Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jan 1.
Published in final edited form as: J Mol Cell Cardiol. 2007 Sep 20;44(1):14–30. doi: 10.1016/j.yjmcc.2007.09.006

Pathogenic Mechanisms of Pulmonary Arterial Hypertension

Stephen Y Chan 1,3, Joseph Loscalzo 2,3
PMCID: PMC2234575  NIHMSID: NIHMS38868  PMID: 17950310

Abstract

Pulmonary arterial hypertension (PAH)1 is a complex disease that causes significant morbidity and mortality and is clinically characterized by an increase in pulmonary vascular resistance. The histopathology is marked by vascular proliferation/fibrosis, remodeling, and vessel obstruction. Development of PAH involves the complex interaction of multiple vascular effectors at all anatomic levels of the arterial wall. Subsequent vasoconstriction, thrombosis, and inflammation ensue, leading to vessel wall remodeling and cellular hyperproliferation as the hallmarks of severe disease. These processes are influenced by genetic predisposition as well as diverse endogenous and exogenous stimuli. Recent studies have provided a glimpse at certain molecular pathways that contribute to pathogenesis; these have led to the identification of attractive targets for therapeutic intervention. We will review our current understanding of the mechanistic underpinnings of the genetic and exogenous/acquired triggers of PAH. The resulting imbalance of vascular effectors provoking pathogenic vascular changes will also be discussed, with an emphasis on common and overarching regulatory pathways that may relate to the primary triggers of disease. The current conceptual framework should allow for future studies to refine our understanding of the molecular pathogenesis of PAH and improve the therapeutic regimen for this disease.

Keywords: pulmonary arterial hypertension, molecular mechanism, vasoconstriction, thrombosis, proliferation, genetic association, acquired risk, vascular effector

Introduction

Pulmonary arterial hypertension (PAH) is clinically characterized by increasing pulmonary arterial pressure in the absence of elevated left heart pressure. If untreated, PAH leads to right ventricular failure, volume overload, and death [1]. No cure is available for PAH, but recent advances in our understanding of the molecular mechanisms of disease progression have led to therapeutic approaches that improve survival and quality of life [2].

Previously, PAH had been classified as a primary form (“idiopathic”) or secondary forms, associated with a variety of diverse clinical entities. Recently, investigators have increasingly recognized that secondary disease states may precipitate pulmonary vascular disease resembling idiopathic PAH in histopathologic changes. This suggests a degree of commonality in mechanisms of disease progression. Currently, the term PAH comprises the idiopathic forms (formerly, primary PAH) as well as manifestations resulting from other disease, such as collagen vascular disease, hemoglobinopathies, congenital cardiovascular disease with systemic-to-pulmonary shunts, human immunodeficiency virus infection, portal hypertension, drugs and toxins, persistent pulmonary hypertension of the newborn, and myeloproliferative disorders, among others [3]. While right ventricular dysfunction certainly contributes to clinical decompensation, the inciting pathogenic processes appear localized to the pulmonary vasculature itself. Common histologic features in nearly all types of PAH occur at the level of the small peripheral pulmonary arteries; these include intimal fibrosis, distal localization and proliferation of vascular smooth muscle, and pulmonary arterial occlusion [4]. Furthermore, a hallmark of severe, end-stage disease is the formation of a vessel “neointima,” characterized by increased deposition of extracellular matrix and myofibroblasts [5]. Plexiform lesions can predominate, characterized by over-proliferation of endothelial-like cells encroaching upon the vessel lumen [6].

Multiple cell types in the pulmonary arterial wall and pulmonary arterial circulation contribute to the specific response to injury and the development of vessel remodeling (Figure 1) [5]. The endothelium serves as a central sensor of injurious stimuli, such as hypoxia, shear stress, inflammation, and toxins. Dysregulation of downstream vascular effectors may be the result of initial endothelial cell injury or dysfunction; however, more recent studies suggest that dysfunction of alternative vascular components, such as adventitial fibroblasts or components of the extracellular matrix, may also initiate pulmonary vascular disease [7]. Regardless of the initiating process, an imbalance of secreted vasoactive factors ensues and directs vascular remodeling via pathologic cellular processes. These include excessive cell proliferation, vasoconstriction, and thrombosis, which are associated with more complex patterns of inflammation and angiogenesis. Transdifferentiation of endothelial cells to vascular smooth muscle cells may also contribute [8, 9]. Furthermore, the identification of monoclonal amplification in plexiform populations [10] has led to speculation of a model of end-stage PAH similar to that of progression to cancer, with dysregulation of the cell-cycle and apoptosis as predominant features [11]. Inflammatory cells and activated platelets appear to predominate in later stages of PAH [12]; yet, our understanding is limited regarding the mechanistic role of these cellular populations in disease progression [13]. Finally, circulating or resident progenitor cells have been proposed to factor significantly in vessel wall injury and repair; dysregulation of these functions may also contribute to PAH [14].

Figure 1. Pulmonary Arterial Pathobiology Involves the Coordinate Action of Multiple Cell Types.

Figure 1

The histologic progression of the pulmonary vasculature from quiescence to pathogenic activation in PAH involves numerous vascular cell types and phenotypic responses. Initial injury to the endothelium and/or adventitial fibroblasts may activate pathogenic signaling pathways. These result in an imbalance of secreted vascular effectors that drive the vascular responses of vasoconstriction, proliferation, and dysregulation of apoptosis, leading to the formation of a layer of “neointima” (red arrow) and, in some cases, plexiform lesions. Activation of platelets (blue thrombus) and extravasation/migration of blood-borne inflammatory cells (green arrow) likely play prominent roles in these processes, but their exact mechanistic actions are unclear. Engraftment and differentiation of vascular progenitor cells may influence disease progression (purple arrow). Transdifferentiation of endothelial cells to vascular smooth muscle cells may contribute as well (blue arrow). Micrographs of pulmonary arteries are courtesy of www.scleroderma.org and Humbert et al., Treatment of Pulmonary Arterial Hypertension, New England Journal of Medicine, 2004, 351(14):1425-1436; Copyright 2004, Massachusetts Medical Society. All rights reserved.

Based on observation of these histologic changes in humans and animal models, a number of recent advances have allowed for the development of a conceptual framework to better delineate the molecular pathways of PAH. In this review, we will highlight our current understanding of the complex and incompletely characterized interplay of genetic and exogenous upstream stimuli with downstream vascular effectors that ultimately lead to clinical PAH.

Genetic Association

An understanding of the mechanism of genetic predisposition to PAH is of paramount importance for identification of the root pathogenesis. The familial variety of idiopathic PAH accounts for at least 6 percent of all cases of PAH [15-17]. Pedigree analysis has demonstrated an autosomal dominant inheritance but with variable penetrance, as only 10-20% of putative genetic carriers develop clinical PAH. Genetic anticipation is present, as each successive generation of affected families is afflicted at a younger age and greater severity compared with the preceding generation.

Mutations in the transforming growth factor-β receptor (TGF-β receptor) superfamily have been genetically linked to PAH and likely play a causative role in the development of disease. A rare group of patients with hereditary hemorrhagic telangiectasia and idiopathic PAH harbor specific mutations in ALK1 or endoglin, genes encoding for two such members of the TGF- β receptor superfamily [18, 19]. However, a more prevalent cohort of patients carries mutations in another member, the bone morphogenetic protein receptor type 2 (BMPR2 gene which encodes for BMPR-II) [20, 21]. Over 140 mutations in BMPR2 have been reported in patients with familial PAH [22], mainly located in the extracellular ligand-binding domain, in the cytoplasmic serine/threonine kinase domain, or in the long carboxyterminal domain [23]. These account for 70% of all familial pedigrees of PAH and 10-30% of idiopathic PAH cases [16, 20, 23-26]. BMPR2 loss-of-function mutations have only been found in the heterozygous state. This fact likely reflects the critical role of the BMP pathway in vascular development, as demonstrated by the embryonic lethality of the BMPR2-homozygous null mouse [27]. The absence of BMPR2 mutations in some familial cohorts and in most of the sporadic cases indicates that additional, unidentified genetic mutations can also predispose to development of PAH. Furthermore, the presence of incomplete penetrance and genetic anticipation suggests that BMPR2 mutations are necessary but insufficient alone to result in clinically significant disease.

The mechanism of action of BMPR-II is complex, and its role in PAH progression is still unclear. It functions as a receptor with serine/threonine kinase activity, and it activates a broad and complex range of intracellular signaling pathways (as reviewed in [28]). Upon binding one of many possible BMP ligands, BMPR-II forms a heterodimer with one of three type-I receptors. BMPR-II phosphorylates the bound type-I receptor, which, in turn, phosphorylates one of the Smad family of proteins to allow for nuclear translocation, binding to DNA, and regulation of gene transcription. Alternatively, BMPR-II activation can also lead to signaling via the LIM kinase pathway, the p38/MAP kinase/ERK/JNK pathways, or the c-Src pathway [29], independent of Smad activation [30, 31].

Correlating with these complexities of signaling, the cellular effects of BMPR-II activation are multiform. In the adult, BMPR-II is expressed predominantly in the pulmonary endothelium, medial smooth muscle cells, and macrophages [32]. The mutations in BMPR2 that lead to PAH likely exert their pathogenic action predominantly by modulating function in endothelium and/or smooth muscle; however, it is possible that specific effects in other tissues or at other times in development may, in part, explain the mechanism of genetic predisposition. Under normal conditions, BMP ligands bind BMPR-II to suppress the growth of vascular smooth muscle cells [33, 34]. In contrast, binding of BMP2 and BMP7 to BMPR-II in pulmonary endothelium leads to protection from apoptosis [35]. While certain BMPR2 mutations may affect receptor function differently [36-39], altered function in vivo does not stem from somatic mutations leading to homozygous loss-of-function mutations in both BMPR2 alleles [40]. Rather, patients harboring such mutations generally suffer from haploinsufficiency with decreased expression of BMPR-II in pulmonary tissue [22, 26, 32, 41]. As a result, BMP signaling appears altered but not completely abolished [42, 43].

Given these findings, a widely held hypothesis contends that failure of the suppressive effects of BMP ligands on vascular smooth muscle [44] and failure of the protective effects of BMP ligands on endothelium may trigger vascular proliferation and remodeling. Accordingly, in vascular smooth muscle cells derived from patients with familial PAH harboring BMPR2 mutations, exposure of BMP ligands does not suppress proliferation [39, 44, 45]. Furthermore, unlike the response in wildtype endothelium, exposure of endothelial cells cultured from patients with idiopathic PAH to BMP2 does not protect against apoptosis [35]. These dysfunctional signaling pathways have been corroborated in some rodent models of PAH [46]. In correlation, pulmonary levels of BMPR-II are reduced both in familial cases of PAH without any BMPR2 mutation and in cases of secondary PAH [32]. As a result, abnormal growth responses to BMP/TGF- β stimulation have been noted in pulmonary vascular smooth muscle cells derived from PAH patients [47]. Furthermore, response of pulmonary vascular smooth muscle cells to BMP signaling appears regulated by hypoxia, a known precipitant of pulmonary hypertension [48]. Thus, dysregulation of the BMP signaling pathway may be a common pathogenic finding in multiple types of PAH due to genetic or exogenous stimuli (Figure 2).

Figure 2. Common Mechanisms Promoting PAH May Rely upon the Intersecting BMPR-II and Serotoninergic Pathways.

Figure 2

In the serotoninergic pathway, both genetic predisposition (i.e., platelet storage pool disease, perhaps 5-HT2B mutations, or perhaps the L-allele variant of 5-HTT) and specific exogenous stimuli (i.e., hypoxia, anorexigens) lead to vessel remodeling via increased serotonin levels, increased 5-HTT activity, and/or increased serotonin receptor signaling. In the BMPR-II pathway, genetic predisposition (such as heterozygous loss-of-function +/− BMPR2 mutations) and, perhaps, exogenous stimuli lead to decreased receptor expression, alteration of downstream signaling, and resulting vessel remodeling. Additional pathogenic mechanisms may upregulate the angiopoietin-1/Tie2 receptor pathway with resulting repression of BMPR-1A function and alteration of BMPR-II signaling. Interestingly, serotonin can directly upregulate angiopoietin-1 and can modulate downstream Smad function, thereby influencing the BMP signaling cascade. This functional intersection has yet to be fully described, but may represent a common, unifying mechanism of pathogenesis.

Despite these advances, a clear mechanistic explanation of the impact of BMPR2 mutations on pathogenesis is still lacking, owing to a number of factors. First, the cellular effects of the BMP pathway vary in the setting of diverse combinations of extracellular ligands, receptors, and intracellular signaling cascades [43]. Therefore, responses in cell culture may not accurately reflect in vivo events that are coordinately regulated by multiple intersecting signaling pathways. Second, the definitive in vivo effects of these mutations have been difficult to decipher. While adenoviral overexpression of wildtype BMPR-II appears to ameliorate pulmonary hypertension in a hypoxic rat model [49], overexpression of BMPR-II fails to attenuate PAH that is chemically induced (via monocrotaline) in a rat model [50]. More definitive in vivo studies have been limited. Specifically, mouse models harboring specific BMPR2 heterozygous mutations have failed to exhibit robust PAH under static conditions [38, 51], again suggesting that dysfunctional BMPR-II is likely insufficient alone to cause disease. Provocation with hypoxia has led to minimally increased pulmonary vascular reaction, while exposure to an experimental inflammatory stimulus or additional cytokines has provoked graded vascular responses [52]. Murine models harboring different BMPR2 mutations may show promise with a more robust phenotype [53]. Yet, until genetically tractable animal models of disease become readily available, the precise pathogenic contribution of the BMP pathway to PAH will likely remain elusive.

Similar to the BMP pathway, the serotonin (5-hydroxytryptamine or 5-HT) signaling pathway has also been implicated as a potential causative factor in PAH through observation of genetic and physiologic influences [54]. Serotonin is both a vasoconstrictor and mitogen that promotes smooth muscle hyperplasia and hypertrophy [55]. Primarily stored in platelet granules, secreted serotonin binds G-protein-coupled serotonin receptors present on pulmonary artery smooth muscle cells. Activation of these receptors leads to a decrease in adenylyl cyclase and cyclic AMP, resulting in an increase in contraction. Furthermore, the cell-surface serotonin transporter (5-HTT) allows for transport of extracellular serotonin into the cytoplasm of smooth muscle cells, thereby activating cellular proliferation directly through the action of serotonin or indirectly via potential pleiotropic mechanisms [56].

A number of observations in idiopathic and familial disease, congenital disease, and environmental exposure have implicated the proliferative effects of serotonin in PAH. In idiopathic PAH, pulmonary expression of serotonin receptors is increased [57, 58], and plasma levels of serotonin are chronically elevated [59]. A mouse model of hypoxic pulmonary hypertension parallels these changes [58]. A positive association has been noted among patients with congenital platelet defects in serotonin uptake (i.e., delta storage pool disease) and development of PAH [60]. Chronic exposure to anorexigens, such dexfenfluramine (an inhibitor of serotonin reuptake and stimulator of serotonin secretion), has led to increased levels of circulating, free serotonin. In mice, these changes are accompanied by increased 5-HT receptor type 2B response [58] and inhibition of 5-HTT responses. In turn, these changes correlate with an increased risk for development of PAH in humans [61]. Finally, the L-allelic variant of 5-HTT is associated with increased expression of the transporter and enhanced smooth muscle proliferation; in homozygous form, this variant has been reported in 65% of idiopathic PAH patients but only in 27% of controls, suggesting an increased risk of PAH in the homozygous population [62, 63]. A larger study of 5-HTT polymorphisms, however, has not confirmed this association [64].

Animal models of pulmonary hypertension have also implicated the activated serotonin pathway in disease progression. Treatment with serotonin and chronic hypoxia in a rat model has led to worsened hemodynamics and increased vessel remodeling [65]. Similarly, overexpression of the 5-HTT gene in mice has resulted in spontaneous development of PAH in the absence of hypoxia and exaggeration of pulmonary hypertension after hypoxic stimulus [66, 67]. Conversely, vessel remodeling and hypoxic pulmonary hypertension are reduced in a 5-HT1B receptor-null mouse [68] and are abrogated in a 5-HT2B receptor-null mouse [58].

As a result, serotonin signaling modulates pulmonary smooth muscle function in both normal and disease states and likely contributes to disease progression of PAH. However, the exact contribution to this mechanism remains to be characterized. For example, selective serotonin re-uptake inhibitors (SSRIs), which increase serotonin levels but inhibit serotonin transport, abrogate, rather than enhance, development of PAH in the setting of hypoxia [69]. In contrast, a possible causative association between maternal SSRI use and persistent pulmonary hypertension of the newborn has been reported [70]. As a result, it is currently unclear if serotonin itself or its associated effectors primarily drive PAH development. Serotonin can also influence other regulatory pathways involved in PAH progression; notably, serotonin can inhibit BMP signaling via modulation of downstream Smad proteins. In correlation, exposure to increased serotonin has led to worsened pulmonary hypertension in a BMPR2 +/− heterozygote murine model [71]. Serotonin may also regulate the angiopoietin-1/Tie2 signaling pathway, another potential pathogenic contributor (see discussion below). This, in turn, can modulate signaling of BMPR-II via inhibition of endothelial expression of one of its dimerized partners, BMPR-IA [72]. Taken together, these data not only further implicate the serotoninergic and BMP pathways as pathogenic factors, but also represent useful evidence of potential common regulatory pathways in this complex disease (Figure 2).

Acquired/Exogenous Factors

In addition to genetic predisposition, development of PAH likely depends on a variety of physiologic, acquired, and/or exogenous stimuli. Some of these factors have been studied to a sufficient degree to hypothesize a potential pathogenic mechanism(s) (Figure 3). These include chronic hypoxia, hemoglobinopathies, autoimmune vascular disease, viral infections, congenital heart disease with systemic-to-pulmonary shunt, and “serotoninergic” anorexigen use (see discussion above). Factors associated with PAH that perhaps carry even less defined mechanisms of disease include thrombocytosis, central nervous system stimulants, portal hypertension, persistent pulmonary hypertension of the newborn, and female gender predilection.

Figure 3. Physiologic/Exogenous Triggers of Pulmonary Arterial Hypertension Are Poorly Understood at the Molecular Level.

Figure 3

While most of these clinical risk factors have been known for years, the mechanisms that lead to the imbalance of vascular mediators and dysregulated cellular phenotypes are unclear.

The pulmonary vascular response to hypoxia has been well studied in cell culture and in animal models (as reviewed in [73, 74]), but its impact on PAH is unclear. In general, pulmonary vascular responses in acute and chronic hypoxia likely allow for the propagation of PAH, and, therefore, may contribute to the later stages of disease. Acute hypoxia induces vasodilatation in systemic vessels, but induces vasoconstriction in pulmonary arteries. This acute and reversible effect is mediated in part by upregulation of vasoconstrictors, such as endothelin-1 and serotonin, and, in part, by hypoxia- and redox-sensitive potassium channel activity in pulmonary vascular smooth muscle cells. Coordinately, these events lead to membrane depolarization in smooth muscle cells, increase in cytosolic calcium, and vasoconstriction [75]. In contrast, chronic hypoxia induces vascular remodeling and less reversible changes, including migration and proliferation of vascular smooth muscle cells and deposition of extracellular matrix. These cellular events in chronic hypoxia correlate with the remodeling events in end-stage PAH. However, because the histopathology of hypoxic pulmonary hypertension does not recapitulate all aspects of PAH, some mechanistic differences in pathogenesis certainly exist but have not yet been fully identified. These are important considerations, especially in the context of interpreting studies of hypoxic pulmonary hypertension and extrapolating those findings to the pathogenesis of PAH.

PAH is associated with hemoglobinopathies, especially thalessemias [76, 77] and sickle cell anemia [78-80]. Hemolysis accompanying these disorders may lead to destruction of bioactive nitric oxide by free hemoglobin [81] or reactive oxygen species [82, 83]. Furthermore, reactive oxygen species may lead to increased levels of oxyhemoglobin, which further impairs the delivery of nitric oxide to the vessel wall. As a result of the lack of available nitric oxide, an inflammatory and proliferative cascade may ensue with culmination in PAH. Accordingly, decreased nitric oxide bioavailability with development of pulmonary hypertension has been reported after hemolysis in a murine model of sickle cell disease [84]. In vivo correlation to human disease is pending.

Pulmonary arteriopathy complicates autoimmune diseases, especially in the setting of the CREST variant of limited systemic sclerosis and, to a lesser degree, in mixed connective tissue disease, systemic lupus erythematosis, and rheumatoid arthritis [85-88]. The occurrence of PAH in each disease has been associated with Raynaud's phenomenon, suggesting at least some similarities in pathogenesis [89]. The presence of interstitial lung disease and pulmonary fibrosis, seen at varying frequency in these autoimmune syndromes, may represent a common pathogenic factor in the development of PAH. Accordingly, in the setting of pulmonary fibrosis and hypoxia, significant inflammation and deposition of extracellular matrix have been observed, which may increase vasoconstriction, proliferation, and vessel remodeling. Murine models of interstitial lung disease utilizing chemical (bleomycin) exposure may prove important in further elucidating the pathogenic mechanisms [90, 91].

An association between the human immunodeficiency virus (HIV) infection and pulmonary hypertension has been noted in approximately 0.5% of all patients with HIV infection, a rate 6 to 12 times higher than the general population [92, 93]. Notably, HIV does not infect pulmonary arterial endothelium [94]; however, mechanisms of disease have been proposed that directly stem from effects of HIV infection (as reviewed by [95]). These include infection of smooth muscle cells with subsequent dysregulation of proliferation, imbalance of vascular mitogens in response to systemic HIV infection, and endothelial injury precipitated by HIV-infected T cells [96]. Recently, the direct actions of HIV-encoded proteins have been implicated in PAH development. The HIV gp120 protein may induce pulmonary endothelial dysfunction and apoptosis [97]. In a macaque model of simian immunodeficiency virus infection, a pathogenic interaction of the viral Nef protein with the pulmonary vessel wall has been reported, leading to pulmonary arteriopathy [98]. Cell culture studies have also demonstrated a role for the HIV Tat protein in repression of BMPR-II transcription, potentially provoking a proliferative response in the vessel wall [99]. Finally, it has been proposed that human herpes virus 8 (HHV-8), the causative agent of Kaposi's sarcoma and an opportunistic pathogen highly associated with HIV infection, may play a role in PAH development with progression to plexiform lesions. Although it was initially reported that HHV-8 infection is associated with idiopathic PAH [100], that link has not been consistently validated after study of additional populations [101-106]. Nonetheless, PAH in the setting of HIV infection likely results from multifactorial effects, and the underlying pathogenesis may involve both direct results of viral infection and indirect consequences of associated pathogens.

Increased flow through the pulmonary circulation has long been associated with development of PAH. Certain types of congenital heart disease with functional systemic-to-pulmonary shunts, such as unrestricted ventricular septal defects (VSD) and large patent ductus arteriosus (PDA), invariably lead to pulmonary vascular remodeling and the clinical syndrome of PAH during childhood (Eisenmenger's syndrome). Presence of atrial septal defects (ASD) with systemic-to pulmonary shunts may also lead to PAH over time [107]. Yet, in contrast to the cases of unrestricted VSD and PDA, only 10-20% of all persons with atrial septal defects progress to PAH [108]. This observation may reflect differences in the response of the pulmonary vasculature to pressure overload (as seen in shunts with VSD and PDA) as compared to volume overload (as seen in shunts with ASD); however, the underlying mechanisms are unclear. Furthermore, a widely held hypothesis suggests that patients with ASD may harbor a specific, unidentified genetic predisposition to the development of PAH that may work in concert with or independently of the increased volume load to the pulmonary circulation. This putative predisposition does not appear to rely upon BMPR2 function, as BMPR2 mutations do not specifically cluster with the presence of ASD [109]. At the molecular level, the physiologic flow patterns of laminar shear stress, turbulent flow, and cyclic strain are all recognized by endothelial cells, leading to transduction of intracellular signals and modulation of a wide variety of phenotypic changes (as reviewed in [110]). Significant prior work has focused mainly on the endothelium of the peripheral vasculature, suggesting that laminar flow induces a vasoprotective, quiescent vascular state while turbulent flow leads to a pro-inflammatory and thrombogenic state. It is unclear if these flow-dependent phenotypes are recapitulated in the pulmonary vasculature. In part, this stems from the difficulty of directly studying the in vivo flow patterns at the anatomic level of the pulmonary arteriole. Recently, ex vivo modeling of pulsatile flow with high levels of shear stress and chronic VEGF inhibition has demonstrated apoptosis of pulmonary artery endothelial cells followed by outgrowth and selection for proliferating apoptosis-resistant cells [111]. Therefore, chronically elevated flow may allow for selection of cells with dysregulated endothelial cell growth and resulting clonal or polyclonal expansion to plexiform lesions. A number of animal models have been developed to study “flow-dependent” PAH, notably involving surgical creation of a shunt in rodents and larger mammalian species (as reviewed in [112]). In addition, attempts at computer modeling have shown promise [113, 114]. These may play important roles in the future for better delineating the molecular effects of increased flow in the pulmonary vasculature.

PAH has been reported in patients suffering from other clinical syndromes such as chronic myelodysplastic syndromes with thrombocytosis [115, 116] and idiopathic thrombocythemia [117, 118]; as well as in persons exposed to stimulants of the central nervous system such as methamphetamines and cocaine [119-121]. Dysregulation of serotoninergic signaling may contribute but does not explain these associations entirely. PAH can also develop in patients suffering from portal hypertension [122-124], and in neonates with failure of the normal fetal-to-neonatal circulatory transition (persistent pulmonary hypertension of the newborn). Finally, idiopathic PAH demonstrates a gender predilection, with a high predominance of affected females [125]. It remains unknown if these associated states alone act as primary triggers of PAH or if confounding factors otherwise predominate pathogenesis.

Vascular Effectors

Downstream of the genetic and acquired triggers of PAH, the histopathologic processes that predominate later stages of disease include vasoconstriction, cellular proliferation, and thrombosis. These processes are influenced by a complex and dysregulated balance of vascular effectors controlling vasodilatation and vasoconstriction, growth suppressors and growth factors, and pro- versus anti-thrombotic mediators. Most of these effectors have been described in previous comprehensive reviews and will be described here specifically in regard to their known roles in the pathogenesis of PAH (Table 1).

Table 1. Dysregulation of Vascular Effectors in Pulmonary Arterial Hypertension.

An imbalance of mediators of pulmonary vascular response has been observed in PAH. The predominant pathogenic consequences of these imbalances are tabulated; these influence vasoconstriction, cellular proliferation, and thrombosis. Less significant effects are denoted by (N/A); unclear effects are indicated by (?).

Vascular Effector Change in
Activity in PAH
Effect on
Vasoconstriction
Effect on Cell
Proliferation
Effect on
Thrombosis
Serotonin Increased Increased Increased Increased
Nitric Oxide Decreased
(Increased in
plexiform
lesions)
Increased (?) Increased
in plexiform
lesions
Increased
Thromboxane A2 Increased Increased Increased Increased
Prostacyclin Decreased Increased Increased Increased
Endothelin-1 Increased Increased Increased (N/A)
Vasoactive Intestinal Peptide (VIP) Decreased Increased Increased Increased
Vascular Endothelial Growth Factor (VEGF) Increased (N/A) Increased (N/A)
Rho-Kinase Increased Increased Increased Increased
Kv Channel Decreased Increased Increased (N/A)
Angiopoietin-1 Increased (N/A) Increased (N/A)
Caveolae Decreased (?) (?) (?)
5-lipoxygenase (5-LO) Increased Increased Increased Increased
Vascular Elastase Increased Increased Increased Increased

Gaseous vasoactive molecules regulate pulmonary vascular homeostasis, and alterations in their endogenous production have been linked to the progression of PAH. Nitric oxide (NO) is the best described of these factors. It is a potent pulmonary arterial vasodilator as well as a direct inhibitor of platelet activation and vascular smooth muscle cell proliferation. The synthesis of NO is mediated by a family of NO synthase enzymes (NOS). The endothelial isoform (eNOS) is regulated by a multitude of vasoactive factors and physiologic stimuli including hypoxia, inflammation, and oxidative stress. Reduced levels of eNOS have been demonstrated in the pulmonary vasculature of patients with PAH, suggesting a mechanism of dysregulated vasoconstriction [126, 127]. Correspondingly, a murine model that genetically lacks eNOS is more susceptible to developing pulmonary hypertension in response to other endogenous stimuli, as compared with the wildtype control [128, 129]. Furthermore, the impact of NO has been reflected in its therapeutic role in PAH, as inhaled NO (as reviewed in [130]) and the NO-dependent phosphodiesterase type-5 inhibitor sildenafil [131] (and as reviewed in [132]). Nonetheless, the exact mechanisms of eNOS and NO dysregulation in PAH have not been fully elucidated. First, increased levels of eNOS have been described specifically in plexiform lesions in idiopathic PAH [133]. Increased NO levels may provoke endothelial cell proliferation in these lesions; however, this link has not been verified. Second, dysregulation of NO depends not only upon NOS activity but also upon still incompletely characterized processes of NO transport in blood [134]. Third, NO levels are intricately associated with oxidative stress, another likely but incompletely described regulator of pulmonary hypertension [135]. Finally, specific polymorphisms of NOS have been associated with pulmonary hypertension [136, 137]; however, mechanistic details are lacking. Again, further characterization of the dysregulated action of NO and potential interactions with other pathogenic factors may depend upon the development of a genetically tractable animal model of severe PAH.

Similar to NO, carbon monoxide (CO) and hydrogen sulfide (H2S) are endogenously produced gaseous vasodilators, deficiencies of which may promote development of PAH. Mice that are null in heme oxygenase-1 (HO-1), a primary enzyme that produces CO in the pulmonary vasculature, exhibit less tolerance for hypoxia with resulting right ventricular dysfunction [138]. In contrast, overexpression of HO-1 in the lung prevents development of pulmonary hypertension in murine models of chronic hypoxia [139] and in rat models of monocrotaline-induced PAH [140]. H2S also functions as a vasodilator and inhibitor of vessel wall proliferation, which can protect against the development of PAH in rat models [141, 142]. However, studies elucidating the precise regulation of CO and H2S production during disease progression are currently lacking.

The arachidonic acid metabolites prostacyclin and thromboxane A2 also play crucial roles in vasoconstriction, thrombosis, and, to a certain degree, vessel wall proliferation. Prostacyclin (prostaglandin I2) activates cyclic adenosine monophosphate (cAMP)-dependent pathways and serves as a vasodilator, an antiproliferative agent for vascular smooth muscle, and an inhibitor of platelet activation and aggregation. In contrast, thromboxane A2 increases vasoconstriction and activates platelets [143]. Protein levels of prostacyclin synthase are decreased in small and medium-sized pulmonary arteries in patients with PAH, particularly with the idiopathic form [144]. Biochemical analysis of urine in patients with PAH has shown decreased levels of a breakdown product of prostacyclin (6-keto-prostacyclin F2alpha), accompanied by increased levels of a metabolite of thromboxane A2 (thromboxane B2) [145]. Therefore, it appears that production of these effectors is coordinately regulated with the imbalance toward thromboxane A2 favored in the development of PAH. The mode of this regulation remains to be characterized. Nonetheless, recognition of this imbalance has led to the success of prostacyclin therapy and improvement of hemodynamics, clinical status, and survival in patients with severe PAH (as reviewed in [146]).

Endothelin-1 (ET-1) is expressed by pulmonary endothelial cells and has been identified as a significant vascular mediator in PAH. It acts as both a potent pulmonary arterial vasoconstrictor and mitogen of pulmonary smooth muscle cells [147, 148]. The vasoconstrictor response relies upon binding to the endothelin receptor A (ETA receptor) on vascular smooth muscle cells. This leads to an increase in intracellular calcium, along with activation of protein kinase C, mitogen-activated protein kinase, and the early growth response genes c-fos and c-jun [5]. The mitogenic action of ET-1 on pulmonary vascular smooth muscle cells can occur through either the ETA receptor and/or the ETB receptor subtype, depending on the anatomic location of cells. The ETA receptor predominantly mediates mitogenesis in the main pulmonary artery, whereas mitogenesis in resistance arteries relies upon contributions from both subtypes. The resulting vasoconstriction, mitogenesis, and vascular remodeling are thought to lead to significant hemodynamic changes in the pulmonary vasculature and to PAH. Plasma levels of endothelin-1 are increased in animal and human subjects suffering from PAH due to a variety of etiologies [149, 150]. Again, improvement in hemodynamics, clinical status, and survival of PAH patients treated with chronic ET receptor antagonists highlights the significance of these effects (as reviewed in [151]). However, the complete upstream pathogenic mechanisms that result in elevated levels of endothelin-1 and dysregulated signaling have not been fully clarified.

Downregulation of vasoactive intestinal peptide (VIP) may also play a pathogenic role. VIP is a pulmonary vasodilator, an inhibitor of proliferation of vascular smooth muscle cells, and an inhibitor of platelet aggregation [152, 153]. Decreased concentrations of VIP have been reported in serum and lung tissue of patients with PAH [154]. VIP-null mice suffer from moderate pulmonary hypertension [155]. Furthermore, both pulmonary arterial pressure and pulmonary vascular resistance decrease after treatment with VIP [156-158]. Improvement of hemodynamics and clinical course has also been observed with inhaled VIP in a small number of PAH patients [154]. Key questions regarding the mode(s) of regulation of VIP expression and its putative causative role in PAH remain unanswered.

Vascular endothelial growth factor (VEGF) is a well-studied endothelial cell mitogen and angiogenic factor. In the pulmonary circulation, it binds endothelial cells via tyrosine kinase receptors (two subtypes: VEGFR-1/KDR and VEGFR-2/Flt). Production of VEGF and its receptors is upregulated in human pulmonary tissue in both acute and chronic hypoxia [159]. Increased VEGF expression has also been observed in PAH, accompanied by elevated levels of VEGFR-1 in the affected pulmonary endothelium and specifically elevated levels of VEGFR-2 in plexiform lesions [6, 160]; however, other signaling molecules that are typically involved in the VEGF angiogenic signaling cascade appear to decrease: phosphoinositide-3-kinase, Akt, and Src. As a result, a dysregulated response to VEGF has been proposed to critically influence endothelial cell survival, proliferation, and apoptosis. Correspondingly, mice with homozygous deletions in the B isoform of VEGF (VEGF-B) demonstrate less vascular remodeling compared to wildtype controls when exposed to hypoxia [161], indicating that VEGF-B can exacerbate vascular proliferative changes. In rat models, a combination of chronic blockade of VEGFR-2 and hypoxia leads to pulmonary endothelial cell apoptosis and to the outgrowth of selected apoptotic-resistant, proliferating endothelial cells with severe PAH [162]. Apoptosis followed by selection of apoptosis-resistant cells may be a crucial event in PAH, triggered by VEGFR-2 blockade. Correlation of these findings to in vivo human disease is pending. In aggregate, however, these observations may offer a partial mechanistic explanation of disordered angiogenic response, clonal expansion of endothelial cells, and resulting plexiform lesions in PAH.

The above vascular effectors all appear to be active in the progression of different forms of PAH. Interestingly, it has been recently reported that peroxisome proliferator-activated receptor gamma (PPARγ) can ameliorate PAH in insulin-resistant, apolipoprotein E-deficient mice [163]. This suggests not only a link between insulin resistance/obesity and PAH but also highlights a novel protective role for PPARγ in pulmonary vasculopathy. Other potential contributing factors to PAH progression include phosphodiesterase I [164], survivin [165], the calcium binding protein S100A4/Mts1[166, 167], the transient receptor potential channels [168], and adrenomedullin [169-171]. These may represent important but yet incompletely described pathogenic contributors. Furthermore, other vascular growth factors such platelet derived growth factor, basic fibroblast growth factor, insulin-like growth factor-1, and epidermal growth factor all may play downstream roles in later stages of PAH. Yet, while our understanding of the specific action of each individual effector has improved, the in vivo vascular responses to their coordinate action remain unclear. In fact, none of these factors has yet been definitively linked to the root pathogenesis of disease. Insight into this topic is offered by the fact that the above effectors are likely subject to upstream, over-arching regulatory pathways that affect the action of multiple vasoactive molecules [172]. Characterization of these regulatory mechanisms may eventually allow for identification of primary molecular triggers of disease and offer novel therapeutic targets for drug development. Some examples of potential overarching and overlapping regulatory pathways may include those that function through rho-kinase, voltage activated potassium channels, angiopoietin-1, caveolae, 5-lipoxygenase (5-LO), and vascular elastase.

Multiple cell types in the vascular wall rely upon the rho-kinase signaling pathway for homeostatic function and response to injury (as reviewed in [173]). These cell types include endothelial and vascular smooth muscle cells, inflammatory cells, and fibroblasts. Rho is a guanosine triphosphate (GTP) binding protein that activates its downstream target, rho-kinase, in response to activation of a variety of G-protein coupled receptors. When activated, rho-kinase inhibits myosin phosphatase and conversely upregulates the ERM family of kinases. In vitro activation of these signaling cascades results in modulation of multiple cellular processes, including enhanced vasoconstriction, proliferation, impaired endothelial response to vasodilators, chronic pulmonary remodeling, and upregulation of vasoactive cytokines via the NF-κB transcription pathway. Rho-kinase activity has also been linked specifically to a number of known effectors of PAH, including endothelin-1 [174, 175], serotonin [176, 177], and eNOS [178], among others. Recently, elevated rho-kinase activity has been demonstrated in animal models of PAH [179, 180]. Furthermore, intravenous fasudil, a selective rho-kinase inhibitor, has induced pulmonary vasodilatation and regression of PAH in various animal models [179-186] as well as in patients with severe PAH who were otherwise refractory to conventional therapies [187, 188]. Interestingly, in a mouse model of chronic hypoxia, pulmonary hypertension improved after therapy with simvastatin, a 3-hydroxy-3-methylglutaryl CoA reductase inhibitor known also to inhibit rho-kinase activity [189, 190]. Taken together, these data suggest that rho-kinase may control a master molecular “switch” in the pulmonary artery, initiating an activated state in disease from a quiescent state in health. As a result, rho-kinase represents an attractive and novel “upstream” therapeutic target for treatment of PAH.

Modulation of voltage-gated potassium channels (Kv) may also represent an overarching pathogenic mechanism of PAH. Kv channels are inhibited in the smooth muscle cells of resistance vessels in the pulmonary arterial tree in response to hypoxia, and they regulate hypoxic pulmonary vasoconstriction (as reviewed in [191]). Subsequent depolarization leads to the opening of voltage-gated calcium channels, an increase in intracellular calcium, and the initiation of a number of intracellular signaling cascades promoting vasoconstriction and proliferation and inhibiting apoptosis. Expression array analysis has demonstrated a depletion of Kv1.5 channels in pulmonary tissue derived from PAH patients [192]. It is currently unknown if these Kv channel abnormalities are congenital or acquired; however, a number of polymorphisms in the Kv1.5 channel gene (KCNA5) have been described, which may suggest a genetic predisposition to channel depletion [193]. Appetite suppressants, such as dexfenfluramine and aminorex that are risk factors for development of PAH, can also directly inhibit Kv1.5 and Kv2.1 [194]. Inhibition of Kv currents in pulmonary smooth muscle cells may be regulated by serotonin [195], thromboxane A2 [196], and, perhaps, nitric oxide [197]. Furthermore, BMP signaling can regulate Kv receptor expression [198, 199]. Taken together, the Kv pathway may represent a common point of regulation in pathogenesis. Accordingly, augmentation of Kv activation would be predicted to induce vasodilatation and, perhaps, allow for regression of vessel remodeling. In vivo gene transfer of Kv channels in chronically hypoxic rats has led to improvement of pulmonary hypertension and suggests its therapeutic potential [200].

Angiopoietin-1 is an angiogenic factor that has been linked to the regulation of multiple vascular effectors of PAH (as reviewed in [201]). Angiopoietin-1 is produced by smooth muscle cells and pericytes during vascular development; it binds the Tie2 receptor expressed on endothelium, which then activates smooth muscle proliferation and migration. After development, pulmonary expression is dramatically reduced; however, most non-familial forms of PAH are characterized by up-regulation of Tie2, and these levels correlate with severity of histologic disease [45]. The level of angiopoietin-1 itself may also be upregulated in some cases [202, 203]. In this context, angiopoietin-1 can stimulate pulmonary arterial endothelial cells to secrete mitogenic factors such as serotonin [204] and endothelin-1 [203]. As discussed previously, angiopoietin-1 also represses endothelial expression of BMPR-IA, a receptor that dimerizes with BMPR-II [45]. It is conceivable that angiopoietin-1 may serve as a crucial link between the serontoninergic and BMP pathways with resulting smooth muscle cell hyperplasia [205]. In correlation, rodents expressing transgenic angiopoietin-1 in pulmonary tissue develop pulmonary vascular remodeling with diffuse smooth muscle cell hyperplasia in small pulmonary vessels [72]. Furthermore, gene transfer of a Tie2 receptor antagonist ameliorates monocrotaline-induced PAH in rats [204]. Paradoxically, angiopoietin-1 has also demonstrated a protective role in some forms of PAH, based on separate rodent studies [206]. As a result, while angiopoietin-1 likely influences the function of multiple vascular mediators, it is unclear if it primarily acts as a direct pathogenic factor or as a homeostatic response mediator in the human manifestation of PAH.

The actions of caveolae and its main coat protein caveolin-1 (CAV-1) may also represent a possible upstream regulatory pathway of the development of PAH. Caveolae are flask-shaped invaginations found on the surface of the plasma membrane in a variety of vascular cell types, including pulmonary endothelium, vascular smooth muscle cells, and fibroblasts (as reviewed in [207]). Caveolae are potential regulators of signaling function that spatially organize and concentrate signaling molecules. CAV-1 is depleted in plexiform lesions and muscularized pulmonary arterioles from patients with PAH [208]. Furthermore, CAV-1-null mice develop a dilated cardiomyopathy and pulmonary hypertension [209] with impaired NO and calcium signaling [210]. A number of possible pathogenic mechanisms can be envisioned. Both eNOS [211] and endothelin-1 [212] are targeted to caveolae; disruption of these or other trafficking pathways could lead to increased inflammation and proliferation in the vessel wall [213]. BMPR-II may traffic through cholesterol-rich membrane rafts and caveolae, suggesting a possible regulatory role of caveolae for the BMP pathway and potential PAH development [214]. Serotonin signaling may depend upon caveolae to downregulate Kv channels [195]. Moreover, CAV-1 itself can function as a tumor suppressor gene, which, if depleted in the vasculature, may directly lead to increased proliferation [215]. Definitive proof, however, of the specific pathogenic actions of caveolae and CAV-1 in PAH is lacking.

A severe inflammatory state predominates end-stage PAH and may play a significant role in pathogenesis. A number of soluble chemoattractants and pro-inflammatory cytokines from the pulmonary artery are upregulated in human and animal models of severe PAH. These include interleukin-1β [216], transforming growth factor- β1 [217], bradykinin [218], monocyte chemotactic protein-1 [219], fractalkine [220], RANTES [221], and leukotrienes [222], among others. 5-lipoxygenase (5-LO) regulates the synthesis of leukotrienes, which in turn can promote cytokine release. 5-LO may represent a possible upstream factor involved in inciting this pro-inflammatory state [223]. 5-LO promotes endothelial cell proliferation in cell culture, and elevated levels of 5-LO have been detected in macrophages and pulmonary endothelium derived from patients suffering from idiopathic PAH [224]. In both a monocrotaline-treated rat model [225] and a genetically susceptible BMPR2 +/− heterozygote mouse model [52] of PAH, overexpression of 5-LO via adenoviral delivery has worsened pulmonary hypertension and vascular remodeling. Furthermore, 5-LO inhibitors have attenuated pulmonary hypertension in both of these models. It is tempting to speculate that 5-LO may possess an upstream regulatory role in PAH progression. Yet, it is also possible that 5-LO and its enzymatic products and derivatives may represent only the severity of the end-stage inflammatory state.

Finally, vascular-specific serine elastase activity has been implicated in the pathogenesis of PAH via regulation of the remodeling response in the extracellular matrix (as reviewed in [226]). In pulmonary arterioles, serine elastases are secreted into the extracellular space to activate matrix metalloproteinases (MMP) and to inhibit tissue inhibitors of MMP (TIMP). Both MMP and elastases degrade most components of the extracellular matrix leading to an upregulation of fibronectin and subsequent enhancement of cellular migration. Matrix degradation also leads to increased integrin signaling with resulting expression of the glycoprotein tenascin C. Tenascin C acts cooperatively with other growth factors (i.e., epidermal growth factor) to enhance smooth muscle proliferation. Increased degradation of elastin [227] has been observed in pulmonary arteries from patients suffering from congenital heart disease and resultant pulmonary hypertension. In addition, rat models of pulmonary hypertension have demonstrated increased production and activity of vascular elastases [228] and tenascin C [229]. Tenascin C also is induced in pulmonary tissue of patients harboring BMPR2 mutations and suffering from familial PAH [230]. This upregulation of elastase function may be induced by a number of vascular effectors implicated in PAH, including NO [231], serotonin [58], and theoretically, the BMP pathway [4]. Elastase inhibitors can induce apoptosis of smooth muscle cells in cell culture and can improve PAH in animal models [232-234]. While elastase function may not represent a primary trigger of PAH, its function in the extracellular matrix likely coordinates multiple downstream signaling pathways involved in disease propagation and, for this reason, may also represent a novel therapeutic target.

Conclusions and Future Directions

Pulmonary arterial hypertension refers to a clinical syndrome of vascular disease with a stereotyped pattern of histopathology and is related to a variety of secondary disease states. It has become increasingly clear that development of PAH entails a complex, multifactorial pathophysiology. Although genetic mutations, exogenous exposures, and acquired disease states can predispose to PAH, no one factor identified thus far is sufficient alone to drive fully the pathogenic process. Similar to carcinogenesis in which a susceptible person with a specific genetic mutation requires additional injuries to manifest disease, a “multiple-hit” hypothesis has emerged to explain the progression to clinical PAH (Figure 4).

Figure 4. Paradigm for the “Multiple-Hit” Hypothesis Promoting Pulmonary Arterial Hypertension.

Figure 4

Susceptible persons with genetic or acquired traits do not progress to PAH without suffering from additional insults that are synergistic in the pathogenesis of disease.

More delineated mechanisms of disease have focused on the end-stage condition and the effects of the imbalance of multiple vascular effectors. Some unifying mechanism(s) of disease have become more apparent, linking the regulation of these effectors into a more cohesive model. However, a complete understanding of these complex cellular processes at the molecular level is incomplete. In part, this fact stems from the lack of a small animal model that can be followed and genetically manipulated from initiation through progression to severe disease. There has been some success in physiologic study of PAH in larger mammals; however, identification of contributors in the molecular process of pulmonary remodeling has been difficult, owing to incomplete genetic data and tools specific for these models [112, 235]. Conversely, while genetic and molecular studies in the mouse are more tractable, induction of significant PAH in murine models has been challenging. Nonetheless, recent advances have hinted at the possibility of more tractable animal models of PAH, which would be useful for combining genetic study and pathophysiologic exposures.

Our continued lack of understanding of PAH may also stem from the limitations of utilizing a reductionist approach to define a complex clinical syndrome based on end-stage pathologic observations. In the post-genomic era, it is conceivable that complex clinical syndromes such as PAH will become redefined, based on growing genomic, transcriptional, and proteomic data. A “systems-based” network analysis can be envisioned where specific genetic determinants of disease (such as BMPR2 mutations) may interact with environmental or acquired perturbations (such as hypoxia or anorexigen use), “disease” modifying genes (such as 5-HTT), and “stress response” modifying genes (such as those that modulate angiogenesis or inflammation) to lead to one of several pathophenotypes (such as PAH, right ventricular failure, or thromboembolic disease) [172]. With the aid of the genomic database, this resulting human disease network would offer several unique opportunities: to provide a mechanistic rather than an observational description to pulmonary vascular remodeling, to predict risk factors for PAH, and to identify and quantitate regulatory determinants that otherwise would be ignored. By combining a networks-based approach with more tractable animal models of PAH, the results would certainly improve our ability to identify common, overarching pathways of pathogenesis and aid in our understanding of the mechanistic links among primary disease triggers and end-stage disease. Such information would be valuable not only for prevention of disease for at-risk individuals but also, perhaps, for identification of therapeutic targets useful for regression of the pathogenic process itself.

Acknowledgments

This work was supported in part by NIH grants HL61795, HV28718, and HL81587. We thank Tim Lis for composition of graphic illustrations. We thank Stephanie Tribuna and Susan Vignolo-Collazzo for expert secretarial assistance.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Abbreviations

Atrial septal defect (ASD); B isoform of vascular endothelial growth factor (VEGF-B); bone morphogenetic protein receptor type 2 (BMPR2 gene which encodes for BMPR-II protein); carbon monoxide (CO); caveolin-1 (CAV-1); cyclic adenosine monophosphate (cAMP); endothelin-1 (ET-1); endothelial isoform of nitric oxide synthase (eNOS); 5-lipoxygenase (5-LO); guanosine triphosphate (GTP); heme oxygenase-1 (HO-1); human herpes virus 8 (HHV-8); human immunodeficiency virus (HIV); hydrogen sulfide (H2S); Kv1.5 channel gene (KCNA5 gene); matrix metalloproteinase (MMP); nitric oxide (NO); nitric oxide synthase enzymes (NOS); patent ductus arteriosus (PDA); peroxisome proliferator-activated receptor gamma (PPARγ); pulmonary arterial hypertension (PAH); serotonin (5-HT); selective serotonin re-uptake inhibitor (SSRI); serotonin transporter (5-HTT); tissue inhibitor of MMP (TIMP); transforming growth factor-β (TGF-β); vascular endothelial growth factor (VEGF); vascular endothelial growth factor receptor-1 (VEGFR-1/KDR); vascular endothelial growth factor receptor-2 (VEGFR-2/Flt); vasoactive intestinal peptide (VIP); ventricular septal defect (VSD); voltage-gated potassium channel (Kv).

References

  • 1.Farber H, Loscalzo J. Pulmonary arterial hypertension. N Eng J Med. 2004;351:1655–1665. doi: 10.1056/NEJMra035488. PMID: 15483284. [DOI] [PubMed] [Google Scholar]
  • 2.McLaughlin V, McGoon M. Pulmonary arterial hypertension. Circulation. 2006;114:1417–1431. doi: 10.1161/CIRCULATIONAHA.104.503540. doi:10.1161/CIRCULATIONAHA.104.503540. [DOI] [PubMed] [Google Scholar]
  • 3.Simonneau G, Galie N, Rubin L, Langleben D, Seeger W, Domenighetti G, Gibbs S, Lebrec D, Speich R, Beghetti M, Rich S, Fishman A. Clinical classification of pulmonary hypertension. J Am Coll Cardiol. 2004;43(12 Suppl S):5S–12S. doi: 10.1016/j.jacc.2004.02.037. doi:10.1016/j.jacc.2004.02.037. [DOI] [PubMed] [Google Scholar]
  • 4.Humbert M, Morrell N, Archer S, Stenmark K, MacLean M, Lang I, Christman B, Weir E, Eickelberg O, Voelkel N, Rabinovitch M. Cellular and molecular pathobiology of pulmonary arterial hypertension. J Am Coll Cardiol. 2004;43(12 Suppl S):13S–24S. doi: 10.1016/j.jacc.2004.02.029. doi:10.1016/j.jacc.2004.02.029. [DOI] [PubMed] [Google Scholar]
  • 5.Jeffery T, Morrell N. Molecular and cellular basis of pulmonary vascular remodeling in pulmonary hypertension. Prog Cardiovasc Dis. 2002;45:173–202. doi: 10.1053/pcad.2002.130041. doi:10.1053/pcad.2002.130041. [DOI] [PubMed] [Google Scholar]
  • 6.Cool C, Stewart J, Werahera P, Miller G, Williams R, Voelkel N, Tuder R. Three-dimensional reconstruction of pulmonary arteries in plexiform pulmonary hypertension using cell-specific markers. Evidence for a dynamic and heterogeneous process of pulmonary endothelial cell growth. Am J Pathol. 1999;155:411–419. doi: 10.1016/S0002-9440(10)65137-1. PMID: 10433934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Stenmark K, Gerasimovskaya E, Nemenoff R, Das M. Hypoxic activation of adventitial fibroblasts: role in vascular remodeling. Chest. 2002;122(6 Suppl):326S–334S. doi: 10.1378/chest.122.6_suppl.326s. doi:10.1378/chest.122.6_suppl.326S. [DOI] [PubMed] [Google Scholar]
  • 8.Zhu P, Huang L, Ge X, Yan F, Wu R, Ao Q. Transdifferentiation of pulmonary arteriolar endothelial cells into smooth muscle-like cells regulated by myocardin involved in hypoxia-induced pulmonary vascular remodelling. Int J Exp Pathol. 2006;87:463–474. doi: 10.1111/j.1365-2613.2006.00503.x. doi:10.1111/j.1365-2613.2006.00503.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sakao S, Taraseviciene-Stewart L, Cool C, Tada Y, Kasahara Y, Kurosu K, Tanabe N, Takiguchi Y, Tatsumi K, Kuriyama T, Voelkel N. VEGF-R blockade causes endothelial cell apoptosis, expansion of surviving CD34+ precursor cells and transdifferentiation to smooth muscle-like and neuronal-like cells. FASEB J. 2007 doi: 10.1096/fj.07-8432com. Epub ahead of print. doi: fj.07-8432com. [DOI] [PubMed] [Google Scholar]
  • 10.Lee S, Shroyer K, Markham N, Cool C, Voelkel N, Tuder R. Monoclonal endothelial cell proliferation is present in primary but not secondary pulmonary hypertension. J Clin Invest. 1998;101:927–934. doi: 10.1172/JCI1910. PMID: 9486960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yeager M, Halley G, Golpon H, Voelkel N, Tuder R. Microsatellite instability of endothelial cell growth and apoptosis genes within plexiform lesions in primary pulmonary hypertension. Circ Res. 2001;88:E2–E11. doi: 10.1161/01.res.88.1.e2. PMID: 11139485. [DOI] [PubMed] [Google Scholar]
  • 12.Wagenvoort C. Lung biopsy specimens in the evaluation of pulmonary vascular disease. Chest. 1980;77:614–625. doi: 10.1378/chest.77.5.614. PMID: 7363679. [DOI] [PubMed] [Google Scholar]
  • 13.Taraseviciene-Stewart L, Nicolls M, Kraskauskas D, Scerbavicius R, Burns N, Cool C, Wood K, Parr J, Boackle S, Voelkel N. Absence of T cells confers increased pulmonary arterial hypertension and vascular remodeling. Am J Respir Crit Care Med. 2007;175:1280–1289. doi: 10.1164/rccm.200608-1189OC. doi:10.1164/rccm.200608-1189OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sata M. Role of circulating vascular progenitors in angiogenesis, vascular healing, and pulmonary hypertension: lessons from animal models. Arterio Thromb Vasc Biol. 2006;26:1008–1014. doi: 10.1161/01.ATV.0000206123.94140.f3. doi:10.1161/01.ATV.0000206123.94140.f3. [DOI] [PubMed] [Google Scholar]
  • 15.Loscalzo J. Genetic clues to the cause of primary pulmonary hypertension. N Engl J Med. 2001;345:367–371. doi: 10.1056/NEJM200108023450511. PMID: 11484696. [DOI] [PubMed] [Google Scholar]
  • 16.Deng Z, Morse J, Slager SC,N, Moore K, Venetos G, Kalachikov S, Cayanis E, Fischer S, Barst R, Hodge S, Knowles J. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Gen. 2000;67:737–744. doi: 10.1086/303059. PMID: 10903931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Newman J, Trembath R, Morse J, Grunig E, Loyd J, Adnot S, Coccolo F, Ventura C, Phillips J, Knowles J, Janssen B, Eickelberg O, Eddahibi S, Herve P, Nichols W, Elliott G. Genetic basis of pulmonary arterial hypertension: current understanding and future directions. J Am Coll Cardiol. 2004;43(12 Suppl S):33S–39S. doi: 10.1016/j.jacc.2004.02.028. doi:10.1016/j.jacc.2004.02.028. [DOI] [PubMed] [Google Scholar]
  • 18.Trembath R, Thomson J, Machado R, Morgan N, Atkinson C, Winship I, Simonneau G, Galie N, Loyd J, Humbert M, Nichols W, Morrell N, Berg J, Manes A, McGaughran J, Pauciulo M, Wheeler L. Clinical and molecular genetic features of pulmonary hypertension in patients with hereditary hemorrhagic telangiectasia. N Engl J Med. 2001;345:325–334. doi: 10.1056/NEJM200108023450503. PMID: 11484689. [DOI] [PubMed] [Google Scholar]
  • 19.Chaouat A, Coulet F, Favre C, Simonneau G, Weitzenblum E, Soubrier F, Humbert M. Endoglin germline mutation in a patient with hereditary haemorrhagic telangiectasia and dexfenfluramine associated pulmonary arterial hypertension. Thorax. 2004;59:446–448. doi: 10.1136/thx.2003.11890. doi:10.1136/thx.2003.11890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lane K, Machado R, Pauciulo M, Thomson J, Phillips Jr, Loyd J, Nichols W, Trembath R. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. The International PPH Consortium. Nat Gen. 2000;26:81–84. doi: 10.1038/79226. doi:10.1038/79226. [DOI] [PubMed] [Google Scholar]
  • 21.Newman J, Wheeler L, Lane K, Loyd E, Gaddipati R, Phillips Jr, Loyd J. Mutation in the gene for bone morphogenetic protein receptor II as a cause of primary pulmonary hypertension in a large kindred. N Engl J Med. 2001;345:319–324. doi: 10.1056/NEJM200108023450502. [Erratum, N Engl J Med 2001; 2345:1506, 2002; 2346:1258.]. PMID: 11484688. [DOI] [PubMed] [Google Scholar]
  • 22.Aldred M, Machado R, James V, Morrell N, Trembath R. Characterization of the BMPR2 5′-untranslated region and a novel mutation in pulmonary hypertension. Am J Respir Crit Care Med. 2007 doi: 10.1164/rccm.200701-164OC. Epublication. doi:10.1164/rccm.200701-164OC. [DOI] [PubMed] [Google Scholar]
  • 23.Machado R, Aldred M, James V, Harrison R, Patel B, Schwalbe E, Gruenig E, Janssen B, Koehler R, Seeger W, Eickelberg O, Olschewski H, Elliott C, Glissmeyer E, Carlquist J, Kim M, Torbicki A, Fijalkowska A, Szewczyk G, Parma J, Abramowicz M, Galie N, Morisaki H, Kyotani S, Nakanishi N, Morisaki T, Humbert M, Simonneau G, Sitbon O, Soubrier F, Coulet F, Morrell N, Trembath R. Mutations of the TGF-beta type II receptor BMPR2 in pulmonary arterial hypertension. Human Mut. 2006;27:121–132. doi: 10.1002/humu.20285. doi:10.1002/humu.20285. [DOI] [PubMed] [Google Scholar]
  • 24.Thomson J, Machado R, Pauciulo M, Morgan N, Humbert M, Elliott G, Ward K, Yacoub M, Mikhail G, Rogers P, Newman J, Wheeler L, Higenbottam T, Gibbs J, Egan J, Crozier A, Peacock A, Allcock R, Corris P, Loyd J, Trembath R, Nichols W. Sporadic primary pulmonary hypertension is associated with germline mutations of the gene encoding BMPR-II, a receptor member of the TGF-beta family. J Med Gen. 2000;37:741–745. doi: 10.1136/jmg.37.10.741. PMID: 11015450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cogan J, Vnencak-Jones C, Phillips Jr, Lane K, Wheeler L, Robbins I, Garrison G, Hedges L, Loyd J. Gross BMPR2 gene rearrangements constitute a new cause for primary pulmonary hypertension. Gen in Med. 2005;7:169–174. doi: 10.1097/01.gim.0000156525.09595.e9. doi:10.1097/01.GIM.000156525.09595.E9. [DOI] [PubMed] [Google Scholar]
  • 26.Aldred M, Vijayakrishnan J, James V, Soubrier F, Gomez-Sanchez M, Martensson, Galie N, Manes A, Corris P, Simonneau G, Humbert M, Morrell N, Trembath R. BMPR2 gene rearrangements account for a significant proportion of mutations in familial and idiopathic pulmonary arterial hypertension. Hum Mutat. 2006;27:212–213. doi: 10.1002/humu.9398. doi:10.1002/humu.9398. [DOI] [PubMed] [Google Scholar]
  • 27.Beppu H, Kawabata M, Hamamoto T, Chytil A, Minowa O, Noda T, Miyazono K. BMP type II receptor is required for gastrulation and early development of mouse embryos. Develop Biol. 2000;221:249–258. doi: 10.1006/dbio.2000.9670. doi:10.1006/dbio.2000.9670. [DOI] [PubMed] [Google Scholar]
  • 28.Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to nucleus. Cell. 2003;113:685–700. doi: 10.1016/s0092-8674(03)00432-x. doi:10.1016/S0092-8674(03)00432-X. [DOI] [PubMed] [Google Scholar]
  • 29.Wong W, Knowles J, Morse J. Bone morphogenetic protein receptor type II C-terminus interacts with c-Src: implication for a role in pulmonary arterial hypertension. Am J Respir Cell Molec Biol. 2005;33:438–446. doi: 10.1165/rcmb.2005-0103OC. doi: 10.1165/rcmb.2005-0103OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Massagué J, Chen Y. Controlling TGF-beta signaling. Genes Dev. 2000;14:627–644. PMID: 10733523. [PubMed] [Google Scholar]
  • 31.Jeffery T, Upton P, Trembath R, Morrell N. BMP4 inhibits proliferation and promotes myocyte differentiation of lung fibroblasts via Smad1 and JNK pathways. Am J Physiol. Lung Cell Molec Physiol. 2005;288:L370–L378. doi: 10.1152/ajplung.00242.2004. doi:10.1152/ajplung.00242.2004. [DOI] [PubMed] [Google Scholar]
  • 32.Atkinson C, Stewart S, Upton P, Machado R, Thomson J, Trembath R, Morrell N. 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. doi:10.1161/01.CIR.0000012754.72951.3D. [DOI] [PubMed] [Google Scholar]
  • 33.Zhang S, Fantozzi I, Tigno D, Yi E, Platoshyn O, Thistlethwaite P, Kriett J, Yung G, Rubin L, Yuan J. Bone morphogenetic proteins induce apoptosis in human pulmonary vascular smooth muscle cells. Am J Physiol. Lung Cell Molec Physiol. 2003;285:L740–L754. doi: 10.1152/ajplung.00284.2002. doi:10.1152/ajplung.00284.2002. [DOI] [PubMed] [Google Scholar]
  • 34.Zakrzewicz A, Hecker M, Marsh L, Kwapiszewska G, Nejman B, Long L, Seeger W, Schermuly R, Morrell N, Morty R, Eickelberg O. Receptor for activated C-kinase 1, a novel interaction partner of type II bone morphogenetic protein receptor, regulates smooth muscle cell proliferation in pulmonary arterial hypertension. Circulation. 2007;115:2957–2968. doi: 10.1161/CIRCULATIONAHA.106.670026. doi:10.1161/CIRCULATIONAHA.106.670026. [DOI] [PubMed] [Google Scholar]
  • 35.Teichert-Kuliszewska K, Kutryk M, Kuliszewski M, Karoubi G, Courtman D, Zucco L, Granton J, Stewart D. Bone morphogenetic protein receptor-2 signaling promotes pulmonary arterial endothelial cell survival: implications for loss-of-function mutations in the pathogenesis of pulmonary hypertension. Circ Res. 2006;98:209–217. doi: 10.1161/01.RES.0000200180.01710.e6. doi:10.1161/01.RES.0000200180.01710.e6. [DOI] [PubMed] [Google Scholar]
  • 36.Thomas A, Carneal J, Markin C, Lane K, Phillips J, Loyd J, Gaddipati R. Specific bone morphogenic protein receptor II mutations found in primary pulmonary hypertension cause different biochemical phenotypes in vitro. Chest. 2002;121(3 Suppl):83S. PMID: 11893707. [PubMed] [Google Scholar]
  • 37.Machado R, Pauciulo M, Thomson J, Lane K, Morgan N, Wheeler L, Phillips J, Newman J, Williams D, Galiè N, Manes A, McNeil K, Yacoub M, Mikhail G, Rogers P, Corris P, Humbert M, Donnai D, Martensson G, Tranebjaerg L, Loyd J, Trembath R, Nichols W. BMPR2 haploinsufficiency as the inherited molecular mechanism for primary pulmonary hypertension. Am J Hum Gen. 2001;68:92–102. doi: 10.1086/316947. PMID: 11115378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.West J, Fagan K, Steudel W, Fouty B, Lane K, Harral J, Hoedt-Miller M, Tada Y, Ozimek J, Tuder R, Rodman D. Pulmonary hypertension in transgenic mice expressing a dominant-negative BMPRII gene in smooth muscle. Circ Res. 2004;94:1109–1114. doi: 10.1161/01.RES.0000126047.82846.20. doi:10/1161/01.RES.0000126047.82846.20. [DOI] [PubMed] [Google Scholar]
  • 39.Lagna G, Nguyen P, Ni W, Hata A. BMP-dependent activation of caspase-9 and caspase-8 mediates apoptosis in pulmonary artery smooth muscle cells. Am J Physiol. Lung Cell Molec Physiol. 2006;291:L1059–L1067. doi: 10.1152/ajplung.00180.2006. doi:10.1152/ajplung.00180.2006. [DOI] [PubMed] [Google Scholar]
  • 40.Machado R, James V, Southwood M, Harrison R, Atkinson C, Stewart S, Morrell N, Trembath R, Aldred M. Investigation of second genetic hits at the BMPR2 locus as a modulator of disease progression in familial pulmonary arterial hypertension. Circulation. 2005;111:607–613. doi: 10.1161/01.CIR.0000154543.07679.08. doi: 10.1161/01.CIR.0000154543.07679.08. [DOI] [PubMed] [Google Scholar]
  • 41.Rudarakanchana N, Flanagan J, Chen H, Upton P, Machado R, Patel D, Trembath R, Morrell N. Functional analysis of bone morphogenetic protein type II receptor mutations underlying primary pulmonary hypertension. Hum Molec Gen. 2002;11:1517–1525. doi: 10.1093/hmg/11.13.1517. PMID: 12045205. [DOI] [PubMed] [Google Scholar]
  • 42.Fantozzi I, Huang W, Zhang J, Zhang S, Platoshyn O, Remillard C, Thistlethwaite P, Yuan J. Divergent effects of BMP-2 on gene expression in pulmonary artery smooth muscle cells from normal subjects and patients with idiopathic pulmonary arterial hypertension. Exp Lung Res. 2005;31:783–806. doi: 10.1080/01902140500461026. doi: 10.1080/01902140500461026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yu P, Beppu H, Kawai N, Li E, Bloch K. Bone morphogenetic protein (BMP) type II receptor deletion reveals BMP ligand-specific gain of signaling in pulmonary artery smooth muscle cells. J Biol Chem. 2005;280:24443–24450. doi: 10.1074/jbc.M502825200. doi:10.1074/jbc.M502825200. [DOI] [PubMed] [Google Scholar]
  • 44.Yang X, Long L, Southwood M, Rudarakanchana N, Upton P, Jeffery T, Atkinson C, Chen H, Trembath R, Morrell N. Dysfunctional Smad signaling contributes to abnormal smooth muscle cell proliferation in familial pulmonary arterial hypertension. Circ Res. 2005;96:1053–1063. doi: 10.1161/01.RES.0000166926.54293.68. doi:10.1161/01.RES.0000166926.54293.68. [DOI] [PubMed] [Google Scholar]
  • 45.Du L, Sullivan C, Chu D, Cho A, Kido M, Wolf P, Yuan J, Deutsch R, Jamieson S, Thistlethwaite P. Signaling molecules in nonfamilial pulmonary hypertension. N Engl J Med. 2003;348:500–509. doi: 10.1056/NEJMoa021650. PMID: 12571257. [DOI] [PubMed] [Google Scholar]
  • 46.Morty R, Nejman B, Kwapiszewska G, Hecker M, Zakrzewicz A, Kouri F, Peters D, Dumitrascu R, Seeger W, Knaus P, Schermuly R, Eickelberg O. Dysregulated bone morphogenetic protein signaling in monocrotaline-induced pulmonary arterial hypertension. Arterio Thromb Vasc Biol. 2007;27:1072–1078. doi: 10.1161/ATVBAHA.107.141200. doi:10.1161/ATVBAHA.107.141200. [DOI] [PubMed] [Google Scholar]
  • 47.Morrell N, Yang X, Upton P, Jourdan K, Morgan N, Sheares K, Trembath R. Altered growth responses of pulmonary artery smooth muscle cells from patients with primary pulmonary hypertension to transforming growth factor-beta(1) and bone morphogenetic proteins. Circulation. 2001;104:790–795. doi: 10.1161/hc3201.094152. doi:10.1161/hc3201.094152. [DOI] [PubMed] [Google Scholar]
  • 48.Wu X, Chang M, Mitsialis S, Kourembanas S. Hypoxia regulates bone morphogenetic protein signaling through C-terminal-binding protein 1. Circ Res. 2006;99:240–247. doi: 10.1161/01.RES.0000237021.65103.24. doi:10.1161/01.RES.0000237021.65103.24. [DOI] [PubMed] [Google Scholar]
  • 49.Reynolds A, Xia W, Holmes M, Hodge S, Danilov S, Curiel D, Morrell N, Reynolds P. Bone morphogenetic protein type 2 receptor gene therapy attenuates hypoxic pulmonary hypertension. Am J Physiol Lung Cell Molec Physiol. 2007;292:L1182–L1192. doi: 10.1152/ajplung.00020.2006. doi:10.1152/ajplung.00020.2006. [DOI] [PubMed] [Google Scholar]
  • 50.McMurtry M, Moudgil R, Hashimoto K, Bonnet S, Michelakis E, Archer S. Overexpression of human bone morphogenetic protein receptor 2 does not ameliorate monocrotaline pulmonary arterial hypertension. Am J Physiol Lung Cell Molec Physiol. 2007;292:L872–L878. doi: 10.1152/ajplung.00309.2006. doi:10.1152/ajplung.00309.2006. [DOI] [PubMed] [Google Scholar]
  • 51.Beppu H, Ichinose F, Kawai N, Jones R, Yu P, Zapol W, Miyazono K, Li E, Bloch K. BMPR-II heterozygous mice have mild pulmonary hypertension and an impaired pulmonary vascular remodeling response to prolonged hypoxia. Am J Physiol Lung Cell Molec Physiol. 2004;287:L1241–L1247. doi: 10.1152/ajplung.00239.2004. doi:10.1152/ajplung.00239.2004. [DOI] [PubMed] [Google Scholar]
  • 52.Song Y, Jones J, Beppu H, Keaney JJ, Loscalzo J, Zhang Y. Increased susceptibility to pulmonary hypertension in heterozygous BMPR2-mutant mice. Circulation. 2005;112:553–562. doi: 10.1161/CIRCULATIONAHA.104.492488. doi:10.1161/CIRCULATIONAHA.104.492488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.West J, Tada Y, Fagan K, Steudel W, Fouty B, Harral J, Miller M, Ozimek J, Tuder R, Rodman D. Suppression of type II bone morphogenic protein receptor in vascular smooth muscle induces pulmonary arterial hypertension in transgenic mice. Chest. 2005;128(6 Suppl):553S. doi: 10.1378/chest.128.6_suppl.553S. doi:10.1378/chest.128.6_suppl.553S. [DOI] [PubMed] [Google Scholar]
  • 54.Eddahibi S, Adnot S. The serotonin pathway in pulmonary hypertension. Arch Mal Coeur Vaiss. 2006;99:621–625. PMID: 16878724. [PubMed] [Google Scholar]
  • 55.Lee S, Wang W, Lanzillo J, Fanburg B. Serotonin produces both hyperplasia and hypertrophy of bovine pulmonary artery smooth muscle cells in culture. Am J Physiol. 1994;266:L46–52. doi: 10.1152/ajplung.1994.266.1.L46. PMID: 8304469. [DOI] [PubMed] [Google Scholar]
  • 56.Liu Y, Li M, Warburton R, Hill N, Fanburg B. The 5-HT transporter transactivates the PDGF{beta} receptor in pulmonary artery smooth muscle cells. FASEB J. 2007;21:2725–2734. doi: 10.1096/fj.06-8058com. Epub ahead of print. doi: 10.1096/fj.06-8058com. [DOI] [PubMed] [Google Scholar]
  • 57.Morecroft I, Heeley R, Prentice H, Kirk A, MacLean M. 5-hydroxytryptamine receptors mediating contraction in human small muscular pulmonary arteries: importance of the 5-HT1B receptor. Br J Pharmacol. 1999;128:730–734. doi: 10.1038/sj.bjp.0702841. doi:10.1038/sj.bjp.0702841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Launay J, Hervé P, Peoc'h KT,C, Callebert J, Nebigil C, Etienne N, Drouet L, Humbert M, Simonneau G, Maroteaux L. Function of the serotonin 5-hydroxytryptamine 2B receptor in pulmonary hypertension. Nat Med. 2002;8:1129–1135. doi: 10.1038/nm764. doi:10.1038/nm764. [DOI] [PubMed] [Google Scholar]
  • 59.Hervé P, Launay J, Scrobohaci M, Brenot F, Simonneau G, Petitpretz P, Poubeau P, Cerrina J, Duroux P, Drouet L. Increased plasma serotonin in primary pulmonary hypertension. Am J Med. 1995;99:249–254. doi: 10.1016/s0002-9343(99)80156-9. doi:10.1016/S0002-9343(99)80156-9. [DOI] [PubMed] [Google Scholar]
  • 60.Herve P, Drouet L, Dosquet C, Launay J, Rain B, Simonneau G, Caen J, Duroux P. Primary pulmonary hypertension in a patient with a familial platelet storage pool disease: role of serotonin. Am J Med. 1990;89:117–120. doi: 10.1016/0002-9343(90)90111-p. doi:10.1016/0002-9343(90)90111-P. [DOI] [PubMed] [Google Scholar]
  • 61.Abenhaim L, Moride Y, Brenot F, Rich S, Benichou J, Kurz X, Higenbottam T, Oakley C, Wouters E, Aubier M, Simonneau G, Bégaud B. Appetite-suppressant drugs and the risk of primary pulmonary hypertension. International Primary Pulmonary Hypertension Study Group. N Engl J Med. 1996;335:609–616. doi: 10.1056/NEJM199608293350901. PMID: 8692238. [DOI] [PubMed] [Google Scholar]
  • 62.Eddahibi S, Humbert M, Fadel E, Raffestin B, Darmon M, Capron F, Simonneau G, Dartevelle P, Hamon M, Adnot S. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J Clin Invest. 2001;108:1141–1150. doi: 10.1172/JCI12805. doi:10.1172/JCI200112805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Eddahibi S, Chaouat A, Morrell N, Fadel E, Fuhrman C, Bugnet A, Dartevelle P, Housset B, Hamon M, Weitzenblum E, Adnot S. Polymorphism of the serotonin transporter gene and pulmonary hypertension in chronic obstructive pulmonary disease. Circulation. 2003;108:1839–1844. doi: 10.1161/01.CIR.0000091409.53101.E8. doi:10.1161/01.CIR.0000091409.53101.E8. [DOI] [PubMed] [Google Scholar]
  • 64.Machado R, Koehler R, Glissmeyer E, Veal C, Suntharalingam J, Kim M, Carlquist J, Town M, Elliott C, Hoeper M, Fijalkowska A, Kurzyna M, Thomson J, Gibbs S, Wilkins M, Seeger W, Morrell N, Gruenig E, Trembath R, Janssen B. Genetic association of the serotonin transporter in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;173:793–797. doi: 10.1164/rccm.200509-1365OC. doi: 10.1164/rccm.200509-1365OC. [DOI] [PubMed] [Google Scholar]
  • 65.Eddahibi S, Raffestin B, Launay J, Sitbon M, Adnot S. Effect of dexfenfluramine treatment in rats exposed to acute and chronic hypoxia. Am J Respir Crit Care Med. 1998;157:1111–1119. doi: 10.1164/ajrccm.157.4.9704095. PMID: 9563727. [DOI] [PubMed] [Google Scholar]
  • 66.MacLean M, Deuchar G, Hicks M, Morecroft I, Shen S, Sheward J, Colston J, Loughlin L, Nilsen M, Dempsie Y, Harmar A. Overexpression of the 5-hydroxytryptamine transporter gene: effect on pulmonary hemodynamics and hypoxia-induced pulmonary hypertension. Circulation. 2004;109:2150–2155. doi: 10.1161/01.CIR.0000127375.56172.92. doi:10.1161/01.CIR.0000127375.56172.92. [DOI] [PubMed] [Google Scholar]
  • 67.Guignabert C, Izikki M, Tu L, Li Z, Zadigue P, Barlier-Mur A, Hanoun N, Rodman D, Hamon M, Adnot S, Eddahibi S. Transgenic mice overexpressing the 5-hydroxytryptamine transporter gene in smooth muscle develop pulmonary hypertension. Circ Res. 2006;98:1323–1330. doi: 10.1161/01.RES.0000222546.45372.a0. doi:10.1161/01.RES.0000222546.45372.a0. [DOI] [PubMed] [Google Scholar]
  • 68.Keegan A, Morecroft I, Smillie D, Hicks M, MacLean M. Contribution of the 5-HT(1B) receptor to hypoxia-induced pulmonary hypertension: converging evidence using 5-HT(1B)-receptor knockout mice and the 5-HT(1B/1D)-receptor antagonist GR127935. Circ Res. 2001;89:1231–1239. doi: 10.1161/hh2401.100426. doi:10.1161/hh2401.100426. [DOI] [PubMed] [Google Scholar]
  • 69.Marcos E, Adnot S, Pham M, Nosjean A, Raffestin B, Hamon M, Eddahibi S. Serotonin transporter inhibitors protect against hypoxic pulmonary hypertension. Am J Respir Crit Care Med. 2003;168:487–493. doi: 10.1164/rccm.200210-1212OC. doi:10.1164/rccm.200210-12120C. [DOI] [PubMed] [Google Scholar]
  • 70.Chambers C, Hernandez-Diaz S, Van Marter L, Werler M, Louik C, Jones K, Mitchell A. Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med. 2006;354:579–587. doi: 10.1056/NEJMoa052744. PMID: 16467545. [DOI] [PubMed] [Google Scholar]
  • 71.Long L, MacLean M, Jeffery T, Morecroft I, Yang X, Rudarakanchana N, Southwood M, James V, Trembath R, Morrell N. Serotonin increases susceptibility to pulmonary hypertension in BMPR2-deficient mice. Circ Res. 2006;98:818–827. doi: 10.1161/01.RES.0000215809.47923.fd. doi:10.1161/01.RES.0000215809.47923.fd. [DOI] [PubMed] [Google Scholar]
  • 72.Sullivan C, Du L, Chu D, Cho A, Kido M, Wolf P, Jamieson S, Thistlethwaite P. Induction of pulmonary hypertension by an angiopoietin 1/TIE2/serotonin pathway. Proc Natl Acad Sci (USA) 2003;100:12331–12336. doi: 10.1073/pnas.1933740100. doi:10.1073/pnas.1933740100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Dumas J, Bardou M, Goirand F, Dumas M. Hypoxic pulmonary vasoconstriction. Gen Pharmacol. 1999;33:289–297. doi: 10.1016/s0306-3623(99)00026-9. doi:10.1016/S0306-3623(99)00026-9. [DOI] [PubMed] [Google Scholar]
  • 74.Moudgil R, Michelakis E, Archer S. Hypoxic pulmonary vasoconstriction. J Appl Physiol. 2005;98:390–403. doi: 10.1152/japplphysiol.00733.2004. doi:10.1152/japplphysiol.00733.2004. [DOI] [PubMed] [Google Scholar]
  • 75.Sweeney M, Yuan J. Hypoxic pulmonary vasoconstriction: role of voltage-gated potassium channels. Respir Res. 2000;1:40–48. doi: 10.1186/rr11. doi:10.1186/rr11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Grisaru D, Rachmilewitz E, Mosseri M, Gotsman M, Lafair J, Okon E, Goldfarb A, Hasin Y. Cardiopulmonary assessment in beta-thalassemia major. Chest. 1990;98:1138–1142. doi: 10.1378/chest.98.5.1138. PMID: 2225958. [DOI] [PubMed] [Google Scholar]
  • 77.Koren A, Garty I, Antonelli D, Katzuni E. Right ventricular cardiac dysfunction in beta-thalassemia major. Am J Dis Child. 141:93–96. PMID: 3788890. [PubMed] [Google Scholar]
  • 78.Minter K, Gladwin M. Pulmonary complications of sickle cell anemia. A need for increased recognition, treatment, and research. Am J Respir Crit Care Med. 2001;164:2016–2019. doi: 10.1164/ajrccm.164.11.2104101. doi:10.1164/rccm2104101. [DOI] [PubMed] [Google Scholar]
  • 79.Castro O, Hoque M, Brown B. Pulmonary hypertension in sickle cell disease: cardiac catheterization results and survival. Blood. 2003;101:1257–1261. doi: 10.1182/blood-2002-03-0948. doi:10.1182/blood-2002-03-0948. [DOI] [PubMed] [Google Scholar]
  • 80.Gladwin M, Sachdev V, Jison M, Shizukuda Y, Plehn J, Minter K, Brown B, Coles W, Nichols J, Ernst I, Hunter L, Blackwelder W, Schechter A, Rodgers G, Castro O, Ognibene F. Pulmonary hypertension as a risk factor for death in patients with sickle cell disease. N Engl J Med. 2004;350:886–895. doi: 10.1056/NEJMoa035477. PMID: 14985486. [DOI] [PubMed] [Google Scholar]
  • 81.Reiter C, Wang X, Tanus-Santos J, Hogg N, Cannon R, Schechter A, Gladwin M. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med. 2002;8:1383–1389. doi: 10.1038/nm1202-799. doi:10.1038/nm1202-799. [DOI] [PubMed] [Google Scholar]
  • 82.Klings E, Farber H. Role of free radicals in the pathogenesis of acute chest syndrome in sickle cell disease. Respir Res. 2001;2:280–285. doi: 10.1186/rr70. doi:10.1186/rr70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Aslan M, Ryan T, Adler B, Townes T, Parks D, Thompson J, Tousson A, Gladwin M, Patel R, Tarpey M, Batinic-Haberle I, White C, Freeman B. Oxygen radical inhibition of nitric oxide-dependent vascular function in sickle cell disease. Proc Natl Acad Sci (USA) 2001;98:15215–15220. doi: 10.1073/pnas.221292098. doi:10.1073/pnas.221292098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hsu L, Champion H, Campbell-Lee S, Bivalacqua T, Manci E, Diwan B, Schimel D, Cochard A, Wang X, Schechter A, Noguchi C, Gladwin M. Hemolysis in sickle cell mice causes pulmonary hypertension due to global impairment in nitric oxide bioavailability. Blood. 2007;109:3088–3098. doi: 10.1182/blood-2006-08-039438. doi:10.1182/blood-2006-08-039438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Silver R. Scleroderma. Clinical problems. The lungs. Rheum Dis Clin N Am. 1996;22:825–840. doi: 10.1016/s0889-857x(05)70303-3. PMID: 8923598. [DOI] [PubMed] [Google Scholar]
  • 86.Bolster M, Silver R. Lung disease in systemic sclerosis (scleroderma) Baillières Clin Rheum. 1993;7:79–97. doi: 10.1016/s0950-3579(05)80269-9. doi:10.1016/S0950-3579(05)80269-9. [DOI] [PubMed] [Google Scholar]
  • 87.Battle R, Davitt MC,SM, Buckley L, Leib E, Beglin P, Tischler M. Prevalence of pulmonary hypertension in limited and diffuse scleroderma. Chest. 1996;110:1515–1519. doi: 10.1378/chest.110.6.1515. PMID: 8989070. [DOI] [PubMed] [Google Scholar]
  • 88.Launay D, Mouthon L, Hachulla E, Pagnoux C, de Groote P, Remy-Jardin M, Matran R, Lambert M, Queyrel V, Morell-Dubois S, Guillevin L, Hatron P. Prevalence and characteristics of moderate to severe pulmonary hypertension in systemic sclerosis with and without interstitial lung disease. J Rheum. 2007;341:1005–1011. PMID: 17444586. [PubMed] [Google Scholar]
  • 89.Peacock A. Primary pulmonary hypertension. Thorax. 1999;54:1107–1118. doi: 10.1136/thx.54.12.1107. [Erratum in Thorax 2000 55:254]. PMID: 10567632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Williams J, Bodell P, Hosseini S, Tran H, Baldwin K. Haemodynamic sequelae of pulmonary fibrosis following intratracheal bleomycin in rats. Cardiovasc Res. 1992;26:401–408. doi: 10.1093/cvr/26.4.401. PMID: 1379125. [DOI] [PubMed] [Google Scholar]
  • 91.Ortiz L, Champion H, Lasky J, Gambelli F, Gozal E, Hoyle G, Beasley M, Hyman A, Friedman M, Kadowitz P. Enalapril protects mice from pulmonary hypertension by inhibiting TNF-mediated activation of NF-kappaB and AP-1. Am J Physiol Lung Cell Mol Physiol. 2002;282:L1209–L1221. doi: 10.1152/ajplung.00144.2001. doi:10.1152/ajplung.00144.2001. [DOI] [PubMed] [Google Scholar]
  • 92.Speich R, Jenni R, Opravil M, Pfab M, Russi E. Primary pulmonary hypertension in HIV infection. Chest. 1991;100:1268–1271. doi: 10.1378/chest.100.5.1268. PMID: 1935280. [DOI] [PubMed] [Google Scholar]
  • 93.Farber H. HIV-associated pulmonary hypertension. AIDS Clin Care. 2001;13:53–55, 59. PMID: 11710309. [PubMed] [Google Scholar]
  • 94.Mette S, Palevsky H, Pietra G, Williams T, Bruder E, Prestipino A, Patrick A, Wirth J. Primary pulmonary hypertension in association with human immunodeficiency virus infection. A possible viral etiology for some forms of hypertensive pulmonary arteriopathy. Am Rev Respir Dis. 1992;145:1196–1200. doi: 10.1164/ajrccm/145.5.1196. PMID: 1586065. [DOI] [PubMed] [Google Scholar]
  • 95.Galiè N, Manes A, Uguccioni L, Serafini F, De Rosa M, Branzi A, Magnani B. Primary pulmonary hypertension: insights into pathogenesis from epidemiology. Chest. 1998;114(3 Suppl):184S–194S. doi: 10.1378/chest.114.3_supplement.184s. PMID: 9741567. [DOI] [PubMed] [Google Scholar]
  • 96.Cool C, Kennedy D, Voelkel N, Tuder R. Pathogenesis and evolution of plexiform lesions in pulmonary hypertension associated with scleroderma and human immunodeficiency virus infection. Hum Pathol. 1997;28:434–442. doi: 10.1016/s0046-8177(97)90032-0. doi:10.1016/S0046-8177(97)90032-0. [DOI] [PubMed] [Google Scholar]
  • 97.Kanmogne G, Primeaux C, Grammas P. Induction of apoptosis and endothelin-1 secretion in primary human lung endothelial cells by HIV-1 gp120 proteins. Biochem Biophys Res Comm. 2005;333(4):1107–1115. doi: 10.1016/j.bbrc.2005.05.198. 333(334) doi:10.1016/j.bbrc.2005.05.198. [DOI] [PubMed] [Google Scholar]
  • 98.Marecki J, Cool C, Parr J, Beckey V, Luciw P, Tarantal A, Carville A, Shannon R, Cota-Gomez A, Tuder R, Voelkel N, Flores S. HIV-1 Nef is associated with complex pulmonary vascular lesions in SHIV-nef-infected macaques. Am J Respir Crit Care Med. 2006;174:437–445. doi: 10.1164/rccm.200601-005OC. doi:10.1164/rccm.200601-0050C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Caldwell R, Gadipatti R, Lane K, Shepherd V. HIV-1 TAT represses transcription of the bone morphogenic protein receptor-2 in U937 monocytic cells. J Leuk Biol. 2006;79:192–201. doi: 10.1189/jlb.0405194. doi:10.1189/jlb.0405194. [DOI] [PubMed] [Google Scholar]
  • 100.Cool C, Rai P, Yeager M, Hernandez-Saavedra D, Serls A, Bull T, Geraci M, Brown K, Routes J, Tuder R, Voelkel N. Expression of human herpesvirus 8 in primary pulmonary hypertension. N Engl J Med. 2003;349:1113–1122. doi: 10.1056/NEJMoa035115. PMID: 13679525. [DOI] [PubMed] [Google Scholar]
  • 101.Daibata M, Miyoshi I, Taguchi H, Matsubara H, Date H, Shimizu N, Ohtsuki Y. Absence of human herpesvirus 8 in lung tissues from Japanese patients with primary pulmonary hypertension. Respir Med. 2004;98:1231–1232. doi: 10.1016/j.rmed.2004.07.020. doi:10.1016/j.rmed.2004.07.020. [DOI] [PubMed] [Google Scholar]
  • 102.Katano H, Ito K, Shibuya K, Saji T, Sato Y, Sata T. Lack of human herpesvirus 8 infection in lungs of Japanese patients with primary pulmonary hypertension. J Infec Dis. 2005;191:743–745. doi: 10.1086/427824. doi:10.1086/427824. [DOI] [PubMed] [Google Scholar]
  • 103.Henke-Gendo C, Mengel M, Hoeper M, Alkharsah K, Schulz T. Absence of Kaposi's sarcoma-associated herpesvirus in patients with pulmonary arterial hypertension. Am J Respir Crit Care Med. 2005;172:1581–1585. doi: 10.1164/rccm.200504-546OC. doi:10.1164/rccm.20054-5460C. [DOI] [PubMed] [Google Scholar]
  • 104.Galambos C, Montgomery J, Jenkins F. No role for kaposi sarcoma-associated herpesvirus in pediatric idiopathic pulmonary hypertension. Ped Pulmonol. 2006;41:122–125. doi: 10.1002/ppul.20349. doi:10.1002/ppul.20349. [DOI] [PubMed] [Google Scholar]
  • 105.Bresser P, Cornelissen M, van der Bij W, van Noesel C, Timens W. Idiopathic pulmonary arterial hypertension in Dutch Caucasian patients is not associated with human herpes virus-8 infection. Respir Med. 2007;101:854–856. doi: 10.1016/j.rmed.2006.08.015. doi:10.1016/j.rmed.2006.08.015. [DOI] [PubMed] [Google Scholar]
  • 106.Bendayan D, Sarid R, Cohen A, Shitrit D, Shechtman I, Kramer M. Absence of human herpesvirus 8 DNA sequences in lung biopsies from Israeli patients with pulmonary arterial hypertension. Respiration. 2006 doi: 10.1159/000097495. Electronic publication; doi:10.1159/000097495. [DOI] [PubMed] [Google Scholar]
  • 107.Webb G, Gatzoulis M. Atrial septal defects in the adult: recent progress and overview. Circulation. 2006;114:1645–1653. doi: 10.1161/CIRCULATIONAHA.105.592055. doi:10.1161/CIRCULATIONAHA.105.592055. [DOI] [PubMed] [Google Scholar]
  • 108.Steele P, Fuster V, Cohen M, Ritter D, McGoon D. Isolated atrial septal defect with pulmonary vascular obstructive disease--long-term follow-up and prediction of outcome after surgical correction. Circulation. 1987;76:1037–1042. doi: 10.1161/01.cir.76.5.1037. PMID: 3664992. [DOI] [PubMed] [Google Scholar]
  • 109.Therrien J, Rambihar S, Newman B, Siminovitch K, Langleben D, Webb G, Granton J. Eisenmenger syndrome and atrial septal defect: nature or nurture. Can J Cardiol. 2006;22:1133–1136. doi: 10.1016/s0828-282x(06)70950-3. PMID: 17102831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Topper J, Gimbrone MJ. Blood flow and vascular gene expression: fluid shear stress as a modulator of endothelial phenotype. Molec Med Today. 1999;5:40–46. doi: 10.1016/s1357-4310(98)01372-0. doi:10.1016/S1357-4310(98)01372-0. [DOI] [PubMed] [Google Scholar]
  • 111.Sakao S, Taraseviciene-Stewart L, Lee J, Wood K, Cool C, Voelkel N. Initial apoptosis is followed by increased proliferation of apoptosis-resistant endothelial cells. FASEB J. 2005;19:1178–1180. doi: 10.1096/fj.04-3261fje. doi:10.1096/fj.04-3261fje. [DOI] [PubMed] [Google Scholar]
  • 112.Botney M. Role of hemodynamics in pulmonary vascular remodeling: implications for primary pulmonary hypertension. Am J Respir Crit Care Med. 1999;159:361–364. doi: 10.1164/ajrccm.159.2.9805075. PMID: 9927344. [DOI] [PubMed] [Google Scholar]
  • 113.Cool C, Groshong S, Oakey J, Voelkel N. Pulmonary hypertension: cellular and molecular mechanisms. Chest. 2005;128(6 Suppl):565S–571S. doi: 10.1378/chest.128.6_suppl.565S. doi:10.1378/chest.128.6_suppl.565S. [DOI] [PubMed] [Google Scholar]
  • 114.Ghorishi Z, Milstein J, Poulain F, Moon-Grady A, Tacy T, Bennett S, Fineman J, Eldridge M. Shear stress paradigm for perinatal fractal arterial network remodeling in lambs with pulmonary hypertension and increased pulmonary blood flow. Am J Physiol Heart Circ Physiol. 2007;292:H3006–H3018. doi: 10.1152/ajpheart.01012.2006. doi:10.1152/ajpheart.01012.2006. [DOI] [PubMed] [Google Scholar]
  • 115.García-Manero G, Schuster S, Patrick H, Martinez J. Pulmonary hypertension in patients with myelofibrosis secondary to myeloproliferative diseases. Am J Hematol. 1999;60:130–135. doi: 10.1002/(sici)1096-8652(199902)60:2<130::aid-ajh8>3.0.co;2-z. PMID: 9929105. [DOI] [PubMed] [Google Scholar]
  • 116.Dingli D, Utz J, Krowka M, Oberg A, Tefferi A. Unexplained pulmonary hypertension in chronic myeloproliferative disorders. Chest. 2001;120:801–808. doi: 10.1378/chest.120.3.801. PMID: 11555513. [DOI] [PubMed] [Google Scholar]
  • 117.Shishido N, Inoue A, Morishita H, Kodama M, Hasegawa K, Ishihara H, Aitani M, Kawabata S, Kimura K, Araki Y, Takada Y, Kikui M. A case of chronic thromboembolic pulmonary hypertension based on essential thrombocythemia. Nihon Kokyūki Gakkai Zasshi (J Jap Respir Soc) 2002;40:574–578. [Article in Japanese]. PMID: 12382421. [PubMed] [Google Scholar]
  • 118.Hill G, McClean D, Fraser R, Hart D, Crozier IS,J, Gibbons S. Pulmonary hypertension as a consequence of alveolar capillary plugging by malignant megakaryocytes in essential thrombocythaemia. Aust N Z J Med. 1996;26:852–853. doi: 10.1111/j.1445-5994.1996.tb00642.x. PMID: 9028525. [DOI] [PubMed] [Google Scholar]
  • 119.Albertson T, Walby W, Derlet R. Stimulant-induced pulmonary toxicity. Chest. 1995;108:1140–1149. doi: 10.1378/chest.108.4.1140. PMID: 7555128. [DOI] [PubMed] [Google Scholar]
  • 120.Murray R, Smialek J, Golle M, Albin R. Pulmonary artery medial hypertrophy in cocaine users without foreign particle microembolization. Chest. 1989;96:1050–1053. doi: 10.1378/chest.96.5.1050. PMID: 2805835. [DOI] [PubMed] [Google Scholar]
  • 121.Chin K, Channick R, Rubin L. Is methamphetamine use associated with idiopathic pulmonary arterial hypertension. Chest. 2006;130:1657–1663. doi: 10.1378/chest.130.6.1657. PMID: 17166979. [DOI] [PubMed] [Google Scholar]
  • 122.McDonnell P, Toye P, Hutchins G. Primary pulmonary hypertension and cirrhosis: are they related? Am Rev Respir Dis. 1983;127:437–441. doi: 10.1164/arrd.1983.127.4.437. PMID: 6838050. [DOI] [PubMed] [Google Scholar]
  • 123.Castro M, Krowka M, Schroeder D, Beck K, Plevak D, Rettke S, Cortese D, Wiesner R. Frequency and clinical implications of increased pulmonary artery pressures in liver transplant patients. Mayo Clin Proc Mayo Clinic. 1996;71:543–551. doi: 10.4065/71.6.543. PMID: 8642882. [DOI] [PubMed] [Google Scholar]
  • 124.Yang Y, Lin H, Lee WH,MC, Lee F, Chang F, Lee S. Portopulmonary hypertension: distinctive hemodynamic and clinical manifestations. J Gastroenterol. 2001;36:181–186. doi: 10.1007/s005350170126. doi:10.1007/s005350170126. [DOI] [PubMed] [Google Scholar]
  • 125.Robles A, Shure D. Gender issues in pulmonary vascular disease. Clin Chest Med. 2004;25:373–377. doi: 10.1016/j.ccm.2004.01.007. doi:10.1016/j.ccm.2004.01.007. [DOI] [PubMed] [Google Scholar]
  • 126.McQuillan L, Leung G, Marsden P, Kostyk S, Kourembanas S. Hypoxia inhibits expression of eNOS via transcriptional and posttranscriptional mechanisms. Am J Physiol. 1994;267(5 Pt 2):H1921–1927. doi: 10.1152/ajpheart.1994.267.5.H1921. [Erratum, Am J Physiol 1995;1268]. PMID: 7526714. [DOI] [PubMed] [Google Scholar]
  • 127.Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med. 1995;333:214–221. doi: 10.1056/NEJM199507273330403. PMID: 7540722. [DOI] [PubMed] [Google Scholar]
  • 128.Fagan K, Fouty B, Tyler R, Morris K, Hepler L, Sato K, LeCras T, Abman S, Weinberger H, Huang P, McMurtry I, Rodman D. The pulmonary circulation of homozygous or heterozygous eNOS-null mice is hyperresponsive to mild hypoxia. J Clin Invest. 1999;103:291–299. doi: 10.1172/JCI3862. PMID: 9916141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Champion H, Bivalacqua T, Greenberg S, Giles T, Hyman A, Kadowitz P. Adenoviral gene transfer of endothelial nitric-oxide synthase (eNOS) partially restores normal pulmonary arterial pressure in eNOS-deficient mice. Proc Natl. Acad Sci (USA) 2002;99:13248–13253. doi: 10.1073/pnas.182225899. doi:10.1073/pnas.182225899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Griffiths M, Evans T. Inhaled nitric oxide therapy in adults. N Engl J Med. 2005;353:2683–2695. doi: 10.1056/NEJMra051884. PMID: 16371634. [DOI] [PubMed] [Google Scholar]
  • 131.Galiè N, Ghofrani H, Torbicki A, Barst R, Rubin L, Badesch D, Fleming T, Parpia T, Burgess G, Branzi A, Grimminger F, Kurzyna M, Simonneau G, Group SUiPAHSS Sildenafil citrate therapy for pulmonary arterial hypertension. N Engl J Med. 2005;353:2148–2157. doi: 10.1056/NEJMoa050010. PMID: 16291984. [DOI] [PubMed] [Google Scholar]
  • 132.Barnett C, Machado R. Sildenafil in the treatment of pulmonary hypertension. Vasc Health Risk Manag. 2006;2:411–422. doi: 10.2147/vhrm.2006.2.4.411. PMID: 17323595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Mason N, Springall DB, Burke M, Pollock J, Mikhail G, Yacoub M, Polak J. High expression of endothelial nitric oxide synthase in plexiform lesions of pulmonary hypertension. J Pathol. 1998;185:313–318. doi: 10.1002/(SICI)1096-9896(199807)185:3<313::AID-PATH93>3.0.CO;2-8. doi:10.1002/(SICI)1096-9896(199807)185:3<313::AID-PATH93>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
  • 134.Gladwin M, Schechter A. NO contest: nitrite versus S-nitroso-hemoglobin. Circ Res. 2004;94:851–855. doi: 10.1161/01.RES.0000126697.64381.37. doi:10.1161/01.RES.0000126697.64381.37. [DOI] [PubMed] [Google Scholar]
  • 135.Bowers R, Cool C, Murphy R, Tuder R, Hopken M, Flores S, Voelkel N. Oxidative stress in severe pulmonary hypertension. Am J Respir Crit Care Med. 2004;169:764–769. doi: 10.1164/rccm.200301-147OC. doi:10.1164/rccm.200301-1470C. [DOI] [PubMed] [Google Scholar]
  • 136.Yildiz P, Oflaz H, Cine N, Erginel-Unaltuna N, Erzengin F, Yilmaz V. Gene polymorphisms of endothelial nitric oxide synthase enzyme associated with pulmonary hypertension in patients with COPD. Respir Med. 2003;97:1282–1288. doi: 10.1016/j.rmed.2003.06.001. doi:10.1016/j.rmed.2003.06.001. [DOI] [PubMed] [Google Scholar]
  • 137.Kawaguchi Y, Tochimoto A, Hara M, Kawamoto M, Sugiura T, Katsumata Y, Okada J, Kondo H, Okubo M, Kamatani N. NOS2 polymorphisms associated with the susceptibility to pulmonary arterial hypertension with systemic sclerosis: contribution to the transcriptional activity. Arthr Res Ther. 2006;8:R104. doi: 10.1186/ar1984. doi:10.1186/ar1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Yet S, Perrella M, Layne M, Hsieh C, Maemura K, Kobzik L, Wiesel P, Christou H, Kourembanas S, Lee M. Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. J Clin Invest. 1999;103:R23–R29. doi: 10.1172/JCI6163. PMID: 10207174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Christou H, Morita T, Hsieh C, Koike H, Arkonac B, Perrella M, Kourembanas S. Prevention of hypoxia-induced pulmonary hypertension by enhancement of endogenous heme oxygenase-1 in the rat. Circ Res. 2000;86:1224–1229. doi: 10.1161/01.res.86.12.1224. PMID: 10864912. [DOI] [PubMed] [Google Scholar]
  • 140.Zuckerbraun B, Chin B, Wegiel B, Billiar T, Czsimadia E, Rao J, Shimoda L, Ifedigbo E, Kanno S, Otterbein L. Carbon monoxide reverses established pulmonary hypertension. J Exp Med. 2006;203:2109–2119. doi: 10.1084/jem.20052267. doi:10.1084/jem.20052267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chunyu Z, Junbao D, Dingfang B, Hui Y, Xiuying T, Chaoshu T. The regulatory effect of hydrogen sulfide on hypoxic pulmonary hypertension in rats. Biochem Biophys Res Comm. 2003;302:810–816. doi: 10.1016/s0006-291x(03)00256-0. doi:10.1016/S0006-291X(03)00256-0. [DOI] [PubMed] [Google Scholar]
  • 142.Li X, Du J, Ding Y, Jin H, Bu D, Tang X, Tang C. Impact of hydrogen sulfide donor on experimental pulmonary hypertension induced by high pulmonary flow and endogenous carbon monoxide/heme oxygenase pathway. J Peking Univ Health Sci. 2006;38:135–139. PMID: 16617353. [PubMed] [Google Scholar]
  • 143.Gerber J, Voelkel N, Nies A, McMurtry I, Reeves J. Moderation of hypoxic vasoconstriction by infused arachidonic acid: role of PGI2. J Appl Physiol. 1980;49:107–112. doi: 10.1152/jappl.1980.49.1.107. PMID: 6995413. [DOI] [PubMed] [Google Scholar]
  • 144.Tuder R, Cool C, Geraci M, Wang J, Abman S, Wright L, Badesch D, Voelkel N. Prostacyclin synthase expression is decreased in lungs from patients with severe pulmonary hypertension. Am J Respir Crit Care Med. 1999;159:1925–1932. doi: 10.1164/ajrccm.159.6.9804054. PMID: 10351941. [DOI] [PubMed] [Google Scholar]
  • 145.Christman B, McPherson C, Newman J, King G, Bernard G, Groves B, Loyd J. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med. 1992;327:70–75. doi: 10.1056/NEJM199207093270202. PMID: 1603138. [DOI] [PubMed] [Google Scholar]
  • 146.Strauss W, Edelman J. Prostanoid therapy for pulmonary arterial hypertension. Clin Chest Med. 2007;28:127–142. doi: 10.1016/j.ccm.2006.11.008. ix. doi:10.1016/j.ccm.2006.11.008. [DOI] [PubMed] [Google Scholar]
  • 147.Hassoun P, Thappa V, Landman M, Fanburg B. Endothelin 1 mitogenic activity on pulmonary artery smooth muscle cells and release from hypoxic endothelial cells. Proc Soc Exper Biol Med Soc Exper Biol Med. 1992;199:165–170. doi: 10.3181/00379727-199-43342. PMID: 1741408. [DOI] [PubMed] [Google Scholar]
  • 148.Stelzner T, O'Brien R, Yanagisawa M, Sakurai T, Sato K, Webb S, Zamora M, McMurtry I, Fisher J. Increased lung endothelin-1 production in rats with idiopathic pulmonary hypertension. Am J Physiol. 1992;262:L614–620. doi: 10.1152/ajplung.1992.262.5.L614. PMID: 1534203. [DOI] [PubMed] [Google Scholar]
  • 149.Allen S, Chatfield B, Koppenhafer S, Schaffer M, Wolfe R, Abman S. Circulating immunoreactive endothelin-1 in children with pulmonary hypertension. Association with acute hypoxic pulmonary vasoreactivity. Am Rev Respir Dis. 1993;148:519–522. doi: 10.1164/ajrccm/148.2.519. PMID: 8342919. [DOI] [PubMed] [Google Scholar]
  • 150.Giaid A, Yanagisawa M, Langleben D, Michel R, Levy R, Shennib H, Kimura S, Masaki T, Duguid W, Stewart D. Expression of endothelin-1 in the lungs of patients with pulmonary hypertension. N Engl J Med. 1993;328:1732–1739. doi: 10.1056/NEJM199306173282402. PMID: 8497283. [DOI] [PubMed] [Google Scholar]
  • 151.Langleben D. Endothelin receptor antagonists in the treatment of pulmonary arterial hypertension. Clin Chest Med. 2007;28:117–125. doi: 10.1016/j.ccm.2006.11.002. viii (review). doi:10.1016/j.ccm.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 152.Cox C, Linden J, Said S. VIP elevates platelet cyclic AMP (cAMP) levels and inhibits in vitro platelet activation induced by platelet-activating factor (PAF) Peptides. 1984;5:325–328. doi: 10.1016/0196-9781(84)90228-6. doi:10.1016/0196-9781(84)90228-6. [DOI] [PubMed] [Google Scholar]
  • 153.Maruno K, Absood A, Said S. VIP inhibits basal and histamine-stimulated proliferation of human airway smooth muscle cells. Am J Physiol. 1995;268:L1047–1051. doi: 10.1152/ajplung.1995.268.6.L1047. PMID: 7541947. [DOI] [PubMed] [Google Scholar]
  • 154.Petkov V, Mosgoeller W, Ziesche R, Raderer M, Stiebellehner L, Vonbank K, Funk G, Hamilton G, Novotny C, Burian B, Block L. Vasoactive intestinal peptide as a new drug for treatment of primary pulmonary hypertension. J Clin Invest. 2003;111:1339–1346. doi: 10.1172/JCI17500. doi:10.1172/JCI200317500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Said S, Hamidi S, Dickman K, Szema A, Lyubsky S, Lin R, Jiang Y, Chen J, Waschek J, Kort S. Moderate pulmonary arterial hypertension in male mice lacking the vasoactive intestinal peptide gene. Circulation. 2007;115:1260–1268. doi: 10.1161/CIRCULATIONAHA.106.681718. doi:10.1161/CIRCULATIONAHA.106.681718. [DOI] [PubMed] [Google Scholar]
  • 156.Söderman C, Eriksson L, Juhlin-Dannfelt A, Lundberg J, Broman L, Holmgren A. Effect of vasoactive intestinal polypeptide (VIP) on pulmonary ventilation-perfusion relationships and central haemodynamics in healthy subjects. Clin Physiol. 1993;13:677–685. doi: 10.1111/j.1475-097x.1993.tb00482.x. PMID: 8119061. [DOI] [PubMed] [Google Scholar]
  • 157.Gunaydin S, Imai Y, Takanashi Y, Seo K, Hagino I, Chang D, Shinoka T. The effects of vasoactive intestinal peptide on monocrotaline induced pulmonary hypertensive rabbits following cardiopulmonary bypass: a comparative study with isoproteronol and nitroglycerine. Cardiovasc Surg. 2002;10:138–145. doi: 10.1016/s0967-2109(01)00126-0. doi:10.1016/S0967-2109(01)00126-0. [DOI] [PubMed] [Google Scholar]
  • 158.Haydar S, Sarti J, Grisoni E. Intravenous vasoactive intestinal polypeptide lowers pulmonary-to-systemic vascular resistance ratio in a neonatal piglet model of pulmonary arterial hypertension. J Ped Surg. 2007;42:758–764. doi: 10.1016/j.jpedsurg.2006.12.026. doi:10.1016/j.jpedsurg.2006.12.026. [DOI] [PubMed] [Google Scholar]
  • 159.Tuder R, Flook B, Voelkel N. Increased gene expression for VEGF and the VEGF receptors KDR/Flk and Flt in lungs exposed to acute or to chronic hypoxia. Modulation of gene expression by nitric oxide. J Clin Invest. 1995;95:1798–1807. doi: 10.1172/JCI117858. PMID: 7706486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Tuder R, Chacon M, Alger L, Wang J, Taraseviciene-Stewart L, Kasahara Y, Cool C, Bishop A, Geraci M, Semenza G, Yacoub M, Polak J, Voelkel N. Expression of angiogenesis-related molecules in plexiform lesions in severe pulmonary hypertension: Evidence for a process of disordered angiogenesis. J Pathol. 2001;195:367–374. doi: 10.1002/path.953. doi:10.1002/path.953. [DOI] [PubMed] [Google Scholar]
  • 161.Wanstall J, Gambino A, Jeffery T, Cahill M, Bellomo D, Hayward N, Kay G. Vascular endothelial growth factor-B-deficient mice show impaired development of hypoxic pulmonary hypertension. Cardiovasc Res. 2002;55:361–368. doi: 10.1016/s0008-6363(02)00440-6. doi:10.1016/S0008-6363(02)00440-6. [DOI] [PubMed] [Google Scholar]
  • 162.Taraseviciene-Stewart L, Kasahara Y, Alger L, Hirth P, Mc Mahon GW,J, Voelkel N, Tuder R. Inhibition of the VEGF receptor 2 combined with chronic hypoxia causes cell death-dependent pulmonary endothelial cell proliferation and severe pulmonary hypertension. FASEB J. 2001;15:427–438. doi: 10.1096/fj.00-0343com. PMID: 11156958. [DOI] [PubMed] [Google Scholar]
  • 163.Hansmann G, Wagner R, Schellong S, Perez V, Urashima T, Wang L, Sheikh A, Suen R, Stewart D, Rabinovitch M. Pulmonary arterial hypertension is linked to insulin resistance and reversed by peroxisome proliferator-activated receptor-gamma activation. Circulation. 2007;115:1275–1284. doi: 10.1161/CIRCULATIONAHA.106.663120. doi:10.1161/CIRCULATION AHA.106.663. [DOI] [PubMed] [Google Scholar]
  • 164.Schermuly R, Pullamsetti S, Kwapiszewska G, Dumitrascu R, Tian X, Weissmann N, Ghofrani H, Kaulen C, Dunkern T, Schudt C, Voswinckel R, Zhou J, Samidurai A, Klepetko W, Paddenberg R, Kummer W, Seeger W, Grimminger F. Phosphodiesterase 1 upregulation in pulmonary arterial hypertension: target for reverse-remodeling therapy. Circulation. 2007;115:2331–2339. doi: 10.1161/CIRCULATIONAHA.106.676809. doi:10.1161/CIRCULATIONAHA.106.676809. [DOI] [PubMed] [Google Scholar]
  • 165.McMurtry M, Archer S, Altieri D, Bonnet S, Haromy A, Harry G, Bonnet S, Puttagunta L, Michelakis E. Gene therapy targeting survivin selectively induces pulmonary vascular apoptosis and reverses pulmonary arterial hypertension. J Clin Invest. 2005;115:1479–1491. doi: 10.1172/JCI23203. doi:10.1172/JCI23203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Greenway S, van Suylen R, Du Marchie Sarvaas G, Kwan E, Ambartsumian N, Lukanidin E, Rabinovitch M. S100A4/Mts1 produces murine pulmonary artery changes resembling plexogenic arteriopathy and is increased in human plexogenic arteriopathy. Am J Pathol. 2004;164:253–262. doi: 10.1016/S0002-9440(10)63115-X. PMID: 14695338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Merklinger S, Wagner R, Spiekerkoetter E, Hinek A, Knutsen R, Kabir M, Desai K, Hacker S, Wang L, Cann G, Ambartsumian N, Lukanidin E, Bernstein D, Husain M, Mecham R, Starcher B, Yanagisawa H, Rabinovitch M. Increased fibulin-5 and elastin in S100A4/Mts1 mice with pulmonary hypertension. Circ Res. 2005;97:596–604. doi: 10.1161/01.RES.0000182425.49768.8a. doi:10.1161/01.RES.0000182425.49768.8a. [DOI] [PubMed] [Google Scholar]
  • 168.Yu Y, Fantozzi I, Remillard C, Landsberg J, Kunichika N, Platoshyn O, Tigno D, Thistlethwaite P, Rubin L, Yuan J. Enhanced expression of transient receptor potential channels in idiopathic pulmonary arterial hypertension. Proc Natl Acad Sci (USA) 2004;101:13861–13866. doi: 10.1073/pnas.0405908101. doi:10.1073/pnas.0405908101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Shimokubo T, Sakata J, Kitamura K, Kangawa K, Matsuo H, Eto T. Augmented adrenomedullin concentrations in right ventricle and plasma of experimental pulmonary hypertension. Life Sci. 1995;57:1771–1779. doi: 10.1016/0024-3205(95)02155-c. doi:10.1016/0024-3205(95)02155-C. [DOI] [PubMed] [Google Scholar]
  • 170.Nishikimi T, Nagata S, Sasaki T, Tomimoto S, Matsuoka H, Takishita S, Kitamura K, Miyata A, Matsuo H, Kangawa K. Plasma concentrations of adrenomedullin correlate with the extent of pulmonary hypertension in patients with mitral stenosis. Heart. 1997;78:390–395. doi: 10.1136/hrt.78.4.390. PMID: 9404257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Kakishita M, Nishikimi T, Okano Y, Satoh T, Kyotani S, Nagaya N, Fukushima K, Nakanishi N, Takishita S, Miyata A, Kangawa K, Matsuo H, Kunieda T. Increased plasma levels of adrenomedullin in patients with pulmonary hypertension. Clini Sci. 1999;96:33–39. PMID: 9857104. [PubMed] [Google Scholar]
  • 172.Loscalzo J, Kohane I, Barabasi A. Human disease classification in the postgenomic era: A complex systems approach to human pathobiology. Molec Syst Biol. 2007;3:124. doi: 10.1038/msb4100163. doi:10.1038/msb4100163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Shimokawa H, Takeshita A. Rho-kinase is an important therapeutic target in cardiovascular medicine. Arterio Thromb Vasc Biol. 2005;25:1767–1775. doi: 10.1161/01.ATV.0000176193.83629.c8. doi:10.1161/01.ATV.0000176193.83629.c8. [DOI] [PubMed] [Google Scholar]
  • 174.Weigand L, Sylvester J, Shimoda L. Mechanisms of endothelin-1-induced contraction in pulmonary arteries from chronically hypoxic rats. Am J Physiol Lung Cell Molec Physiol. 2006;290:L284–L290. doi: 10.1152/ajplung.00449.2004. doi:10.1152/ajplung.00449.2004. [DOI] [PubMed] [Google Scholar]
  • 175.Barman S. Vasoconstrictor effect of endothelin-1 on hypertensive pulmonary arterial smooth muscle involves Rho kinase and protein kinase C. Am J Physiol Lung Cell Molec Physiol. 2007 doi: 10.1152/ajplung.00101.2006. Electronic publication. doi:10.1152/ajplung.00101.2006. [DOI] [PubMed] [Google Scholar]
  • 176.Liu Y, Suzuki Y, Day R, Fanburg B. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res. 2004;95:579–586. doi: 10.1161/01.RES.0000141428.53262.a4. doi:10.1161/01.RES.0000141428.53262.a4. [DOI] [PubMed] [Google Scholar]
  • 177.Li M, Liu Y, Dutt P, Fanburg B, Toksoz D. Inhibition of serotonin-induced mitogenesis, migration, and ERK MAPK nuclear translocation in vascular smooth muscle cells by atorvastatin. Am J Physiol Lung Cell Molec Physiol. 2007;293:L463–L471. doi: 10.1152/ajplung.00133.2007. Electronic publication ahead of print. doi:10.1152/ajplung.00133.2007. [DOI] [PubMed] [Google Scholar]
  • 178.Takemoto M, Sun J, Hiroki J, Shimokawa H, Liao J. Rho-kinase mediates hypoxia-induced downregulation of endothelial nitric oxide synthase. Circulation. 2002;106:57–62. doi: 10.1161/01.cir.0000020682.73694.ab. doi:10.1161/01.CIR.0000020682.73694.AB. [DOI] [PubMed] [Google Scholar]
  • 179.Nagaoka T, Morio Y, Casanova N, Bauer N, Gebb S, McMurtry I, Oka M. Rho/Rho kinase signaling mediates increased basal pulmonary vascular tone in chronically hypoxic rats. Am J Physiol Lung Cell Molec Physiol. 2004;287:L665–L672. doi: 10.1152/ajplung.00050.2003. doi:10.1152/ajplung.00050.2003. [DOI] [PubMed] [Google Scholar]
  • 180.Oka M, Homma N, Taraseviciene-Stewart L, Morris K, Kraskauskas D, Burns N, Voelkel N, McMurtry I. Rho kinase-mediated vasoconstriction is important in severe occlusive pulmonary arterial hypertension in rats. Circ Res. 2007;100:923–929. doi: 10.1161/01.RES.0000261658.12024.18. doi:10.1161/01.RES.0000261658.12024.08. [DOI] [PubMed] [Google Scholar]
  • 181.Fagan K, Oka M, Bauer N, Gebb S, Ivy D, Morris K, McMurtry I. Attenuation of acute hypoxic pulmonary vasoconstriction and hypoxic pulmonary hypertension in mice by inhibition of Rho-kinase. Am J Physiol Lung Cell Molec Physiol. 2004;287:L656–L664. doi: 10.1152/ajplung.00090.2003. doi:10.1152/ajplung.00090.2003. [DOI] [PubMed] [Google Scholar]
  • 182.Abe K, Shimokawa H, Morikawa K, Uwatoku T, Oi K, Matsumoto Y, Hattori T, Nakashima Y, Kaibuchi K, Sueishi K, Takeshit A. Long-term treatment with a Rho-kinase inhibitor improves monocrotaline-induced fatal pulmonary hypertension in rats. Circ Res. 2004;94:385–393. doi: 10.1161/01.RES.0000111804.34509.94. doi:10.1161/01.RES.0000111804.34509.94. [DOI] [PubMed] [Google Scholar]
  • 183.Abe K, Tawara S, Oi K, Hizume T, Uwatoku T, Fukumoto Y, Kaibuchi K, Shimokawa H. Long-term inhibition of Rho-kinase ameliorates hypoxia-induced pulmonary hypertension in mice. J CardiovascPharmacol. 2006;48:280–285. doi: 10.1097/01.fjc.0000248244.64430.4a. doi:10.1097/01.fjc.0000248244.64430.4a. [DOI] [PubMed] [Google Scholar]
  • 184.Jiang B, Tawara S, Abe K, Takaki A, Fukumoto Y, Shimokawa H. Acute vasodilator effect of fasudil, a Rho-kinase inhibitor, in monocrotaline-induced pulmonary hypertension in rats. J Cardiovasc Pharmacol. 2007;49:85–89. doi: 10.1097/FJC.0b013e31802df112. doi: 10.1097/FJC.0b013e31802df112. [DOI] [PubMed] [Google Scholar]
  • 185.Li F, Xia W, Li A, Zhao C, Sun R. Inhibition of rho kinase attenuates high flow induced pulmonary hypertension in rats. Chinese Med J. 2007;120:22–29. PMID: 17254483. [PubMed] [Google Scholar]
  • 186.Li F, Xia W, Li A, Zhao C, Sun R. Long-term inhibition of Rho kinase with fasudil attenuates high flow induced pulmonary artery remodeling in rats. Pharmacol Res. 2007;55:64–71. doi: 10.1016/j.phrs.2006.10.009. doi:10.1016/j.phrs.2006.10.009. [DOI] [PubMed] [Google Scholar]
  • 187.Fukumoto Y, Matoba T, Ito A, Tanaka H, Kishi T, Hayashidani S, Abe K, Takeshita A, Shimokawa H. Acute vasodilator effects of a Rho-kinase inhibitor, fasudil, in patients with severe pulmonary hypertension. Heart. 2005;91:391–392. doi: 10.1136/hrt.2003.029470. doi:10.1136/hrt.2003.029470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Ishikura K, Yamada N, Ito M, Ota S, Nakamura M, Isaka N, Nakano T. Beneficial acute effects of rho-kinase inhibitor in patients with pulmonary arterial hypertension. Circ J. 2006;70:174–178. doi: 10.1253/circj.70.174. doi:10.1253/circj.70.174. [DOI] [PubMed] [Google Scholar]
  • 189.Nishimura T, Faul J, Berry G, Vaszar L, Qiu D, Pearl R, Kao P. Simvastatin attenuates smooth muscle neointimal proliferation and pulmonary hypertension in rats. Am J Respir Crit Care Med. 2002;166:1403–1408. doi: 10.1164/rccm.200203-268OC. doi:10.1164/rccm.200203-2680C. [DOI] [PubMed] [Google Scholar]
  • 190.Hu H, Sung A, Zhao G, Shi L, Qiu D, Nishimura T, Kao P. Simvastatin enhances bone morphogenetic protein receptor type II expression. Biochem Biophys Res Comm. 2006;339:59–64. doi: 10.1016/j.bbrc.2005.10.187. doi:10.1016/j.bbrc.2005.10.187. [DOI] [PubMed] [Google Scholar]
  • 191.Moudgil R, Michelakis E, Archer S. The role of k+ channels in determining pulmonary vascular tone, oxygen sensing, cell proliferation, and apoptosis: implications in hypoxic pulmonary vasoconstriction and pulmonary arterial hypertension. Microcirculation. 2006;13:615–632. doi: 10.1080/10739680600930222. doi:10.1080/10739680600930222. [DOI] [PubMed] [Google Scholar]
  • 192.Yuan X, Wang J, Juhaszova M, Gaine S, Rubin L. Attenuated K+ channel gene transcription in primary pulmonary hypertension. Lancet. 1998;351:726–727. doi: 10.1016/S0140-6736(05)78495-6. PMID: 9504523. [DOI] [PubMed] [Google Scholar]
  • 193.Remillard C, Tigno D, Platoshyn O, Burg E, Brevnova E, Conger D, Nicholson A, Rana B, Channick R, Rubin L, O'Connor D, Yuan J. Function of Kv1.5 channels and genetic variations of KCNA5 in patients with idiopathic pulmonary arterial hypertension. Am J Physiol Cell Physiol. 2007;292:C1837–C1853. doi: 10.1152/ajpcell.00405.2006. doi:10.1152/ajpcell.00405.2006. [DOI] [PubMed] [Google Scholar]
  • 194.Weir E, Reeve H, Huang J, Michelakis E, Nelson D, Hampl V, Archer S. Anorexic agents aminorex, fenfluramine, and dexfenfluramine inhibit potassium current in rat pulmonary vascular smooth muscle and cause pulmonary vasoconstriction. Circulation. 1996;94:2216–2220. doi: 10.1161/01.cir.94.9.2216. PMID: 8901674. [DOI] [PubMed] [Google Scholar]
  • 195.Cogolludo A, Moreno L, Lodi F, Frazziano G, Cobeño L, Tamargo J, Perez-Vizcaino F. Serotonin inhibits voltage-gated K+ currents in pulmonary artery smooth muscle cells: role of 5-HT2A receptors, caveolin-1, and KV1.5 channel internalization. Circ Res. 2006;98:931–938. doi: 10.1161/01.RES.0000216858.04599.e1. doi:10.1161/01.RES.0000216858.04599.el. [DOI] [PubMed] [Google Scholar]
  • 196.Cogolludo A, Moreno L, Bosca L, Tamargo J, Perez-Vizcaino F. Thromboxane A2-induced inhibition of voltage-gated K+ channels and pulmonary vasoconstriction: role of protein kinase Czeta. Circ Res. 2003;93:656–663. doi: 10.1161/01.RES.0000095245.97945.FE. doi:10.1161/01.RES.0000095245.9749.FE. [DOI] [PubMed] [Google Scholar]
  • 197.Michelakis E, McMurtry M, Sonnenberg B, Archer S. The NO - K+ channel axis in pulmonary arterial hypertension. Activation by experimental oral therapies. Adv Exp Biol Med. 2003;543:293–322. doi: 10.1007/978-1-4419-8997-0_21. PMID: 14713130. [DOI] [PubMed] [Google Scholar]
  • 198.Young K, Ivester C, West J, Carr M, Rodman D. BMP signaling controls PASMC KV channel expression in vitro and in vivo. Am J Physiol Lung Cell Mol Physiol. 2006;290:L841–L848. doi: 10.1152/ajplung.00158.2005. doi:10.1152/ajplung.00158.2005. [DOI] [PubMed] [Google Scholar]
  • 199.Fantozzi I, Platoshyn O, Wong A, Zhang S, Remillard C, Furtado M, Petrauskene O, Yuan J. Bone morphogenetic protein-2 upregulates expression and function of voltage-gated K+ channels in human pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2006;291:L993–L1004. doi: 10.1152/ajplung.00191.2005. doi:10.1152/ajplung.00191.2005. [DOI] [PubMed] [Google Scholar]
  • 200.Pozeg Z, Michelakis E, McMurtry M, Thébaud B, Wu X, Dyck J, Hashimoto K, Wang S, Moudgil R, Harry G, Sultanian R, Koshal A, Archer S. In vivo gene transfer of the O2-sensitive potassium channel Kv1.5 reduces pulmonary hypertension and restores hypoxic pulmonary vasoconstriction in chronically hypoxic rats. Circulation. 2003;107:2037–2044. doi: 10.1161/01.CIR.0000062688.76508.B3. doi:10.1161/01.CIR.0000062688.76508.B3. [DOI] [PubMed] [Google Scholar]
  • 201.Brindle N, Saharinen P, Alitalo K. Signaling and functions of angiopoietin-1 in vascular protection. Circ Res. 2006;98:1014–1023. doi: 10.1161/01.RES.0000218275.54089.12. doi:10.1161/01.RES.0000218275.54089.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Kugathasan L, Dutly A, Zhao Y, Deng Y, Robb M, Keshavjee S, Stewart D. Role of angiopoietin-1 in experimental and human pulmonary arterial hypertension. Chest. 2005;128(6 Suppl):633S–642S. doi: 10.1378/chest.128.6_suppl.633S. doi:10.1378/chest.128.6_suppl.633S. [DOI] [PubMed] [Google Scholar]
  • 203.Dewachter L, Adnot S, Fadel E, Humbert M, Maitre B, Barlier-Mur A, Simonneau G, Hamon M, Naeije R, Eddahibi S. Angiopoietin/Tie2 pathway influences smooth muscle hyperplasia in idiopathic pulmonary hypertension. Am J Respir Crit Care Med. 2006;1743:1025–1033. doi: 10.1164/rccm.200602-304OC. doi:10.1164/rccm.200602-3040C. [DOI] [PubMed] [Google Scholar]
  • 204.Kido M, Du L, Sullivan C, Deutsch R, Jamieson S, Thistlethwaite P. Gene transfer of a TIE2 receptor antagonist prevents pulmonary hypertension in rodents. J Thorac Cardiovasc Surg. 2005;129:268–276. doi: 10.1016/j.jtcvs.2004.09.013. doi:10.1016/j.jtcvs.2004.09.013. [DOI] [PubMed] [Google Scholar]
  • 205.Eickelberg O, Yeager M, Grimminger F. The tantalizing triplet of pulmonary hypertension-BMP receptors, serotonin receptors, and angiopoietins. Cardiovasc Res. 2003;60:465–467. doi: 10.1016/j.cardiores.2003.10.017. doi:10.1016/j.cardiores.2003.10.017. [DOI] [PubMed] [Google Scholar]
  • 206.Zhao Y, Campbell A, Robb M, Ng D, Stewart D. Protective role of angiopoietin-1 in experimental pulmonary hypertension. Circ Res. 2003;92:984–991. doi: 10.1161/01.RES.0000070587.79937.F0. doi:10.1161/01. RES.0000070587.79939.FO. [DOI] [PubMed] [Google Scholar]
  • 207.Li X, Everson W, Smart E. Caveolae, lipid rafts, and vascular disease. Trends Cardiovasc Med. 2005;15:92–96. doi: 10.1016/j.tcm.2005.04.001. doi:10.1016/j.tcm.2005.04.001. [DOI] [PubMed] [Google Scholar]
  • 208.Achcar R, Demura Y, Rai P, Taraseviciene-Stewart L, Kasper M, Voelkel N, Cool C. Loss of caveolin and heme oxygenase expression in severe pulmonary hypertension. Chest. 2006;129:696–705. doi: 10.1378/chest.129.3.696. doi:10.1378/chest.129.3.696. [DOI] [PubMed] [Google Scholar]
  • 209.Zhao Y, Liu Y, Stan R, Fan L, Gu Y, Dalton N, Chu P, Peterson K, Ross J, Chien K. Defects in caveolin-1 cause dilated cardiomyopathy and pulmonary hypertension in knockout mice. Proc Natl Acad Sci (USA) 2002;99:11375–11380. doi: 10.1073/pnas.172360799. doi: 10.1073/pnas.172360799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Drab M, Verkade P, Elger M, Kasper M, Lohn M, Lauterbach B, Menne J, Lindschau C, Mende F, Luft F, Schedl A, Haller H, Kurzchalia T. Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice. Science. 2001;293:2449–2452. doi: 10.1126/science.1062688. PMID: 11498544. [DOI] [PubMed] [Google Scholar]
  • 211.Shaul P, Smart E, Robinson L, German Z, Yuhanna I, Ying Y, Anderson R, Michel T. Acylation targets endothelial nitric-oxide synthase to plasmalemmal caveolae. J Biol Chem. 1996;271:6518–6522. doi: 10.1074/jbc.271.11.6518. PMID: 8626455. [DOI] [PubMed] [Google Scholar]
  • 212.Chun M, Liyanage U, Lisanti M, Lodish H. Signal transduction of a G protein-coupled receptor in caveolae: colocalization of endothelin and its receptor with caveolin. Proc Natl Acad Sci (USA) 1994;91:11728–11732. doi: 10.1073/pnas.91.24.11728. PMID: 7972131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Sehgal P, Mukhopadhyay S, Xu F, Patel K, Shah M. Dysfunction of Golgi tethers, SNAREs, and SNAPs in monocrotaline-induced pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1526–L1542. doi: 10.1152/ajplung.00463.2006. doi:10.1152/ajplung.00463.2006. [DOI] [PubMed] [Google Scholar]
  • 214.Ramos M, Lamé M, Segall H, Wilson D. The BMP type II receptor is located in lipid rafts, including caveolae, of pulmonary endothelium in vivo and in vitro. Vasc Pharmacol. 2006;44(1):50–59. doi: 10.1016/j.vph.2005.09.007. doi:10.1016/j.vph.2005.09.007. [DOI] [PubMed] [Google Scholar]
  • 215.Wiechen K, Sers C, Agoulnik A, Arlt K, Dietel M, Schlag P, Schneider U. Down-regulation of caveolin-1, a candidate tumor suppressor gene, in sarcomas. Am J Pathol. 2001;158:833–839. doi: 10.1016/S0002-9440(10)64031-X. PMID: 11238032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Gillespie M, Goldblum S, Cohen D, McClain C. Interleukin 1 bioactivity in the lungs of rats with monocrotaline-induced pulmonary hypertension. Proc Soc Exp Biol Med. 1988;187(1):26–32. doi: 10.3181/00379727-187-42632. PMID: 3124119. [DOI] [PubMed] [Google Scholar]
  • 217.Perkett E, Lyons R, Moses H, Brigham K, Meyrick B. Transforming growth factor-beta activity in sheep lung lymph during the development of pulmonary hypertension. J Clin Invest. 1990;86:1459–1464. doi: 10.1172/JCI114862. PMID: 2243124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Chand N, Altura B. Acetylcholine and bradykinin relax intrapulmonary arteries by acting on endothelial cells: role in lung vascular diseases. Science. 1981;213:1376–1379. doi: 10.1126/science.7268440. PMID: 7268440. [DOI] [PubMed] [Google Scholar]
  • 219.Itoh T, Nagaya N, Ishibashi-Ueda H, Kyotani S, Oya H, Sakamaki F, Kimura H, Nakanishi N. Increased plasma monocyte chemoattractant protein-1 level in idiopathic pulmonary arterial hypertension. Respirol. 2006;11:158–163. doi: 10.1111/j.1440-1843.2006.00821.x. doi:10.1111/j.1440-1843.2006.00821.x. [DOI] [PubMed] [Google Scholar]
  • 220.Balabanian K, Foussat A, Dorfmüller P, Durand-Gasselin I, Capel F, Bouchet-Delbos L, Portier A, Marfaing-Koka A, Krzysiek R, Rimaniol A, Simonneau G, Emilie D, Humbert M. CX(3)C chemokine fractalkine in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2002;165:1419–1425. doi: 10.1164/rccm.2106007. doi:10.1164/rccm.2106007. [DOI] [PubMed] [Google Scholar]
  • 221.Dorfmüller P, Zarka V, Durand-Gasselin I, Monti G, Balabanian K, Garcia G, Capron F, Coulomb-Lherminé A, Marfaing-Koka A, Simonneau G, Emilie D, Humbert M. Chemokine RANTES in severe pulmonary arterial hypertension. Am J Respir Crit Care Med. 2002;165:534–539. doi: 10.1164/ajrccm.165.4.2012112. doi:10.1164/rccm.2012112. [DOI] [PubMed] [Google Scholar]
  • 222.Stenmark K, James S, Voelkel N, Toews W, Reeves J, Murphy R. Leukotriene C4 and D4 in neonates with hypoxemia and pulmonary hypertension. N Engl J Med. 1983;309:77–80. doi: 10.1056/NEJM198307143090204. PMID: 6855869. [DOI] [PubMed] [Google Scholar]
  • 223.Voelkel N, Tuder R, Wade K, Höper M, Lepley R, Goulet J, Koller B, Fitzpatrick F. Inhibition of 5-lipoxygenase-activating protein (FLAP) reduces pulmonary vascular reactivity and pulmonary hypertension in hypoxic rats. J Clin Invest. 1996;97:2491–2498. doi: 10.1172/JCI118696. PMID: 8647941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Wright L, Tuder R, Wang J, Cool C, Lepley R, Voelkel N. 5-Lipoxygenase and 5-lipoxygenase activating protein (FLAP) immunoreactivity in lungs from patients with primary pulmonary hypertension. Am J Respir Crit Care Med. 1998;157:219–229. doi: 10.1164/ajrccm.157.1.9704003. PMID: 9445303. [DOI] [PubMed] [Google Scholar]
  • 225.Jones J, Walker J, Song Y, Weiss N, Cardoso W, Tuder R, Loscalzo J, Zhang Y. Effect of 5-lipoxygenase on the development of pulmonary hypertension in rats. Am J Physiol Heart Circ Physiol. 2004;286:H1775–H1784. doi: 10.1152/ajpheart.00281.2003. doi:10.1152/ajpheart.00281.2003. [DOI] [PubMed] [Google Scholar]
  • 226.Rabinovitch M. Pathobiology of pulmonary hypertension. Extracellular matrix. Clin Chest Med. 2001;22:433–449. doi: 10.1016/s0272-5231(05)70282-3. viii. PMID: 11590839. [DOI] [PubMed] [Google Scholar]
  • 227.Rabinovitch M. Elastase and the pathobiology of unexplained pulmonary hypertension. Chest. 1998;114(3 Suppl):213S–224S. doi: 10.1378/chest.114.3_supplement.213s. PMID: 9741572. [DOI] [PubMed] [Google Scholar]
  • 228.Zhu L, Wigle D, Hinek A, Kobayashi J, Ye C, Zuker M, Dodo H, Keeley F, Rabinovitch M. The endogenous vascular elastase that governs development and progression of monocrotaline-induced pulmonary hypertension in rats is a novel enzyme related to the serine proteinase adipsin. J Clin Invest. 1994;94:1163–1171. doi: 10.1172/JCI117432. PMID: 8083356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Jones P, Rabinovitch M. Tenascin-C is induced with progressive pulmonary vascular disease in rats and is functionally related to increased smooth muscle cell proliferation. Circ Res. 1996;79:1131–1142. doi: 10.1161/01.res.79.6.1131. PMID: 8943951. [DOI] [PubMed] [Google Scholar]
  • 230.Ihida-Stansbury K, McKean D, Lane K, Loyd J, Wheeler L, Morrell N, Jones P. Tenascin-C is induced by mutated BMP type II receptors in familial forms of pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 2006;291:L694–L702. doi: 10.1152/ajplung.00119.2006. doi:10.1152/ajplung.00119.2006. [DOI] [PubMed] [Google Scholar]
  • 231.Mitani Y, Zaidi S, Dufourcq P, Thompson K, Rabinovitch M. Nitric oxide reduces vascular smooth muscle cell elastase activity through cGMP-mediated suppression of ERK phosphorylation and AML1B nuclear partitioning. FASEB J. 2000;14:805–814. doi: 10.1096/fasebj.14.5.805. PMID: 10744637. [DOI] [PubMed] [Google Scholar]
  • 232.Cowan K, Jones P, Rabinovitch M. Elastase and matrix metalloproteinase inhibitors induce regression, and tenascin-C antisense prevents progression, of vascular disease. J Clin Invest. 2000;105:21–34. doi: 10.1172/JCI6539. PMID: 10619858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Cowan K, Heilbut A, Humpl T, Lam C, Ito S, Rabinovitch M. Complete reversal of fatal pulmonary hypertension in rats by a serine elastase inhibitor. Nat Med. 2000;6:698–702. doi: 10.1038/76282. doi:10.1038/76282. [DOI] [PubMed] [Google Scholar]
  • 234.Zaidi S, You X, Ciura S, Husain M, Rabinovitch M. Overexpression of the serine elastase inhibitor elafin protects transgenic mice from hypoxic pulmonary hypertension. Circulation. 2002;105:516–521. doi: 10.1161/hc0402.102866. doi:10.1161/hc0402.102866. [DOI] [PubMed] [Google Scholar]
  • 235.Medhora M, Bousamra Mn, Zhu D, Somberg L, Jacobs E. Upregulation of collagens detected by gene array in a model of flow-induced pulmonary vascular remodeling. Am J Physiol Heart Circ Physiol. 2002;282:H414–422. doi: 10.1152/ajpheart.00292.2001. doi:10.1152/ajpheart.00292.2001. [DOI] [PubMed] [Google Scholar]

RESOURCES