure and three palmitoylation sites and a scaffolding domain that facilitates interaction with the plasma membrane [59, 60]. Caveolin-1 functions through protein-protein interaction and regulates and stabilizes several proteins including Src family of kinases, G proteins (α-subunits), G protein-coupled receptors, H-Ras, PKC, endothelial NO synthase (eNOS), integrins, and growth factor receptors such as VEGFR2, EGFR and PDGFR in an inhibitory conformation. Importantly, a 20-amino acid membrane proximal region of the cytosolic amino-terminal domain, termed caveolin-scaffolding domain (residue 82–101), is sufficient to mediate these interactions [61, 62]. Caveolin-1 also functions as a suppressor of cytokine signaling (SOCS), the family of proteins that are upregulated by cytokines and that in turn inhibit cytokine signaling via modulating JAK-STAT pathway [63]. Caveolin-2 is present associated with caveolin-1 in all cell types. It requires caveolin-1 for its transport from Golgi body to the plasma membrane. Caveolin-2 is not necessary for caveolae formation or caveolar localization of caveolin-1, but the coexpression results in a more efficient formation of caveolae [64]. In the absence of caveolin-1, caveolin-2 is degraded, and the decreased expression of caveolin-2 promotes increased cell proliferation [65, 66]. Furthermore, caveolin-2 knockout mice display increased proliferation of endothelial cells, hyper-cellular lung parenchyma and cell cycle progression [67].

In addition to caveolin-1, caveolae require polymerase 1 and transcript release factor (PTRF) also known as cavin-1. It is an essential component of caveolae; it regulates membrane curvature by stabilizing caveolin-1 in caveolae. The loss of cavin-1 results in the loss of caveolae and the release of caveolin-1 into the plasma membrane. Importantly, caveolin-1 is required for cavin-1 recruitment to plasma membrane [68, 69]. Loss of caveolin-1 is accompanied by a marked loss of caveolin-2 and partial reduction in cavin-1 expression in the lungs. The re-expression of caveolin-1 rescues both caveolin-2 and cavin-1 [70]. In a carotid artery-injury model, the local loss of cavin-1 is reported to promote neointima formation. Furthermore, in cultured vascular SMC, the overexpression of cavin-1 suppresses SMC proliferation and migration, whereas its inhibition promotes cell proliferation and migration [71]. Cavin-1 knockout mice display lung pathological changes such as remodeled pulmonary vessels, PH and right ventricular hypertrophy. In addition, these mice have altered metabolic phenotype with insulin resistance [72, 73].

Recent studies have shown other accessory proteins required in caveolae biogenesis. The accessory protein pacsin2 also known as syndapin2 contains F-BAR domain associated with generation and maintenance of caveolae. It partially colocalizes with caveolin-1 at plasma membrane level. Loss of pacsin2 function results in the loss of caveolae and accumulation of caveolin-1 within the plasma membrane. Interestingly, overexpression of F-BAR domain can cause loss of caveolae. Another protein EH 15 homology domain 2 (EHD2) is present in caveolae, and it binds to pacsin2 that partially colocalizes with caveolin-1. It is a dynamin-related ATPase that confines caveolae to cell surface. Furthermore, regulation of EHD2 oligomerization in a membrane-bound state is crucial in order to restrict caveolar dynamics in cells [74, 75]. Importantly, caveolar coat controls a large number of signaling circuits; a defect in any of these pathways can lead to several systemic diseases such as vascular dysfunction, cardiomyopathy, cancer, muscular dystrophy and lipodystrophy [76].

The role of caveolin-1 is well established in the pathogenesis of PH. Caveolin-1 knockout mice are viable but have dysregulated NO synthesis, impaired NO and Ca2+ signaling, cell proliferation, increased vascular permeability accompanied by cardiomyopathy and PH. Reconstituting endothelial caveolin-1 has been shown to recover dysregulated NO synthesis, cardiomyopathy and PH [77, 78]. In addition, caveolin-1 knockout mice exhibit low-grade systemic pro-inflammatory status and moderately increased IL-6 and TNFα levels [79]. EC-specific CAV1 knockout mice and LPS-treated wild-type mice exhibit reduced BMPR2 expression and eNOS uncoupling, accompanied by increased TGF-β–promoted TGFβRI-dependent SMAD-2/3 phosphorylation. In addition, human lung sections from patients with ARDS reveal reduced endothelial caveolin-1 expression, increased TGF-β levels and severe pulmonary vascular remodeling. These results further support the view that the loss of endothelial caveolin-1 promotes pulmonary vascular remodeling in inflamed lungs via oxidative stress-mediated reduction in BMPR2 expression [80]. Furthermore, endothelial dysfunction during inflammation leads to endothelium-mesenchymal transition (End MT). These cells lose endothelial characteristics and acquire mesenchymal phenotypes and express mesenchymal specific markers such as smooth muscle α-actin, fibroblast-specific protein 1 and Notch1 [81]. In addition, caveolin-1 is a determinant of oxidative stress and is a regulator of metabolic switch and autophagy [82].

2.2 Vascular relaxation

NO, EDHF and prostacyclin (PGI2) induce endothelium-dependent vascular relaxation. NO is produced by eNOS via its action on L-arginine and oxygen. NO activates guanylate cyclase, which catalyzes the conversion of guanosine triphosphate to cyclic guanylate monophosphate. eNOS expressed in endothelial cells and cardiac myocytes is targeted to caveolae. It directly binds to caveolin-1 scaffolding domain and is held in an inhibitory state. This interaction prevents eNOS activation leading to inappropriate NO production under basal conditions. The eNOS/caveolin-1 regulatory cycle is a reversible protein-protein interaction controlled by Ca2+/calmodulin and by enzyme palmitoylation. Increase in intracellular Ca2+ with calmodulin disrupts the caveolin-1/eNOS complex resulting in eNOS activation and NO production leading to vascular relaxation. Calmodulin is a direct allosteric competitor promoting the caveolin-1 and eNOS dissociation. Heat shock protein (HSP) 90 binds to eNOS in Ca2+/calmodulin-dependent manner and it reduces the inhibitory effects of caveolin scaffolding domain on eNOS, thus promoting eNOS activation [83, 84, 85]. Furthermore, increase in vascular flow and pressure rapidly activates caveolar eNOS with its dissociation from caveolin-1 and association with calmodulin [86]. Thus, caveolin-1 and eNOS have a dynamic relationship. Importantly, caveolin-1 contained within non-caveolar lipid rafts fails to exert its inhibitory effect on eNOS [87]. The loss of endothelial caveolin-1 leads to eNOS uncoupling, oxidative stress and endothelial injury [88]. Interestingly, under conditions of stress, caveolin-1 increases eNOS trafficking in plasma membrane and primes eNOS for flow-mediated activation. Caveolin-1 plays a positive role in shear-induced eNOS activation by targeting eNOS to plasma membrane. Importantly, the coupling of flow stimulus to activate eNOS is lost in the absence of caveolin-1 and caveolae. Thus, caveolin-1 exerts dual role of post-translational regulation of eNOS activity [89]. In addition, caveolin-1 plays a critical role in VEGFR2 stimulation and downstream mediators of angiogenesis, but higher levels of caveolin-1 repress this function [90]. Interestingly, EC migration, tube formation and angiogenesis are impaired both in caveolin-1 and eNOS knockout mice but are fully restored by double knockout [91].

The transient receptor potential (TRP) channels are the link between caveolae and EDHF. TRP channels facilitating the capacitive Ca2+ entry directly interact with caveolin-1 in EC. Ca2+-activated K+ channels play a key role in endothelium-dependent hyperpolarization and vascular tone regulation. Absence of caveolin-1 impairs Ca2+ homeostasis in EC and decreases the activity of TPRV4 cation channels that participate in NO and EDHF activation. Caveolin-1 is required for EDHF-related relaxation, modulating TRPV4 and connexins. Caveolin-1 knockout arteries exhibit fewer gap junctions and altered myoendothelial communication. Furthermore, caveolin-1 deficiency is associated with impaired EDHF-mediated vascular relaxation in response to shear stress and acetylcholine [92, 93, 94]. Colocalization of PGI2 synthase and caveolin-1 regulates angiogenesis [95]. Thus, caveolin-1 interacts with relaxing factors to maintain homeostasis.

2.3 Barrier function

Endothelial barrier controls the passage of fluids, nutrients and cells through vascular wall. Glycocalyx coats the luminal surface of EC and forms an important barrier. It modulates permeability, prevents leukocyte and platelet adhesion to EC, serves as an anti-inflammatory, anti-adhesive and anti-coagulant barrier, and allows selective permeability. In addition, it mediates mechanotransduction of shear stress. Under normal conditions, the apoptosis rate in EC is very low, but the activated EC exhibits a reduction in the EC surface layer, the glycocalyx, and an increased rate of apoptosis [96, 97]. Disturbed flow has been shown to inhibit glycocalyx expression as well as to reduce caveolin-1 expression in systemic arterial EC [98]. Inflammatory mediators lead to the disruption of glycocalyx resulting in the weakening of vascular protection. Integrity of vascular glycocalyx is inversely related to the degree of inflammation. Inflammatory mediators lead to the loss of glycocalyx resulting in the weakening of vascular protection. Furthermore, destruction of glycocalyx has been reported in the MCT model of PH [99].

Ca2+-dependent vascular endothelial cadherin (VE-Cad) and its associated catenins control cell-cell adhesion and paracellular barrier function and are important for the tight junction complexes. VE-Cad is tissue specific for EC and is expressed at the intercellular clefts and mediates cell-cell adhesion, maintains barrier function, and contributes to the inhibition of cell growth. Association of caveolin-1 and VE-Cad catenin complex is essential for barrier function [100, 101, 102]. Depletion of caveolin-1 reduces VE-Cad levels and facilitates endothelial cell permeability [103]. Furthermore, VE-Cad interacts with various growth receptors, regulates endothelial proliferative signaling and mediates contact inhibition of cell growth [104, 105]. In adult EC, VE-Cad and VEGFR2 are physically linked. This maintains VEGFR2 stable and prevents its endocytosis. VEGF-induced permeability is facilitated by decoupling of VE-Cad and VEGFR2 [106]. Loss of VE-Cad and PECAM1 has been shown to occur in the MCT-induced PH [31, 107]. PECAM-1 supports EC integrity and maintains barrier function [108]. Importantly, BMPR2 also plays a role in maintaining vascular integrity by dampening inflammatory signals in pulmonary vasculature [109].

Advertisement

3. Endothelial disruption/dysfunction

Vascular injury from different conditions such as inflammation, hypoxia, increased flow and pressure, and shear stress leads to endothelial dysfunction. Injury can lead to disruption of EC and endothelial caveolin-1 loss or endothelial dysfunction without EC disruption. Both lead to the activation of proliferative pathways, vascular remodeling and PH [96, 110]. Recent studies have shown the loss of myocyte enhancer factor 2 (MEF2) in dysfunctional EC from PAH patients. MEF2 regulates a number of transcription factors involved in pulmonary vascular homeostasis [111]. Furthermore, these dysfunctional ECs exhibit increased production of leptin, and SMCs overexpress leptin receptor contributing to SMC proliferation [112]. In addition, pulmonary arterial ECs from PAH patients have been shown to produce increased FGF2 leading to increased proliferation and survival response by constitutive activation of ERK1/2 and decreased apoptosis associated with the activation of Bcl2 and Bcl-xL. It is thought that FGF2 in PAH may contribute to abnormal EC phenotype [113]. Furthermore, there is evidence that pro-inflammatory cytokine macrophage migration inhibitory factor and its receptor CD74 are markedly increased in idiopathic PAH, which may contribute to pro-inflammatory phenotype of EC [114].

3.1 EC disruption and pulmonary hypertension

Endothelial disruption accompanied by the loss of endothelial caveolin-1 has been reported in several forms of experimental models of PH such as MCT, myocardial infarction and sugen + hypoxia [31, 115, 116]. In the MCT model, progressive loss of endothelial caveolin-1 and reciprocal activation of proliferative and anti-apoptotic pathways such as PY-STAT3 and Bcl-xL occur before the onset of PH. Loss of other membrane proteins such as PECAM-1, Tie2 and soluble guanylate cyclase (α and β) occurs in tandem with caveolin-1 loss indicating extensive disruption of endothelial cell membrane. At 2 weeks, a further loss of endothelial caveolin-1 is accompanied by the loss of cytosolic proteins such as HSP90 and IκB-α and increased pulmonary artery pressure. However, at this stage, eNOS expression is relatively well preserved. In the presence of significant loss of endothelial caveolin-1 and HSP90, eNOS gets uncoupled resulting in an increased production of reactive oxygen species (ROS). By 3 and 4 weeks, there is a significant reduction in eNOS levels, leading to normalization of ROS levels [9, 31, 117]. At 4 weeks post-MCT, extensive endothelial caveolin-1 loss is accompanied by the loss of von Willebrand factor (vWF) in 29% of the arteries; and 23% of arteries exhibit enhanced expression of caveolin-1 in SMC. Enhanced expression of caveolin-1 in SMC occurs only in the arteries with extensive endothelial caveolin-1 and vWF loss. At this stage, the expression of total caveolin-1 in the lungs remains low. In addition, there is a progressive increase in MMP2 expression and activation [117]. The rescue of endothelial caveolin-1 as a preventive measure abrogates MCT-induced PH, but once the PH is established, the treatment does not alter the progression of the disease [118, 119, 120]. Exposing MCT-treated rats to hypoxia accelerates the disease process, and by 4 weeks, extensive endothelial disruption and endothelial caveolin-1 loss are accompanied by enhanced expression of caveolin-1 in SMC in 61% of the arteries, near normalization of lung caveolin-1 expression, and neointima formation. Importantly, neointimal cells exhibit low to no caveolin-1 expression [121, 122]. Extensive loss of endothelial caveolin-1, enhanced expression of caveolin-1 in SMC and neointima formation are also observed in idiopathic and hereditary PAH, PAH associated with CHD and drug toxicity [122, 123, 124, 125]. In in-vitro studies, pulmonary arterial SMCs from idiopathic PAH exhibit increased caveolin-1 expression accompanied by increased capacitive Ca2+ entry and DNA synthesis, which could be abrogated by silencing caveolin-1 [125]. Loss of EC exposes SMC to direct pressure and shear stress, which is likely to result in flattening of caveolae leading to displacement of caveolin-1 to non-caveolar site on the plasma membrane. Recently, it has been shown that in the MCT + hypoxia model, at 4 weeks, the extensive loss of endothelial caveolin-1 as well as VE-Cad loss and enhanced expression of caveolin-1 in SMC are accompanied by cavin-1 loss, tyrosine phosphorylation of caveolin-1 and neointima formation. Loss of VE-Cad is indicative of loss of EC attachment to the junction [126]. Interestingly, p-caveolin-1 in cancer has been shown to make cells pro-migratory [127, 128]. As PH progresses, SMC phenotype changes from contractile to synthetic, facilitating cell migration, and neointima formation resulting in arterial occlusion. Neointima formation leads to the irreversibility of the disease [129]. In addition, increasing pulmonary blood flow either by pneumonectomy or by a shunt procedure (left subclavian and pulmonary artery) in rats treated with MCT leads to the development of neointimal lesions. Pneumonectomy or shunt alone does not lead to neointima formation [130, 131]. Furthermore, in children with significant left to right cardiac shunt, reversal of pulmonary vascular changes were seen after they underwent pulmonary artery banding to restrict the pulmonary flow. Medial hypertrophy and early intimal changes seem reversible, but not during the later stages [132, 133]. These studies demonstrate that EC injury and disruption associated with increased flow or pressure play an important role in determining the pattern of pulmonary vascular remodeling.

Apoptosis of EC in PAH is followed by proliferation of antiapoptotic EC. This concept has been confirmed in in-vitro studies. Sugen (VEGFR antagonist) causes initial apoptosis, and the surviving cells become hyperproliferative [134]. Importantly, increased levels of circulating EC (CEC) have been reported in PAH, and 50% of these cells expressed CD36, a marker of microvascular origin, and 25% exhibited E selectin, a marker of EC activation [135]. In children with CHD and PAH, the increased levels of CEC are reported to be associated with worse prognosis. Pulmonary ECs exhibited high expression of antiapoptotic protein Bcl-2 in cases of irreversible PAH but not in cases of reversible PAH, or in controls. Interestingly, intimal proliferation was observed only in irreversible PAH cases, but not in the reversible PAH [136, 137]. In addition, increased vWF levels in patients with PAH were reported to be associated with worse survival [138]. Interestingly, increased CEC levels were observed in PAH, but not in CTEPH [139]. These studies strongly support the view that the disruption and loss of EC are associated with severe PAH and poor prognosis.

Endothelial mesenchymal transition (EndMT) is a process by which ECs exhibit phenotype alteration. These cells lose endothelial characteristics and acquire the properties of myofibroblasts or mesenchymal cells. They exhibit loss of PECAM-1 and VE-Cad, in addition to caveolin-1, and express smooth muscle α-actin, fibroblast-specific protein 1 and Notch1. PECAM-1 and VE-Cad support EC integrity and junctional stability and maintain barrier function. Thus, their loss leads to the loss of barrier function and junction stability. These transformed ECs also acquire pro-inflammatory phenotype and are primed for proliferation, migration and tissue generation [81, 140]. Neointimal cells exhibit low levels of caveolin-1, but normal eNOS expression in the experimental model of PH and also in human PAH [96, 122], and sustained NO production has been shown to degrade caveolin-1 [141]. Importantly, caveolin-1 deficiency has been shown to induce spontaneous EndMT in pulmonary EC [142]. EndMT plays an important role in vascular remodeling, and it is also linked to the loss of BMPR2 in PH [143, 144, 145]. Furthermore, TGFβ1 plays a significant role in EndMT [146]. In addition, endothelial caveolin-1 depletion leads to eNOS uncoupling and oxidative stress that switches from BMPR2 signaling to TGFβR1 and thus may promote EndMT [80]. Plexogenic lesions contain increased VEGFA and VEGFR expression indicating misguided angiogenesis involving cells of EC origin. EC dysfunction in PAH model is shown to act through DNA methylation, histone protein modification and non-coding RNA [19]. Thus, the initial apoptosis followed by the proliferation of dysfunctional and antiapoptotic EC leads to deregulation of a number of pathways resulting in neointima and plexiform lesion formation and irreversible PAH.

3.2 EC dysfunction without EC disruption and pulmonary hypertension

Exposure to acute hypoxia results in pulmonary arterial contraction and elevated pulmonary artery pressure, while sustained hypoxia leads to pulmonary vascular remodeling [147]. Hypoxia impairs endothelium-dependent relaxation response [148, 149]. In the MCT model of PH, the progressive loss of endothelial caveolin-1 is accompanied by a significant reduction in the expression of HSP90 (2 weeks post-MCT) and eNOS (3 weeks post-MCT) [9]. However, hypoxia does not alter the protein expression of caveolin-1, eNOS or HSP90 in the lungs. During hypoxia, caveolin-1 and eNOS have been shown to form a tight complex in vivo and in vitro, resulting in their dysfunction [110, 150]. Normally, in response to Ca2+ agonists, eNOS dissociates from caveolin-1 and binds to HSP90. Ca2+ activated calmodulin further aids in recruitment of HSP90, thus facilitating the release of eNOS from caveolin-1 [151]. However, hypoxia disrupts eNOS/HSP90 binding [152]. Furthermore, normally functioning caveolin-1 is required for the plasma membrane localization of TRPC4 and endothelial Ca2+ entry [153], and introduction of caveolin-1 scaffolding domain restores Ca2+ entry during chronic hypoxia [154]. Thus, the hypoxia-induced caveolin-1 and eNOS complex formation may in part be responsible for the deregulation of Ca2+ entry and disruption of HSP90/eNOS binding leading to impaired vascular relaxation. Statins have been shown to disrupt hypoxia-induced abnormal coupling of eNOS and caveolin-1, thus restoring eNOS function and attenuating hypoxia-induced PH [155]. Recent studies of hypoxia-induced PH in rats and cows showed no disruption of EC or any alterations in the expression of caveolin-1, VE-Cad or vWF [46, 126]. Not surprisingly, there was no enhanced expression of caveolin-1 in SMC as seen in the MCT model. However, there was evidence of caveolin-1 dysfunction, such as the activation of proliferative pathways such as PY-STAT3, β-catenin and pERK1/2, and a loss of PTEN. PTEN contains a Cav-1–binding motif and, in part, colocalizes in caveolae. Caveolin-1 determines the membrane PTEN levels through its binding sequence. The loss of PTEN during hypoxia further confirms caveolin-1 dysfunction [46].

People living at high altitude develop PH and right ventricular hypertrophy as an adaptive mechanism. Upon return to sea level, PH reverts to normal slowly [156]. These observations suggest that the absence of physical disruption of EC observed in the hypoxia model may be the reason why hypoxia-induced PH is reversible. Although hypoxia plays a role, inflammation and endothelial dysfunction are important factors that determine the development of PH in chronic obstructive pulmonary disease (COPD). The outflow obstruction in COPD results from inflammatory processes affecting airways, lung parenchyma and pulmonary vasculature. PH in COPD can develop independently of underlying parenchymal destruction and loss of lung vessels [157, 158]. Endothelial dysfunction has been observed in mild cases of COPD, and the loss of endothelium-dependent relaxation in the pulmonary vasculature correlates with the severity of the disease [159]. Importantly, the loss of endothelial caveolin-1 accompanied by enhanced expression of caveolin-1 in SMC is reported in COPD associated with PH. COPD without PH had preserved endothelial caveolin-1 [160]. In addition, severe pulmonary arterial lesions such as plexiform and angiomatoid lesions have been documented in explanted lungs after transplantation in COPD associated with severe PH. These lesions were similar to what are seen in IPAH [161]. In infants with respiratory distress syndrome, despite significantly elevated pulmonary artery pressure and significant medial thickening, pulmonary arteries exhibit well-preserved endothelial caveolin-1, without any evidence of EC disruption or enhanced expression of Cav-1 in SMC. In contrast, loss of endothelial Cav-1 and disruption/loss of EC coupled with enhanced expression of Cav-1 in SMC were observed in infants with bronchopulmonary dysplasia and associated inflammation [123]. These results indicate that irrespective of the underlying disease, EC disruption leads to the loss of endothelial caveolin-1 and subsequent enhanced expression of Cav-1 in SMC, followed by neointima formation and irreversible PH. Thus, the EC disruption puts the patients at a higher risk of developing irreversible PH.

Advertisement

4. Conclusions

Under normal conditions, ECs play a key role in maintaining SMCs in quiescent state and vascular homeostasis. Caveolin-1, a major protein constituent of caveolae on the cell membrane, regulates multiple cellular processes including inflammation, molecular transport, cell proliferation, adhesion, migration and signal transduction. Caveolin-1 interacts with protein molecules that are in or are recruited to caveolae and maintains them in inhibitory confirmation. Endothelial caveolin-1 loss and caveolin-1 dysfunction lead to PH.

4.1 EC disruption and caveolin-1 loss

Injury such as inflammation, increased pulmonary blood flow associated with increased pressure, drugs and toxins can cause endothelial disruption, which is usually progressive. Endothelial disruption leads to the progressive loss of endothelial membrane proteins including caveolin-1, PECAM-1 and VE-Cad. These alterations lead to deregulation of multiple pathways. As depicted in Figure 1, (a) the loss of caveolin-1 is accompanied by reciprocal activation of proliferative and antiapoptotic pathways leading to SMC hypertrophy and proliferation. (b) Further loss of EC exposes SMCs to direct pressure resulting in enhanced expression of caveolin-1 in SMCs. Tyrosine phosphorylated caveolin-1 could alter the phenotype and facilitate cell migration leading to neointima formation. (c) Loss of PECAM-1 and VE-Cad results in the loss of barrier function and junction stability. These alterations lead to EndMT. These cells lose endothelial properties and acquire pro-inflammatory phenotype and are primed for proliferation, migration and tissue generation and participate in neointima formation, thus leading to irreversible PH.

Figure 1.

This figure depicts the pathway leading from endothelial cell disruption to irreversible PH. Cav-1 = caveolin-1, EC = endothelial cells, EndMT = endothelial mesenchymal transformation, PECAM-1 = platelet endothelial cell adhesion molecule 1, pCav-1 = tyrosine phosphorylated caveolin-1, PH = pulmonary hypertension, SMC = smooth muscle cells.

4.2 EC and caveolin-1 dysfunction

Hypoxia exposure to EC leads to a tight complex formation between caveolin-1 and eNOS, resulting in the dysfunction of both factors (Figure 2). Importantly, there is no EC disruption or the loss of caveolin-1 or any other membrane proteins. Since there is no loss of EC, medial layer is not exposed to shear stress and pressure. Not surprisingly, there is no enhanced expression of caveolin-1 in SMCs. However, caveolin-1 and eNOS dysfunction lead to SMC proliferation, medial hypertrophy and loss of endothelial-dependent vascular relaxation. Removal of hypoxia results in the disruption of caveolin-1/eNOS tight complex leading to reversal of PH. Slowly, the pulmonary artery pressure and medial hypertrophy return to normal as seen in experimental animals and in people returning to sea level from high altitude. Hypoxia-induced PH is reversible. However, associated inflammation/shear stress in hypoxia-induced PH, resulting in EC disruption, would lead to irreversible PH.

Figure 2.

This figure shows the effect of hypoxia on EC leading to eNOS/caveolin-1 complex formation, endothelial dysfunction and subsequent medial hypertrophy and PH. EC = endothelial cells, Cav-1 = caveolin-1, eNOS = endothelial nitric oxide synthase, SMC = smooth muscle cells, PH = pulmonary hypertension.

In conclusion, EC integrity and caveolin-1 function are important factors that determine reversible vs. irreversible PH.

Advertisement

Acknowledgments

This work is in part supported by Cardiovascular Medical Research and Education Fund.

References

  1. 1. Simonneau G, Gatzoulis MA, Adatia I, Celermajer D, Denton C, Ghofrani A, et al. Updated clinical classification of pulmonary hypertension. Journal of the American College of Cardiology. 2013;62(25 Suppl):D34-D41. DOI: 10.1016/j.jacc.2013.10.029
  2. 2. Best DH, Sumner KL, Smith BP, Damjanovich-Colmenares K, Nakayama I, Brown LM, et al. EIF2AK4 mutations in patients diagnosed with pulmonary arterial hypertension. Chest. 2017;151:821-828. DOI: 10.1016/j.chest.2016.11.014
  3. 3. Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, et al. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. European Respiratory Journal. 2019;53(1). DOI: 10.1183/13993003.01913-2018
  4. 4. Kovacs G, Berghold A, Scheidl S, Olschewski H. Pulmonary arterial pressure during rest and exercise in healthy subjects: A systematic review. The European Respiratory Journal. 2009;34:888-894. DOI: 10.1183/09031936.00145608
  5. 5. Hoeper MM, Humbert M, Souza R, Idrees M, Kawut SM, Sliwa-Hahnle K, et al. A global view of pulmonary hypertension. The Lancet Respiratory Medicine. 2016;4:306-322. DOI: 10.1016/S2213-2600(15)00543-3
  6. 6. Thenappan T, Shah SJ, Rich S, Tian L, Archer SL, Gomberg-Maitland M. Survival in pulmonary arterial hypertension: A reappraisal of the NIH risk stratification equation. The European Respiratory Journal. 2010;35:1079-1087. DOI: 10.1183/09031936.00072709
  7. 7. Pogoriler JE, Rich S, Archer SL, Husain AN. Persistence of complex vascular lesions despite prolonged prostacyclin therapy of pulmonary arterial hypertension. Histopathology. 2012;61:597-609. DOI: 10.1111/j.1365-2559.2012.04246.x
  8. 8. Strange G, Gabbay E, Kermeen F, Williams T, Carrington M, Stewart S, et al. Time from symptoms to definitive diagnosis of idiopathic pulmonary arterial hypertension: The delay study. Pulmonary Circulation. 2013;3:89-94. DOI: 10.4103/2045-8932.109919
  9. 9. Huang J, Wolk JH, Gewitz MH, Mathew R. Progressive endothelial cell damage in an inflammatory model of pulmonary hypertension. Experimental Lung Research. 2010;36:57-66. DOI: 10.3109/01902140903104793
  10. 10. Mathew R. Pulmonary hypertension management challenges in pediatric age group. Annals of Vascular Medicine and Research. 2017;4(3):1057
  11. 11. Muensterer O, Abellar R, Otterburn D, Mathew R. Pulmonary agenesis and associated pulmonary hypertension: A case report and review on variability, therapy, and outcome. European Journal of Pediatric Surgery Reports. 2015;3:33-39. DOI: 10.1055/s-0034-1395986
  12. 12. van Loon RL, Roofthooft MT, Hillege HL, ten Harkel AD, van Osch-Gevers M, Delhass T, et al. Pediatric pulmonary hypertension in Netherlands: Epidemiology and characterization during period 1991 to 2005. Circulation. 2011;124:1755-1764. DOI: 10.1161/CIRCULATIONAHA.110.969584
  13. 13. Naumburg E, Söderström L, Huber D, Axelsson I. Risk factors for pulmonary arterial hypertension in children and young adults. Pediatric Pulmonology. 2017;52:636-641. DOI: 10.1002/ppul.23633
  14. 14. Chida A, Shintani M, Yagi H, Fujiwara M, Kojima Y, Sato H, et al. Outcomes of childhood pulmonary arterial hypertension in BMPR2 and ALK1 mutation carriers. The American Journal of Cardiology. 2012;110:586-593. DOI: 10.1016/j.amjcard.2012.04.035
  15. 15. Galambos C, Mullen MP, Shieh JT, Schwerk N, Kielt MJ, Ullmann N, et al. Phenotype characterisation of TBX4 mutation and deletion carriers with neonatal and pediatric pulmonary hypertension. European Respiratory Journal. 2019;54(2):1801965. DOI: 10.1183/13993003.01965-2018
  16. 16. Rabinovitch M, Keane JF, Norwood WI, Castaneda AR, Reid L. Vascular structure in lung tissue obtained at biopsy correlated with pulmonary hemodynamic findings after repair of congenital heart defects. Circulation. 1984;69:655-667. DOI: 10.1161/01.cir.69.4.655
  17. 17. Yamaki S, Wagenvoort CA. Comparison of primary plexogenic arteriopathy in adults and children. A morphometric study in 40 patients. British Heart Journal. 1985;54:428-434. DOI: 10.1136/hrt.54.4.428
  18. 18. Lee SD, Shroyer KR, Markham NE, Cool CD, Voelkel NF, Tuder RM. Monoclonal endothelial cell proliferation is present in primary but not secondary pulmonary hypertension. The Journal of Clinical Investigation. 1998;101:927-934. DOI: 10.1172/JCI1910
  19. 19. Ranchoux B, Harvey LD, Ayon RJ, Babicheva A, Bonnet S, Chan SY, et al. Endothelial dysfunction in pulmonary arterial hypertension: An evolving landscape (2017 Grover conference series). Pulmonary Circulation. 2018;8:2045893217752912. DOI: 10.1177/2045893217752912
  20. 20. Humbert M, Morrell NW, Archer SL, Stenmark KR, MacLean MR, Lang IM, et al. Cellular and molecular pathobiology of pulmonary arterial hypertension. Journal of the American College of Cardiology. 2004;43(12 Suppl S):13S-24S. DOI: 10.1016/j.jacc.2004.02.029
  21. 21. Mathew R. Inflammation and pulmonary hypertension. Cardiology in Review. 2010;18:67-72. DOI: 10.1097/CRD.0b013e3181cd612f
  22. 22. Balabanian K, Foussat A, Dorfmüller P, Durand-Gasselin I, Capel F, Bouchet-Delbos L, et al. CX (3) C chemokine fractalkine in pulmonary arterial hypertension. American Journal of Respiratory and Critical Care Medicine. 2002;165:1419-1425. DOI: 10.1164/rccm.2106007
  23. 23. Dorfmüller P, Zarka V, Durand-Gasselin I, Monti G, Balabanian K, Garcia G, et al. Chemokine RANTES in severe pulmonary arterial hypertension. American Journal of Respiratory and Critical Care Medicine. 2002;165:534-539. DOI: 10.1164/ajrccm.165.4.2012112
  24. 24. Nie X, Tan J, Dai Y, Liu Y, Zou J, Sun J, et al. CCL5 deficiency rescues pulmonary vascular dysfunction, and reverses pulmonary hypertension via caveolin-1-dependent BMPR2 activation. Journal of Molecular and Cellular Cardiology. 2018;116:41-56. DOI: 10.1016/j.yjmcc.2018.01.016
  25. 25. Tuder RM, Groves B, Badesch DB, Voelkel NF. Exuberant endothelial cell growth and elements of inflammation are present in plexiform lesions of pulmonary hypertension. The American Journal of Pathology. 1994;144:275-285
  26. 26. Stacher E, Graham BB, Hunt JM, Gandjeva A, Groshong SD, McLaughlin VV, et al. Modern age pathology of pulmonary arterial hypertension. American Journal of Respiratory and Critical Care Medicine. 2012;186:261-272. DOI: 10.1164/rccm.201201-0164OC
  27. 27. Humbert M, Monti G, Brenot F, Sitbon O, Portier A, Grangeot-Keros L, et al. Increased interleukin-1 and interleukin-6 serum concentrations in severe primary pulmonary hypertension. American Journal of Respiratory and Critical Care Medicine. 1995;151:1628-q631. DOI: 10.1164/ajrccm.151.5.7735624
  28. 28. Bhargava A, Kumar A, Yuan N, Gewitz MH, Mathew R. Monocrotaline induces interleukin-6 mRNA expression in rat lungs. Heart Disease. 1999;1:126-132
  29. 29. Schermuly RT, Ghofrani HA, Wilkins MR, Grimminger F. Mechanisms of disease: Pulmonary arterial hypertension. Nature Reviews. Cardiology. 2011;8:443-455. DOI: 10.1038/nrcardio.2011.87
  30. 30. Xu W, Erzurum SC. Endothelial cell energy metabolism, proliferation, and apoptosis in pulmonary hypertension. Comprehensive Physiology. 2011;1:357-372. DOI: 10.1002/cphy.c090005
  31. 31. Mathew R, Huang J, Shah M, Patel K, Gewitz M, Sehgal PB. Disruption of endothelial-cell caveolin-1alpha/raft scaffolding during development of monocrotaline-induced pulmonary hypertension. Circulation. 2004;110:1499-1506. DOI: 10.1161/01.CIR.0000141576.39579.23
  32. 32. Masri FA, Xu W, Comhair SA, Asosingh K, Koo M, Vasanji A, et al. Hyperproliferative apoptosis-resistant endothelial cells in idiopathic pulmonary arterial hypertension. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2007;293:L548-L554. DOI: 10.1152/ajplung.00428.2006
  33. 33. Shibata R, Kai H, Seki Y, Kato S, Wada Y, Hanakawa Y, et al. Inhibition of STAT3 prevents neointima formation by inhibiting proliferation and promoting apoptosis of neointimal smooth muscle cells. Human Gene Therapy. 2003;14:601-610. DOI: 10.1089/104303403321618128
  34. 34. Soon E, Crosby A, Southwood M, Yang P, Tajsic T, Toshner M, et al. Bone morphogenetic protein receptor type II deficiency and increased inflammatory cytokine production. A gateway to pulmonary arterial hypertension. American Journal of Respiratory and Critical Care Medicine. 2015;192:859-872. DOI: 10.1164/rccm.201408-1509OC
  35. 35. Brock M, Trenkmann M, Gay RE, Michel BA, Gay S, Fischler M, et al. Interleukin-6 modulates the expression of the bone morphogenic protein receptor type II through a novel STAT3-microRNA cluster 17/92 pathway. Circulation Research. 2009;104:1184-1191. DOI: 10.1161/CIRCRESAHA.109.197491
  36. 36. Atkinson C, Stewart S, Upton PD, Machado R, Thomson JR, Trembath RC, et al. Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor. Circulation. 2002;105:1672-1678. DOI: 10.1161/01.cir.0000012754.72951.3d
  37. 37. Murakami K, Mathew R, Huang J, Farahani R, Peng H, Olson SC, et al. Smurf1 ubiquitin ligase causes downregulation of BMP receptors and is induced in monocrotaline and hypoxia models of pulmonary arterial hypertension. Experimental Biology and Medicine. 2010;235:805-813. DOI: 10.1258/ebm.2010.009383
  38. 38. Rothman AM, Arnold ND, Pickworth JA, Iremonger J, Ciuclan L, Allen RM, et al. MicroRNA-140-5p and SMURF1 regulate pulmonary arterial hypertension. Journal of Clinical Sciences. 2016;126:2495-2508. DOI: 10.1172/JCI83361
  39. 39. Nickel NP, Spiekerkoetter E, Gu M, Li CG, Li H, Kaschwich M, et al. Elafin reverses pulmonary hypertension via caveolin-1-dependent bone morphogenetic protein signaling. American Journal of Respiratory and Critical Care Medicine. 2015;191(11):1273-1286. DOI: 10.1164/rccm.201412-2291OC
  40. 40. Dabral S, Tian X, Kojonazarov B, Savai R, Ghofrani HA, Weissmann N, et al. Notch1 signalling regulates endothelial proliferation and apoptosis in pulmonary arterial hypertension. The European Respiratory Journal. 2016;48:1137-1149. DOI: 10.1183/13993003.00773-2015
  41. 41. Zou J, Li P, Lu F, Liu N, Dai J, Ye J, et al. Notch1 is required for hypoxia-induced proliferation, invasion and chemoresistance of T-cell acute lymphoblastic leukemia cells. Journal of Hematology & Oncology. 2013;6:3. DOI: 10.1186/1756-8722-6-3
  42. 42. Takeshita K, Satoh M, Ii M, Silver M, Limbourg FP, Mukai Y, et al. Critical role of endothelial Notch1 signaling in postnatal angiogenesis. Circulation Research. 2007;100:70-78. DOI: 10.1161/01.RES.0000254788.47304.6e
  43. 43. Mack JJ, Iruela-Arispe ML. NOTCH regulation of the endothelial cell phenotype. Current Opinion in Hematology. 2018;25:212-218. DOI: 10.1097/MOH.0000000000000425
  44. 44. Mack JJ, Mosqueiro TS, Archer BJ, Jones WM, Sunshine H, Faas GC, et al. NOTCH1 is a mechanosensor in adult arteries. Nature Communications. 2017;8:1620. DOI: 10.1038/s41467-017-01741-8
  45. 45. Serra H, Chivite I, Angulo-Urarte A, Soler A, Sutherland JD, Arruabarrena-Aristorena A, et al. PTEN mediates notch-dependent stalk cell arrest in angiogenesis. Nature Communications. 2015;6:7935. DOI: 10.1038/ncomms8935
  46. 46. Huang J, Frid M, Gewitz MH, Fallon JT, Brown D, Krafsur G, et al. Hypoxia-induced pulmonary hypertension and chronic lung disease: Caveolin-1 dysfunction an important underlying feature. Pulmonary Circulation. 2019;9(1):2045894019837876. DOI: 10.1177/2045894019837876
  47. 47. Xia H, Khalil W, Kahm J, Jessurun J, Kleidon J, Henke CA. Pathologic caveolin-1 regulation of PTEN in idiopathic pulmonary fibrosis. The American Journal of Pathology. 2010;176:2626-2637. DOI: 10.2353/ajpath.2010.091117
  48. 48. Sun S, Steinberg BM. PTEN is a negative regulator of STAT3 activation in human papillomavirus-infected cells. The Journal of General Virology. 2002;83:1651-1658. DOI: 10.1099/0022-1317-83-7-1651
  49. 49. Mathew R. Pulmonary hypertension: Endothelial cell function. In: Sulica R, Preston I, editors. Pulmonary Hypertension: From Bench Research to Clinical Challenge. Croatia: IntechOpen; 2011. pp. 1-24. ISBN: 978-953-307-835-9
  50. 50. Dorland YL, Huveneers S. Cell-cell junctional mechanotransduction in endothelial remodeling. Cellular and Molecular Life Sciences. 2017;74:279-292. DOI: 10.1007/s00018-016-2325-8
  51. 51. Palade GE. Fine structure of blood capillaries. Journal of Applied Physiology. 1953;24:1424
  52. 52. Yamada E. The fine structure of the gall bladder epithelium of the mouse. The Journal of Biophysical and Biochemical Cytology. 1955;1:445-458. DOI: 10.1083/jcb.1.5.445
  53. 53. Couet J, Li S, Okamoto T, Ikezu T, Lisanti MP. Identification of peptide and protein ligands for the caveolin-scaffolding domain. Implications for the interaction of caveolin with caveolae-associated proteins. The Journal of Biological Chemistry. 1997;272(10):6525-6533. DOI: 10.1074/jbc.272.10.6525
  54. 54. Parton RG, del Pozo MA. Caveolae as plasma membrane sensors, protectors and organizers. Nature Reviews. Molecular Cell Biology. 2013;14:98-112. DOI: 10.1038/nrm3512
  55. 55. Sinha B, Köster D, Ruez R, Gonnord P, Bastiani M, Abankwa D, et al. Cells respond to mechanical stress by rapid disassembly of caveolae. Cell. 2011;144:402-413. DOI: 10.1016/j.cell.2010.12.031
  56. 56. Frank PG, Woodman SE, Park DS, Lisanti MP. Caveolin, caveolae, and endothelial cell function. Arteriosclerosis, Thrombosis, and Vascular Biology. 2003;23:1161-1168. DOI: 10.1161/01.ATV.0000070546.16946.3A
  57. 57. Shaul PW, Anderson RG. Role of plasmalemmal caveolae in signal transduction. The American Journal of Physiology. 1998;275:L843-L851. DOI: 10.1152/ajplung.1998.275.5.L843
  58. 58. Le PU, Guay G, Altschuler Y, Nabi IR. Caveolin-1 is a negative regulator of caveolae-mediated endocytosis to the endoplasmic reticulum. The Journal of Biological Chemistry. 2002;277:3371-3379. DOI: 10.1074/jbc.M111240200
  59. 59. Parton RG, Simons K. The multiple faces of caveolae. Nature Reviews. Molecular Cell Biology. 2007;8:185-194. DOI: 10.1038/nrm2122
  60. 60. Ariotti N, Rae J, Leneva N, Ferguson C, Loo D, Okano S, et al. Molecular characterization of caveolin-induced membrane curvature. The Journal of Biological Chemistry. 2015;290:24875-24890. DOI: 10.1074/jbc.M115.644336
  61. 61. Okamoto T, Schlegel A, Scherer PE, Lisanti MP. Caveolins, a family of scaffolding proteins for organizing “preassembled signaling complexes” at the plasma membrane. The Journal of Biological Chemistry. 1998;273:5419-5422. DOI: 10.1074/jbc.273.10.5419
  62. 62. Mathew R. PDGF receptor blocker for pulmonary hypertension: A new agent in therapeutic arsenal. Expert Opinion on Investigational Drugs. 2012;21:139-142. DOI: 10.1517/13543784.2012.650861
  63. 63. Jasmin JF, Mercier I, Sotgia F, Lisanti MP. SOCS proteins and caveolin-1 as negative regulators of endocrine signaling. Trends in Endocrinology and Metabolism. 2006;17:150-158. DOI: 10.1016/j.tem.2006.03.007
  64. 64. Fujimoto T, Kogo H, Nomura R, Une T. Isoforms of caveolin-1 and caveolar structure. Journal of Cell Science. 2000;113(Pt 19):3509-3517
  65. 65. Parolini I, Sargiacomo M, Galbiati F, Rizzo G, Grignani F, Engelman JA, et al. Expression of caveolin-1 is required for the transport of caveolin-2 to the plasma membrane. Retention of caveolin-2 at the level of the golgi complex. The Journal of Biological Chemistry. 1999;274:25718-25725. DOI: 10.1074/jbc.274.36.25718
  66. 66. Xie L, Frank PG, Lisanti MP, Sowa G. Endothelial cells isolated from caveolin-2 knockout mice display higher proliferation rate and cell cycle progression relative to their wild-type counterparts. American Journal of Physiology. Cell Physiology. 2010;298:C693-C701. DOI: 10.1152/ajpcell.00401.2009
  67. 67. Razani B, Wang XB, Engelman JA, Battista M, Lagaud G, Zhang XL, et al. Caveolin-2-deficient mice show evidence of severe pulmonary dysfunction without disruption of caveolae. Molecular and Cellular Biology. 2002;22:2329-2344. DOI: 10.1128/MCB.22.7.2329-2344.2002
  68. 68. Liu L, Pilch PF. A critical role of cavin (polymerase I and transcript release factor) in caveolae formation and organization. The Journal of Biological Chemistry. 2008;283:4314-4322. DOI: 10.1074/jbc.M707890200
  69. 69. Hill MM, Bastiani M, Luetterforst R, Kirkham M, Kirkham A, Nixon SJ, et al. PTRF-Cavin, a conserved cytoplasmic protein required for caveola formation and function. Cell. 2008;132:113-124. DOI: 10.1016/j.cell.2007.11.042
  70. 70. Dávalos A, Fernández-Hernando C, Sowa G, Derakhshan B, Lin MI, Lee JY, et al. Quantitative proteomics of caveolin-1-regulated proteins: Characterization of polymerase 1 and transcript release factor/CAVIN-1 in endothelial cells. Molecular & Cellular Proteomics. 2010;9:2109-2124. DOI: 10.1074/mcp.M110.001289
  71. 71. Zhou LJ, Chen XY, Liu SP, Zhang LL, Xu YN, Mu PW, et al. Downregulation of Cavin-1 expression via increasing caveolin-1 degradation prompts the proliferation and migration of vascular smooth muscle cells in balloon injury-induced neointimal hyperplasia. Journal of American Heart Association. 2017;6(8). DOI: 10.1161/JAHA.117.005754
  72. 72. Swärd K, Sadegh MK, Mori M, Erjefält JS, Rippe C. Elevated pulmonary arterial pressure and altered expression of Ddah1 and Arg1 in mice lacking cavin-1/PTRF. Physiological Reports. 2013;1(1):e00008. DOI: 10.1002/PHY2.8
  73. 73. Liu L, Brown D, McKee M, Lebrasseur NK, Yang D, Albrecht KH, et al. Deletion of Cavin/PTRF causes global loss of caveolae, dyslipidemia, and glucose intolerance. Cell Metabolism. 2008;8:310-317. DOI: 10.1016/j.cmet.2008.07.008
  74. 74. Hansen CG, Howard G, Nichols BJ. Pacsin 2 is recruited to caveolae and functions in caveolar biogenesis. Journal of Cell Science. 2011;124:2777-2785. DOI: 10.1242/jcs.084319
  75. 75. Hoernke M, Mohan J, Larsson E, Blomberg J, Kahra D, Westenhoff S, et al. EHD2 restrains dynamics of caveolae by an ATP-dependent, membrane-bound, open conformation. Proceedings of the National Academy of Sciences. 2017;114:E4360-E4369. DOI: 10.1073/pnas.1614066114
  76. 76. Lamaze C, Tardif N, Dewulf M, Vassilopoulos S, Blouin CM. The caveolae dress code: Structure and signaling. Current Opinion in Cell Biology. 2017;47:117-125. DOI: 10.1016/j.ceb.2017.02.014
  77. 77. Drab M, Verkade P, Elger M, Kasper M, Lohn M, Lauterbach B, et al. Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice. Science. 2001;293(5539):2449-2452. DOI: 10.1126/science.1062688
  78. 78. Murata T, Lin MI, Huang Y, Yu J, Bauer PM, Giordano FJ, et al. Re-expression of caveolin-1 in endothelium rescues the vascular, cardiac, and pulmonary defects in global caveolin-1 knockout mice. The Journal of Experimental Medicine. 2007;204:2373-2382. DOI: 10.1084/jem.20062340
  79. 79. Codrici E, Albulescu L, Popescu ID, Mihai S, Enciu AM, Albulescu R, et al. Caveolin-1-knockout mouse as a model of inflammatory diseases. Journal of Immunology Research. 2018;2018:2498576. DOI: 10.1155/2018/2498576. eCollection 2018
  80. 80. Oliveira SDS, Castellon M, Chen J, Bonini MG, Gu X, Elliott MH, et al. Inflammation-induced caveolin-1 and BMPRII depletion promotes endothelial dysfunction and TGF-β-driven pulmonary vascular remodeling. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2017;312:L760-L771. DOI: 10.1152/ajplung.00484.2016
  81. 81. Cho JG, Lee A, Chang W, Lee MS, Kim J. Endothelial to mesenchymal transition represents a key link in the interaction between inflammation and endothelial dysfunction. Frontiers in Immunology. 2018;9:294. DOI: 10.3389/fimmu.2018.00294 eCollection 2018
  82. 82. Shiroto T, Romero N, Sugiyama T, Sartoretto JL, Kalwa H, Yan Z, et al. Caveolin-1 is a critical determinant of autophagy, metabolic switching, and oxidative stress in vascular endothelium. PLoS One. 2014;9:e87871. DOI: 10.1371/journal.pone.0087871. eCollection 2014
  83. 83. Michel JB, Feron O, Sacks D, Michel T. Reciprocal regulation of endothelial nitric-oxide synthase by Ca2+−calmodulin and caveolin. The Journal of Biological Chemistry. 1997;272:15583-15586. DOI: 10.1074/jbc.272.25.15583
  84. 84. Bernatchez PN, Bauer PM, Yu J, Prendergast JS, He P, Sessa WC. Dissecting the molecular control of endothelial NO synthase by caveolin-1 using cell-permeable peptides. Proceedings of the National Academy of Sciences. 2005;102:761-7666. DOI: 10.1073/pnas.0407224102
  85. 85. García-Cardeña G, Martasek P, Masters BS, Skidd PM, Couet J, Li S, et al. Dissecting the interaction between nitric oxide synthase (NOS) and caveolin. Functional significance of the NOS caveolin binding domain in vivo. The Journal of Biological Chemistry. 1997;272:25437-25440. DOI: 10.1074/jbc.272.41.25437
  86. 86. Rizzo V, McIntosh DP, Oh P, Schnitzer JE. In situ flow activates endothelial nitric oxide synthase in luminal caveolae of endothelium with rapid caveolin dissociation and calmodulin association. The Journal of Biological Chemistry. 1998;273:34724-34729. DOI: 10.1074/jbc.273.52.34724
  87. 87. Chidlow JH Jr, Sessa WC. Caveolae, caveolins, and cavins: Complex control of cellular signalling and inflammation. Cardiovascular Research. 2010;86:219-225. DOI: 10.1093/cvr/cvq075
  88. 88. Oliveira SDS, Minshall RD. Caveolin and endothelial NO signaling. Current Topics in Membranes. 2018;82:257-279. DOI: 10.1016/bs.ctm.2018.09.004
  89. 89. Tian J, Hou Y, Lu Q , Wiseman DA, Vasconcelos Fonsesca F, Elms S, et al. A novel role for caveolin-1 in regulating endothelial nitric oxide synthase activation in response to H2O2 and shear stress. Free Radical Biology & Medicine. 2010;49:159-170. DOI: 10.1016/j.freeradbiomed.2010.03.023
  90. 90. Sonveaux P, Martinive P, DeWever J, Batova Z, Daneau G, Pelat M, et al. Caveolin-1 expression is critical for vascular endothelial growth factor-induced ischemic hindlimb collateralization and nitric oxide-mediated angiogenesis. Circulation Research. 2004;95:154-161. DOI: 10.1161/01.RES.0000136344.27825.72
  91. 91. Morais C, Ebrahem Q , Anand-Apte B, Parat MO. Altered angiogenesis in caveolin-1 gene-deficient mice is restored by ablation of endothelial nitric oxide synthase. The American Journal of Pathology. 2012;180:1702-1714. DOI: 10.1016/j.ajpath.2011.12.018
  92. 92. Rath G, Dessy C, Feron O. Caveolae, caveolin and control of vascular tone: Nitric oxide (NO) and endothelium derived hyperpolarizing factor (EDHF) regulation. Journal of Physiology and Pharmacology. 2009;60(Suppl. 4):105-109
  93. 93. Goedicke-Fritz S, Kaistha A, Kacik M, Markert S, Hofmeister A, Busch C, et al. Evidence for functional and dynamic microcompartmentation of Cav-1/TRPV4/K(Ca) in caveolae of endothelial cells. European Journal of Cell Biology. 2015;94:391-400. DOI: 10.1016/j.ejcb.2015.06.002
  94. 94. Saliez J, Bouzin C, Rath G, Ghisdal P, Desjardins F, Rezzani R, et al. Role of caveolar compartmentation in endothelium-derived hyperpolarizing factor-mediated relaxation: Ca2+ signals and gap junction function are regulated by caveolin in endothelial cells. Circulation. 2008;117:1065-1074. DOI: 10.1161/CIRCULATIONAHA.107.731679
  95. 95. Spisni E, Griffoni C, Santi S, Riccio M, Marulli R, Bartolini G, et al. Colocalization prostacyclin (PGI2) synthase—Caveolin-1 in endothelial cells and new roles for PGI2 in angiogenesis. Experimental Cell Research. 2001;266:31-43. DOI: 10.1006/excr.2001.5198
  96. 96. Mathew R. Pathogenesis of pulmonary hypertension: A case for caveolin-1 and cell membrane integrity. American Journal of Physiology. Heart and Circulatory Physiology. 2014;306:H15-H25. DOI: 10.1152/ajpheart.00266.2013
  97. 97. Zeng Y. Endothelial glycocalyx as a critical signalling platform integrating the extracellular haemodynamic forces and chemical signalling. Journal of Cellular and Molecular Medicine. 2017;21:1457-1462. DOI: 10.1111/jcmm.13081
  98. 98. Harding IC, Mitra R, Mensah SA, Herman IM, Ebong EE. Pro-atherosclerotic disturbed flow disrupts caveolin-1 expression, localization, and function via glycocalyx degradation. Journal of Translational Medicine. 2018;16:364. DOI: 10.1186/s12967-018-1721-2
  99. 99. Guo J, Yang ZC, Liu Y. Attenuating pulmonary hypertension by protecting the integrity of glycocalyx in rats model of pulmonary artery hypertension. Inflammation. 2019. DOI: 10.1007/s10753-019-01055-5
  100. 100. Gavard J. Endothelial permeability and VE-cadherin: A wacky comradeship. Cell Adhesion & Migration. 2014;8:158-164. DOI: 10.4161/cam.29026
  101. 101. Kronstein R, Seebach J, Grossklaus S, Minten C, Engelhardt B, Drab M, et al. Caveolin-1 opens endothelial cell junctions by targeting catenins. Cardiovascular Research. 2012;93:130-140. DOI: 10.1093/cvr/cvr256
  102. 102. Caveda L, Martin-Padura I, Navarro P, Breviario F, Corada M, Gulino D, et al. Inhibition of cultured cell growth by vascular endothelial cadherin (cadherin-5/VE-cadherin). The Journal of Clinical Investigation. 1996;98:886-893. DOI: 10.1172/JCI118870
  103. 103. Song L, Ge S, Pachter JS. Caveolin-1 regulates expression of junction-associated proteins in brain microvascular endothelial cells. Blood. 2007;109:1515-1523. DOI: 10.1182/blood-2006-07-034009
  104. 104. Dejana E, Giampietro C. Vascular endothelial-cadherin and vascular stability. Current Opinion in Hematology. 2012;9:218-223. DOI: 10.1097/MOH.0b013e3283523e1c
  105. 105. Vestweber D. VE-cadherin: The major endothelial adhesion molecule controlling cellular junctions and blood vessel formation. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28:223-232. DOI: 10.1161/ATVBAHA.107.158014
  106. 106. Le Guelte A, Dwyer J, Gavard J. Jumping the barrier: VE-cadherin, VEGF and other angiogenic modifiers in cancer. Biology of the Cell. 2011;103:593-605. DOI: 10.1042/BC20110069
  107. 107. Nikitopoulou I, Orfanos SE, Kotanidou A, Maltabe V, Manitsopoulos N, Karras P, et al. Vascular endothelial-cadherin downregulation as a feature of endothelial transdifferentiation in monocrotaline-induced pulmonary hypertension. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2016;311:L352-L363. DOI: 10.1152/ajplung.00156.2014
  108. 108. Privratsky JR, Newman PJ. PECAM-1: Regulator of endothelial junctional integrity. Cell and Tissue Research. 2014;355:607-619. DOI: 10.1007/s00441-013-1779-3
  109. 109. Burton VJ, Ciuclan LI, Holmes AM, Rodman DM, Walker C, Budd DC. Bone morphogenetic protein receptor II regulates pulmonary artery endothelial cell barrier function. Blood. 2011;117:333-341. DOI: 10.1182/blood-2010-05-285973
  110. 110. Mathew R. Cell-specific dual role of caveolin-1 in pulmonary hypertension. Pulmonary Medicine. 2011;2011:573432. DOI: 10.1155/2011/573432
  111. 111. Kim J, Hwangbo C, Hu X, Kang Y, Papangeli I, Mehrotra D, et al. Restoration of impaired endothelial myocyte enhancer factor 2 function rescues pulmonary arterial hypertension. Circulation. 2015;131:190-199. DOI: 10.1161/CIRCULATIONAHA.114.013339
  112. 112. Huertas A, Tu L, Thuillet R, Le Hiress M, Phan C, Ricard N, et al. Leptin signalling system as a target for pulmonary arterial hypertension therapy. The European Respiratory Journal. 2015;45:1066-1080. DOI: 10.1183/09031936.00193014
  113. 113. Tu L, Dewachter L, Gore B, Fadel E, Dartevelle P, Simonneau G, et al. Autocrine fibroblast growth factor-2 signaling contributes to altered endothelial phenotype in pulmonary hypertension. American Journal of Respiratory Cell and Molecular Biology. 2011;45:311-322. DOI: 10.1165/rcmb.2010-0317OC
  114. 114. Le Hiress M, Tu L, Ricard N, Phan C, Thuillet R, Fadel E, et al. Proinflammatory signature of the dysfunctional endothelium in pulmonary hypertension. Role of the macrophage migration inhibitory factor/CD74 complex. American Journal of Respiratory and Critical Care Medicine. 2015;192(8):983-997. DOI: 10.1164/rccm.201402-0322OC
  115. 115. Jasmin JF, Mercier I, Hnasko R, Cheung MW, Tanowitz HB, Dupuis J, et al. Lung remodeling and pulmonary hypertension after myocardial infarction: Pathogenic role of reduced caveolin expression. Cardiovascular Research. 2004;63:747-755. DOI: 10.1016/j.cardiores.2004.05.018
  116. 116. Achcar RO, Demura Y, Rai PR, Taraseviciene-Stewart L, Kasper M, Voelkel NF, et al. Loss of caveolin and heme oxygenase expression in severe pulmonary hypertension. Chest. 2006;129:696-705. DOI: 10.1378/chest.129.3.696
  117. 117. Huang J, Wolk JH, Gewitz MH, Mathew R. Caveolin-1 expression during the progression of pulmonary hypertension. Experimental Biology and Medicine. 2012;237:956-965. DOI: 10.1258/ebm.2012.011382
  118. 118. Mathew R, Guzowski DE, Gloster ES. Effect of verapamil on monocrotaline-induced pulmonary artery hypertension and endothelial cell dysfunction in rats. Experimental Lung Research. 1990;16:627-644
  119. 119. Huang J, Kaminski PM, Edwards JG, Yeh A, Wolin MS, Frishman WH, et al. Pyrrolidine dithiocarbamate restores endothelial cell membrane integrity and attenuates monocrotaline-induced pulmonary artery hypertension. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2008;294:L1250-L1259. DOI: 10.1152/ajplung.00069.2007
  120. 120. Jasmin JF, Mercier I, Dupuis J, Tanowitz HB, Lisanti MP. Short-term administration of a cell-permeable caveolin-1 peptide prevents the development of monocrotaline-induced pulmonary hypertension and right ventricular hypertrophy. Circulation. 2006;114(9):912-920. DOI: 10.1161/CIRCULATIONAHA.106.634709
  121. 121. Mathew R. Possible mechanism of neointima formation in pulmonary arterial hypertension. Blood, Heart and Circulation. 2018;2:1-5. DOI: 10.15761/BHC1000145
  122. 122. Huang J, Wolk JH, Gewitz MH, Loyd JE, West J, Austin ED, et al. Enhanced caveolin-1 expression in smooth muscle cells: Possible prelude to neointima formation. World Journal of Cardiology. 2015;7:671-684. DOI: 10.4330/wjc.v7.i10.671
  123. 123. Dereddy N, Huang J, Erb M, Guzel S, Wolk JH, Sett SS, et al. Associated inflammation or increased flow-mediated shear stress, but not pressure alone, disrupts endothelial caveolin-1 in infants with pulmonary hypertension. Pulmonary Circulation. 2012;2:492-500. DOI: 10.4103/2045-8932.105038
  124. 124. Mathew R, Huang J, Katta US, Krishnan U, Sandoval C, Gewitz MH. Immunosuppressant-induced endothelial damage and pulmonary arterial hypertension. Journal of Pediatric Hematology/Oncology. 2011;33:55-58. DOI: 10.1097/MPH.0b013e3181ec0ede
  125. 125. Patel HH, Zhang S, Murray F, Suda RY, Head BP, Yokoyama U, et al. Increased smooth muscle cell expression of caveolin-1 and caveolae contribute to the pathophysiology of idiopathic pulmonary arterial hypertension. The FASEB Journal. 2007;21(11):2970-2979. DOI: 10.1096/fj.07-8424com
  126. 126. Huang J, Mathew R. Loss of cavin1 and expression of p-caveolin-1 in pulmonary hypertension: Possible role in neointima formation. World Journal of Hypertension. 2019;9:17-29. DOI: 10.5494/wjh.v9.i2.17
  127. 127. Ortiz R, Díaz J, Díaz N, Lobos-Gonzalez L, Cárdenas A, Contreras P, et al. Extracellular matrix-specific Caveolin-1 phosphorylation on tyrosine 14 is linked to augmented melanoma metastasis but not tumorigenesis. Oncotarget. 2016;7:40571-40593. DOI: 10.18632/oncotarget.9738
  128. 128. Joshi B, Strugnell SS, Goetz JG, Kojic LD, Cox ME, Griffith OL, et al. Phosphorylated caveolin-1 regulates rho/ROCK-dependent focal adhesion dynamics and tumor cell migration and invasion. Cancer Research. 2008;68:8210-8220. DOI: 10.1158/0008-5472.CAN-08-0343
  129. 129. Sakao S, Tatsumi K, Voelkel NF. Reversible or irreversible remodeling in pulmonary arterial hypertension. American Journal of Respiratory Cell and Molecular Biology. 2010;43:629-634. DOI: 10.1165/rcmb.2009-0389TR
  130. 130. Okada K, Tanaka Y, Bernstein M, Zhang W, Patterson GA, Botney MD. Pulmonary hemodynamics modify the rat pulmonary artery response to injury. A neointimal model of pulmonary hypertension. The American Journal of Pathology. 1997;151:1019-1025
  131. 131. Tanaka Y, Schuster DP, Davis EC, Patterson GA, Botney MD. The role of vascular injury and hemodynamics in rat pulmonary artery remodeling. The Journal of Clinical Investigation. 1996;98:434-442
  132. 132. Dammann JF Jr, McEachen JA, Thompson WM Jr, Smith R, Muller WH Jr. The regression of pulmonary vascular disease after the creation of pulmonary stenosis. The Journal of Thoracic and Cardiovascular Surgery. 1961;42:722-734
  133. 133. Wagenvoort CA, Wagenvoort N, Draulans-Noë Y. Reversibility of plexogenic pulmonary arteriopathy following banding of the pulmonary artery. The Journal of Thoracic and Cardiovascular Surgery. 1984;87:876-886
  134. 134. Sakao S, Taraseviciene-Stewart L, Lee JD, Wood K, Cool CD, Voelkel NF. Initial apoptosis is followed by increased proliferation of apoptosis-resistant endothelial cells. The FASEB Journal. 2005;19(9):1178-1180. DOI: 10.1096/fj.04-3261fje
  135. 135. Bull TM, Golpon H, Hebbel RP, Solovey A, Cool CD, Tuder RM, et al. Circulating endothelial cells in pulmonary hypertension. Thrombosis and Haemostasis. 2003;90:698-703. DOI: 10.1160/TH03-04-0251
  136. 136. Li X, Qiu J, Pan M, Zheng D, Su Y, Wei M, et al. Correlation between congenital heart disease complicated with pulmonary artery hypertension and circulating endothelial cells as well as endothelin-1. International Journal of Clinical and Experimental Pathology. 2015;8:10743-10751 eCollection 2015
  137. 137. Levy M, Bonnet D, Mauge L, Celermajer DS, Gaussem P, Smadja DM. Circulating endothelial cells in refractory pulmonary hypertension in children: Markers of treatment efficacy and clinical worsening. PLoS One. 2013;8(6):e65114. DOI: 10.1371/journal.pone.0065114
  138. 138. Kawut SM, Horn EM, Berekashvili KK, Widlitz AC, Rosenzweig EB, Barst RJ. von Willebrand factor independently predicts long-term survival in patients with pulmonary arterial hypertension. Chest. 2005;128:2355-2362. DOI: 10.1378/chest.128.4.2355
  139. 139. Smadja DM, Mauge L, Sanchez O, Silvestre JS, Guerin C, Godier A, et al. Distinct patterns of circulating endothelial cells in pulmonary hypertension. The European Respiratory Journal. 2010;36:1284-1293. DOI: 10.1183/09031936.00130809
  140. 140. Yu W, Liu Z, An S, Zhao J, et al. The endothelial-mesenchymal transition (EndMT) and tissue regeneration. Current Stem Cell Research & Therapy. 2014;9:196-204
  141. 141. Bakhshi FR, Mao M, Shajahan AN, Piegeler T, Chen Z, Chernaya O, et al. Nitrosation-dependent caveolin 1 phosphorylation, ubiquitination, and degradation and its association with idiopathic pulmonary arterial hypertension. Pulmonary Circulation. 2013;3(4):816-830. DOI: 10.1086/674753
  142. 142. Li Z, Wermuth PJ, Benn BS, Lisanti MP, Jimenez SA. Caveolin-1 deficiency induces spontaneous endothelial-to-mesenchymal transition in murine pulmonary endothelial cells in vitro. The American Journal of Pathology. 2013;182:325-331
  143. 143. Coll-Bonfill N, Musri MM, Ivo V, Barberà JA, Tura-Ceide O. Transdifferentiation of endothelial cells to smooth muscle cells play an important role in vascular remodelling. American Journal of Stem Cells. 2015;4:13-21
  144. 144. Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Péchoux C, Bogaard HJ, et al. Endothelial-to-mesenchymal transition in pulmonary hypertension. Circulation. 2015;131:1006-1018. DOI: 10.1161/CIRCULATIONAHA.114.008750
  145. 145. Hopper RK, Moonen JR, Diebold L, Cao A, Rhodes CJ, Tojais NF, et al. In pulmonary arterial hypertension, reduced BMPR2 promotes endothelial-to-mesenchymal transition via HMGA1 and its target slug. Circulation. 2016;133:1783-1794. DOI: 10.1161/CIRCULATIONAHA.115.020617
  146. 146. Díez M, Musri MM, Ferrer E, Barberà JA, Peinado VI. Endothelial progenitor cells undergo an endothelial-to-mesenchymal transition-like process mediated by TGFbetaRI. Cardiovascular Research. 2010;88:502-511. DOI: 10.1093/cvr/cvq236
  147. 147. Rabinovitch M, Gamble W, Nadas AS, Miettinen OS, Reid L. Rat pulmonary circulation after chronic hypoxia: Hemodynamic and structural features. The American Journal of Physiology. 1979;236:H818-H827. DOI: 10.1152/ajpheart.1979.236.6.H818
  148. 148. Adnot S, Raffestin B, Eddahibi S, Braquet P, Chabrier PE. Loss of endothelium-dependent relaxant activity in the pulmonary circulation of rats exposed to chronic hypoxia. The Journal of Clinical Investigation. 1991;87:155-162. DOI: 10.1172/JCI114965
  149. 149. Murata T, Yamawaki H, Hori M, Sato K, Ozaki H, Karaki H. Hypoxia impairs endothelium-dependent relaxation in organ cultured pulmonary artery. European Journal of Pharmacology. 2001;421:45-53. DOI: 10.1016/s0014-2999(01)01015-9
  150. 150. Murata T, Sato K, Hori M, Ozaki H, Karaki H. Decreased endothelial nitric-oxide synthase (eNOS) activity resulting from abnormal interaction between eNOS and its regulatory proteins in hypoxia-induced pulmonary hypertension. Journal of Biological Chemistry. 2002;15(277):44085-44092. 3434. DOI: 10.1074/jbc.M205934200
  151. 151. Gratton JP, Fontana J, O’Connor DS, Garcia-Cardena G, McCabe TJ, Sessa WC. Reconstitution of an endothelial nitric-oxide synthase (eNOS), hsp90, and caveolin-1 complex in vitro. Evidence that hsp90 facilitates calmodulin stimulated displacement of eNOS from caveolin-1. The Journal of Biological Chemistry. 2000;275:22268-22272. DOI: 10.1074/jbc.M001644200
  152. 152. Fike CD, Pfister SL, Slaughter JC, Kaplowitz MR, Zhang Y, Zeng H, et al. Protein complex formation with heat shock protein 90 in chronic hypoxia-induced pulmonary hypertension in newborn piglets. The American Journal of Physiology. 2010;299:H1190-H1204. DOI: 10.1152/ajpheart.01207.2009
  153. 153. Murata T, Lin MI, Stan RV, Bauer PM, Yu J, Sessa WC. Genetic evidence supporting caveolae microdomain regulation of calcium entry in endothelial cells. The Journal of Biological Chemistry. 2007;282:16631-16643. DOI: 10.1074/jbc.M607948200
  154. 154. Paffett ML, Naik JS, Riddle MA, Menicucci SD, Gonzales AJ, Resta TC, et al. Altered membrane lipid domains limit pulmonary endothelial calcium entry following chronic hypoxia. American Journal of Physiology—Heart and Circulatory Physiology. 2011;301:H1331-H1340. DOI: 10.1152/ajpheart.00980.2010
  155. 155. Murata T, Kinoshita K, Hori M, Kuwahara M, Tsubone H, Karaki H, et al. Statin protects endothelial nitric oxide synthase activity in hypoxia-induced pulmonary hypertension. Arteriosclerosis, Thrombosis, and Vascular Biology. 2005;25:2335-2342. DOI: 10.1161/01.ATV.0000186184.33537.48
  156. 156. Penaloza D, Arias-Stella J. The heart and pulmonary circulation at high altitudes: Healthy highlanders and chronic mountain sickness. Circulation. 2007;115:1132-1146. DOI: 10.1161/CIRCULATIONAHA.106.624544
  157. 157. Blanco I, Piccari L, Barberà JA. Pulmonary vasculature in COPD: The silent component. Respirology. 2016;21:984-994. DOI: 10.1111/resp.12772
  158. 158. Sakao S, Voelkel NF, Tatsumi K. The vascular bed in COPD: Pulmonary hypertension and pulmonary vascular alterations. European Respiratory Review. 2014;23:350-355. DOI: 10.1183/09059180.00007913
  159. 159. Peinado VI, Barbera JA, Ramirez J, Gomez FP, Roca J, Jover L, et al. Endothelial dysfunction in pulmonary arteries of patients with mild COPD. The American Journal of Physiology. 1998;274:L908-L913. DOI: 10.1152/ajplung.1998.274.6.L908
  160. 160. Huber LC, Soltermann A, Fischler M, Gay S, Weder W, Russi EW, et al. Caveolin-1 expression and hemodynamics in COPD patients. Open Respiratory Medicine Journal. 2009;3:73-78. DOI: 10.2174/1874306400903010073
  161. 161. Carlsen J, Hasseriis Andersen K, Boesgaard S, Iversen M, Steinbrüchel D, Bøgelund AC. Pulmonary arterial lesions in explanted lungs after transplantation correlate with severity of pulmonary hypertension in chronic obstructive pulmonary disease. The Journal of Heart and Lung Transplantation. 2013;32:347-354. DOI: 10.1016/j.healun.2012.11.014

Written By

Rajamma Mathew

Submitted: 02 May 2019 Reviewed: 18 March 2020 Published: 04 May 2020