Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2011 May;163(1):125–140. doi: 10.1111/j.1476-5381.2010.01164.x

New perspectives for the treatment of pulmonary hypertension

Reshma S Baliga 1, Raymond J MacAllister 2, Adrian J Hobbs 1,2
PMCID: PMC3085874  PMID: 21175577

Abstract

Pulmonary hypertension (PH) is a debilitating disease with a poor prognosis. Therapeutic options remain limited despite the introduction of prostacyclin analogues, endothelin receptor antagonists and phosphodiesterase 5 inhibitors within the last 15 years; these interventions address predominantly the endothelial and vascular dysfunctionS associated with the condition, but simply delay progression of the disease rather than offer a cure. In an attempt to improve efficacy, emerging approaches have focused on targeting the pro-proliferative phenotype that underpins the pulmonary vascular remodelling in the lung and contributes to the impaired circulation and right heart failure. Many novel targets have been investigated and validated in animal models of PH, including modulation of guanylate cyclases, phosphodiesterases, tyrosine kinases, Rho kinase, bone morphogenetic proteins signalling, 5-HT, peroxisome proliferator activator receptors and ion channels. In addition, there is hope that combinations of such treatments, harnessing and optimizing vasodilator and anti-proliferative properties, will provide a further, possibly synergistic, increase in efficacy; therapies directed at the right heart may also offer an additional benefit. This overview highlights current therapeutic options, promising new therapies, and provides the rationale for a combination approach to treat the disease.

LINKED ARTICLES

This article is part of a themed issue on Respiratory Pharmacology. To view the other articles in this issue visit http://dx.doi.org/10.1111/bph.2011.163.issue-1

Keywords: pulmonary hypertension, cyclic GMP, guanylate cyclase, phosphodiesterase, endothelin-1, prostacyclin, tyrosine kinase, growth factors, ion channels

Introduction

Pulmonary hypertension (PH) is a multi-factorial, progressive disease with substantial mortality and morbidity. Despite recent improvements in treatment, the mortality associated with PH remains high, with survival at 2 years from diagnosis approximately 85% (Thenappan et al., 2007; National Pulmonary Hypertension Centres of the UK and Ireland, 2008). Unfortunately, there remains no cure and clinical worsening is merely delayed, not prevented, by therapy (McLaughlin et al., 2009a,b;). However, advances in the understanding of PH aetiology and pathology have yielded novel concepts, drug targets and treatment strategies that may improve the management of patients with the disease. This overview will provide a brief overview of the current therapeutic options and highlight some of these emerging therapeutic approaches which hold promise for alleviating this debilitating disorder with an extremely poor prognosis.

Pulmonary arterial hypertension

Since the World Health Organization (WHO) oversaw the initial categorization of PH into ‘primary’ and ‘secondary’ forms in the early 1970 s, based on the presence or absence of identifiable causes or risk factors, the clinical classification of PH has undergone numerous modifications. The goal of the current organization of PH is to group together different manifestations of the disease, sharing similarities in pathophysiological mechanisms, clinical presentation and therapeutic approaches (Figure 1; Simonneau et al., 2009).

Figure 1.

Figure 1

Current classification of pulmonary hypertension.

Pulmonary arterial hypertension (PAH) is a subset of pulmonary hypertensive syndromes, defined by a resting mean pulmonary artery pressure (PAP) > 25 mm Hg, pulmonary vascular resistance (PVR) > 3 Wood units and pulmonary wedge pressure <15 mmHg, in the absence of other causes of PH (Archer et al., 1998). PAH is primarily a disease of the small pulmonary arteries, characterized by vascular proliferation, remodelling and progressive increases in PVR, leading ultimately to right ventricular failure and death (Voelkel et al., 2006). PVR increases are attributed to endothelial dysfunction, resulting in vasoconstriction, remodelling of the pulmonary vessel wall and thrombosis in situ (Budhiraja et al., 2004). However, the haemodynamic aberrations represent only one aspect of PAH, and enhanced proliferation, decreased apoptosis and a shift to glycolytic metabolism in pulmonary artery smooth muscle cell fibroblasts and endothelial cells is now recognized as central to the pathogenesis of the disease. This pro-proliferative phenotype is underpinned, at least in a subset of PAH patients, by aberrations in transforming growth factor (TGF)β signalling. This concept has been brought about by the identification of loss-of-function mutations in the bone morphogenetic protein receptor-2 (BMPR2) gene that promotes cell proliferation and suppress apoptosis in 80% of familial PAH patients (Lane et al., 2000). BMPR2 mutations are, however, uncommon in non-familial PAH (10–20%) and even in familial PAH; disease penetrance is low (only 25% of carriers develop the disease; Newman et al., 2004). Single nucleotide polymorphisms (SNPs) in ion channels (e.g. Kv1.5, TRPC6) and transporter genes (SERT; molecular target nomenclature follows Alexander et al., 2009) can also predispose to PAH and a multi-hit hypothesis, a complex interaction between genes and environment, has been proposed to explain the low disease penetrance of genetic mutations in PAH [Figure 2; (Yuan and Rubin, 2005; Newman et al., 2008)].

Figure 2.

Figure 2

Schematic representation of the current and emerging therapeutic targets for pulmonary hypertension outlined in this review. NP, natriuretic peptide; NPR, natriuretic peptide receptor; PGI2, prostacyclin; ET-1, endothelin-1; BMPR, bone morphogenetic protein receptor; Kv, voltage-sensitive potassium channel; KATP, ATP-sensitive potassium channel; TRPC6, transient receptor potential channel C6; VOCC, voltage operated calcium channel; SERT, 5-HT transporter; NEP, neutral endopeptidase; NEPi, NEP inhibitor; BH4, tetrahydrobiopterin; eNOS, endothelial NO synthase; TKR, tyrosine kinase receptor; ROCK, Rho-associated kinase; sGC, soluble guanylate cyclase; ERA, endothelin receptor antagonist; PDGF, platelet derived growth factor; FGF, fibroblast growth factor; VEGF, vascular endothelial growth factor; AC, adenylate cyclase; IP, prostacyclin receptor; MAPK, mitogen-activated protein kinase; PI3K, phosphoinositide-3-kinase; PKC, protein kinase C; Src, Src kinase; JAK/STAT, Janus kinase/signal transducer and activator of transcription; PDE, phosphodiesterase; PDEi, PDE inhibitor.

Current therapy

To date, clinical evaluation of novel therapies for PH has been confined primarily to the PAH subset; there is only limited randomized clinical trial evidence for other forms of PH, for example, associated with lung disease and chronic thromboembolic PH (CTEPH), and research to establish effectiveness of these therapies across PH classes is needed. Nonetheless, advances made in the treatment of PAH are also likely to be effective, to a greater or lesser extent, in patients with aetiologically distinct forms of PH.

Many PH patients receive a background therapy of warfarin, diuretics, digoxin and oxygen (McLaughlin et al., 2009a,b;). Anti-coagulant therapy with warfarin appears to have beneficial effects on survival, at least based on findings of observational studies (Johnson et al., 2006), while diuretics limit oedema, and digoxin and oxygen provide symptomatic relief. Frontline therapy aims at enhancing vasodilatation, predominantly by inhibiting the bioactivity of endothelin-1 (ET-1), a potent endothelium-derived vasoconstrictor, or by augmenting the vasodilator properties of nitric oxide (NO) and prostacyclin (PGI2).

Ca2+ channel blockers

L-type Ca2+ channel blockers (CCB), such as. nifedipine, diltiazem or amlodipine, can be effective in patients that respond to a one-time vasodilator challenge with a >20% fall in PAP and no decline in cardiac output (Rich and Brundage, 1987). Notably, only 10–15% of patients with iPAH meet these criteria and only half of those will receive sustained clinical and haemodynamic benefit. Patients who respond to CCB therapy, however, have an excellent 5 year survival rate (94%) as compared with those that do not respond [55% survival; (Sitbon et al., 2005)].

Prostacyclin analogues

PGI2 and thromboxane A2 (TXA2) are arachidonic acid metabolites with opposing vasoactivity. In PAH, the balance is shifted towards vasoconstrictor, pro-proliferative TXA2 from vasodilator, anti-proliferative PGI2 (Christman et al., 1992; Tuder et al., 1999). This relative impairment in PGI2-dependent signalling in PAH leads to the development of analogues that would mimic the cytoprotective activity of this prostanoid and restore the balance between PGI2 and TXA2. The beneficial activity of prostacyclin (analogues) in PH is presumed to be via activation of the Gs-coupled IP receptor, despite the fact that these compounds can activate other prostanoid receptors (Narumiya et al., 1999); however, recent evidence also supports a role for peroxisome proliferator activated receptors (PPARs; see below) in the underlying mechanism.

Epoprostenol was the first treatment targeted directly at PAH pathology, and has a proven survival advantage (Rubin et al., 1990; Barst et al., 1996; Badesch et al., 2009). Its poor stability, cost and the need for parenteral infusion, however, have led to the development of more stable analogues with more favourable means of administration and pharmacokinetic profiles; iloprost, trepostinil and beraprost are all used in the clinical management of PAH patients.

Endothelin receptor antagonists (ERAs)

Plasma levels of ET-1, a potent vasoconstrictor and mitogenic agent, are significantly elevated and correlate with disease severity in PAH (Rubens et al., 2001). The action of ET-1 is complex and mediated via two cell-surface, G-protein-coupled receptors; ETA receptors on vascular smooth muscle cells cause vasoconstriction and proliferation, while ETB receptors on endothelial cells stimulate NO and prostacyclin release, but on vascular smooth muscle cells induce vasoconstriction and mitogenesis. Endothelin receptor antagonists such as bosentan (dual ETA/ETB), ambrisentan (ETA > ETB) and sitaxsentan (ET1A > > ETB) have been shown to improve pulmonary haemodynamics, exercise capacity and reduce PAH symptoms (Williamson et al., 2000; Channick et al., 2001; Barst et al., 2004; Galie et al., 2005a); these drugs are of clinical benefit, particularly in PAH associated with connective tissue disease where they are often used as the initial treatment option (Denton et al., 2008). However, a positive survival effect, and relative comparisons between ET-1 receptor selective agents (selective ETA antagonists should possess a theoretical advantage in not preventing the production of NO via ETB receptor activation on endothelial cells) are still lacking. Macitentan, a novel ETA/ETB receptor antagonist, is currently in a phase III trial in PAH (SERAPHIN), after producing a promising haemodynamic profile in a smaller Phase II trial in hypertensive patients (Raja, 2010).

PDE5 inhibitors

PDEs are homologous enzymes that facilitate the breakdown of the second messengers, cAMP and/or cGMP (Bender and Beavo, 2006). There are 11 distinct PDE families, with each typically consisting of several isoforms and/or splice variants. Molecules blocking the activity of this family of enzymes, collectively known as PDE inhibitors, have been a major focus of drug development, particularly for cardiovascular disease. Indeed, in the vasculature, PDE inhibitors exert several favourable effects including vasodilatation, inhibition of smooth muscle proliferation and prevention of platelet aggregation (Bender and Beavo, 2006).

Blockade of PDE5, which metabolizes cGMP exclusively, lowers systemic and pulmonary artery pressure under physiological conditions in animals and humans (Jackson et al., 1999; Madhani et al., 2006). Moreover, in animal models and patients with PH, PDE5 inhibitors cause larger reductions in pulmonary than systemic vascular resistance, thereby exhibiting relative selectivity for the pulmonary vasculature (Klinger et al., 2006; Baliga et al., 2008). In accord, PDE5 is found in abundance in the pulmonary vasculature and both expression and activity are elevated in PAH (Murray et al., 2002; Sebkhi et al., 2003). This favourable vasoactive profile of PDE5 inhibitors has culminated in the development and approval of sildenafil as a first-line therapy for PH; the drug elicits an improvement in several indices of disease severity including pulmonary artery pressure, cardiac index, exercise capacity and WHO functional class (Galie et al., 2005b). Sildenafil also appears to produce an overall beneficial effect on survival (Galie et al., 2009b). Tadalafil, an analogous PDE5 inhibitor with a longer half-life, has also been recently licensed for the treatment of PAH (Galie et al., 2009a). A third PDE5 inhibitor, vardenafil, is currently undergoing Phase III evaluation for the same indication.

Inhaled NO

The inhalation of exogenous NO gas decreases PAP and improves oxygenation in diverse forms of PAH, and is particularly effective in neonates suffering from persistent pulmonary hypertension (PPHN; Roberts et al., 1992; Macrae et al., 2004; Creagh-Brown et al., 2009). However, long-term therapy with inhaled NO is complicated by the instability of NO gas, concerns regarding the development of methaemoglobinaemia (as NO binds avidly to, and oxidizes, the haem moiety) and marked rebound pulmonary hypertension following cessation of therapy (Ichinose et al., 2004).

Novel therapeutic strategies

cGMP signalling

PDE5 inhibitors are an undoubted therapeutic advance, but their effects on PAP are small (approximately 5 mm Hg reduction). A significant cohort of PH patients does not respond to sildenafil treatment and in many individuals indices of disease severity do not differ from placebo approximately 12 months after initiation of therapy. Moreover, in patients who respond well to sildenafil, there is often a dose-dependent systemic hypotension that limits the beneficial effects of the drug. There is no evidence to suggest that the newer PDE5 inhibitors have a substantially greater effect than sildenafil, or that sildenafil resistant patients respond to other PDE5 inhibitors. Thus, there remains considerable opportunity to optimize interventions targeting cGMP-dependent signalling to improve the treatment of PH.

In PH, pulmonary vascular cGMP levels are decreased, either through impairment of NO bioavailability, guanylyl cyclase inactivation [enzymes that generate cGMP in response to NO and natriuretic peptides (Hobbs, 1997; Ahluwalia et al., 2004; Potter et al., 2006) or enhanced cGMP degradation by PDEs (Crawley et al., 1992; Zhao et al., 1992; Steudel et al., 1997; Archer et al., 1998). Accordingly, therapeutics targeted at augmenting cGMP levels have been shown to have therapeutic value in PH, either in animal models or patients with the disease.

NO donors

Attempts have been made to bypass the short half-life and indiscriminate chemical reactivity of (inhaled) NO, by developing more stable NO donors (e.g. NONOates), which spontaneously release defined amounts of NO when exposed to physiological pH. Daily nebulization with NONOates (e.g. diethylenetriamineNONOate; DEA-NO) has shown to be effective in animal models of PH (Vanderford et al., 1994; Hampl et al., 1996). Similarly, older NO donors such as glyceryl trinitrate administered by inhalation have been shown to be effective in reducing PAP in small clinical samples (Goyal et al., 2006). Thus, delivery of NO via more sophisticated donor drugs may still prove to be efficacious in PH patients. Nonetheless, concerns regarding the lack of pulmonary selectivity, cGMP-independent cytotoxic effects and rebound pulmonary hypertension remain relevant.

Endothelial NO synthase augmentation

A further mechanism that may be exploited to treat PH is to improve endogenous NO bioavailability by augmenting the activity of endothelial NO synthase (eNOS). Expression and activity of eNOS, and the availability of a key redox co-factor, tetrahydrobiopterin (BH4), are largely reduced in PH (Giaid and Saleh, 1995; Shaul et al., 1997; Le Cras et al., 1998; Khoo et al., 2005), and mice with gene deletions in these systems are predisposed to the disease (Fagan et al., 1999; Nandi et al., 2005; Leiper et al., 2007). However, under some circumstances, eNOS may be hyperactive in the pulmonary circulation in PH and, as a result of inadequate supply of BH4, the enzyme uncouples to form superoxide rather than NO (Zhao et al., 2009); this has the doubly detrimental effect of scavenging NO and producing direct cytotoxicity.

Several approaches focusing on eNOS/BH4 have been evaluated for efficacy in PH. First, supplementation with BH4 itself, or more promisingly an orally active, more stable form (6R-BH4), is effective in augmenting endogenous BH4 levels, restoring eNOS expression and reversing systemic hypertension (Landmesser et al., 2003); similar effects may be achievable in PH. Second, the ‘eNOS coupling agent’, cicletanine, has shown modest beneficial effects in animal models of PH and humans with the disease (Jin et al., 1992; Saadjian et al., 1998), presumably by coordinating eNOS activity with BH4 supply/binding and favouring the generation of NO over superoxide (although increasing the endogenous formation of PGI2 and natriuretic peptides may also underlie these positive effects). Cicletanine is currently under phase II evaluation in patients with PAH. Thirdly, eNOS transcription enhancers may prove advantageous in PH, as they have shown in animal models to reverse the vascular remodelling and cardiac hypertrophy associated with left-sided heart failure (Westermann et al., 2009), ischaemia-reperfusion injury [i.e. myocardial infarction (Sasaki et al., 2006; Frantz et al., 2009)], and atherosclerosis (Wohlfart et al., 2008). Finally, the Pulmonary Hypertension and Cell Therapy trial, currently recruiting, is designed to test the safety and tolerability of autologous progenitor cell-based gene delivery of human eNOS in patients with severe PAH. This study may pave the way for more cell-based therapies for PAH, particularly because endothelial progenitor cells are thought to play a role in the pathogenesis of the disease (Toshner et al., 2009), are a predictive biomarker and a novel therapeutic target (Yip et al., 2008; Sun et al., 2009; Toshner et al., 2009; Fadini et al., 2010).

Soluble GC activators

In order to harness the beneficial, cytoprotective effects of cGMP while circumventing the potentially detrimental cGMP-independent effects of NO, the development of directly acting sGC ‘agonists’ has progressed in rapid fashion. Soluble GC appears a good target in PH as the expression and activity of the enzyme is up-regulated in order to compensate for decreased NO bioavailability (Black et al., 2001; Schermuly et al., 2008; de Frutos et al., 2009) and genetic deletion of the enzyme results in an exaggerated response to hypoxia-induced PH (Vermeersch et al., 2007).

Two different classes of sGC ‘agonist’ have been developed. First, sGC ‘stimulators’ or ‘haem-dependent activators’ (e.g. BAY 41-2272, BAY 41-8543, BAY 63-2521, riociguat) which stimulate the native Fe2+-sGC and synergize with NO (Stasch et al., 2002a,b;). Second, sGC ‘activators’ or ‘haem-independent activators’ (e.g. BAY 58-2667, cinaciguat; HMR-1766, ataciguat) which activate the proposed Fe3+ or haem-free form of the enzyme and are additive with NO (Belik, 2009; Schmidt et al., 2009; Stasch & Hobbs, 2009).

Both classes of drugs have been shown to have favourable effects on experimental PH (Dumitrascu et al., 2006; Chester et al., 2009; Weissmann et al., 2009). Riociguat, an orally active sGC ‘stimulator’ is currently in Phase III trials for determination of clinical effectiveness in idiopathic PAH and CTEPH (Ghofrani et al., 2010). However, a limitation of this sGC-centric strategy may be its lack of pulmonary selectivity, as shown by the systemic hypotension observed in earlier trials (Grimminger et al., 2009). This is perhaps not unexpected. Soluble GC ‘stimulators’ synergize with NO and will therefore augment NO-dependent dilatation in all vascular beds. Moreover, in PH the bioavailability of NO in the pulmonary vasculature is known to be impaired, entailing that this synergy will predominate in the systemic, rather than pulmonary circulation. Nonetheless, these agents have exhibited a favourable profile in Phase II trials and offer a novel approach to treat PH; this therapeutic value may increase with inhalation or combination therapy to target the sGC ‘stimulators’ to the pulmonary circulation (Evgenov et al., 2007). In addition, Phase III evaluation of sGC ‘activators’ (e.g. cinaciguat) that preferentially trigger the oxidized form of the enzyme, thought to be more prominent in diseased vasculature, may provide a more pulmonary-centred therapeutic approach in PH. Indeed, cinaciguat has already exhibited a favourable profile in patients with left-sided heart failure (Lapp et al., 2009).

Natriuretic peptides

Atrial natriuretic peptide and brain natriuretic peptide are synthesized by and released from cardiac atrial and ventricular tissue, respectively, in response to stretch and elicit falls in blood volume and blood pressure (Ahluwalia et al., 2004; Potter et al., 2006). A third member of the family, C-type natriuretic peptide, is released from the vascular endothelium and regulates local blood flow in a paracrine fashion (Ahluwalia and Hobbs, 2005). Each natriuretic peptide acts on specific cell-surface natriuretic peptide receptors (NPR) in the vasculature which possess guanylate cyclase functionality. The increase in tissue cGMP in response to NPR activation brings about several cytoprotective effects including natriuresis, vasodilatation, and anti-hypertrophic and anti-proliferative activity [particularly in the heart (Oliver et al., 1997)].

Genetic deletion of NPRs is associated with PH (Klinger et al., 1999; Zhao et al., 1999; Kuhn, 2004), while administration of exogenous natriuretic peptides has been shown to reduce hypoxia-induced PH (Klinger et al., 1999); such observations provide the rationale for therapeutic modulation of natriuretic peptide signalling in PH. However, the short plasma half-life and negligible oral bioavailability make natriuretic peptides poor candidates for drug therapy. An alternative strategy is to increase endogenous natriuretic peptide levels by inhibiting the enzyme neutral endopeptidase (NEP), a major degradative pathway for natriuretic peptides (and other bioactive peptides) in the circulation (Okolicany et al., 1992). This strategy has been proven to be effective in animal models both as monotherapy (Klinger et al., 1993) and using the NEP inhibitor racecadotril in combination with a PDE5 inhibitor (Baliga et al., 2008). Indeed, our data, both in vitro and in vivo, suggest that PDE5 is pivotal in terminating the cyclic GMP-dependent signalling in response to natriuretic peptides in the pulmonary vasculature, whereas other PDE isozymes regulate the vasorelaxant activity of natriuretic peptides in the systemic circulation (Baliga et al., 2008). Therefore, by inhibiting PDE5 in PH, in which circulating natriuretic peptide concentrations are raised, it is possible to target the pulmonary vasculature and reduce pulmonary pressure. These observations explain the mechanism underpinning the pulmonary selectivity of PDE5 inhibitors and suggest that in PH, the release of natriuretic peptides represents a cytoprotective mechanism that reduces disease progression. This thesis is in accord with studies reporting increased expression and activity of PDE5 in the pulmonary circulation of patients with PH (Wharton et al., 2005), that the beneficial effects of PDE5 inhibitors in models of PH are blunted in NPR-A knockout mice (Zhao et al., 2003) and that, in patients with PH and animal models of the disease, acute infusion of natriuretic peptides in the presence of sildenafil synergistically reduces pulmonary artery pressure (Preston et al., 2004; Klinger et al., 2006). Thus, the therapeutic potential of manipulating natriuretic peptide bioactivity to reverse the haemodynamic abnormalities associated with PH holds great promise. This is true not only for the haemodynamic dysfunction, but also for attenuating the pulmonary vascular re-modelling that also characterises the disease. Natriuretic peptides inhibit pulmonary vascular smooth muscle proliferation and TGFβ-induced extracellular matrix expression in vitro, and prevent structural changes in vivo in animal models of PH (Jin et al., 1990; Klinger et al., 1998; 1999; Chen et al., 2006; Li et al., 2007).

The strategy of targeting neutral endopeptidase for the treatment of PH may also have the added benefit of slowing the breakdown of other protective peptides that will contribute to efficacy, including adrenomedullin and vasoactive intestinal peptide; both have been shown to be up-regulated in PH and to reverse disease progression in animal models (Shimokubo et al., 1995; Gunaydin et al., 2002; Matsui et al., 2004; Qi et al., 2007; Said et al., 2007). However, NEP is also important in the metabolism of ET-1, which may offset some of its beneficial activity.

Other PDE inhibitors

PDE5 has received considerable attention in the context of PH due to the success of sildenafil and other selective inhibitors. However, other isozymes (e.g. PDE1 and PDE3) are also up-regulated in PAH, and might be suitable targets for therapy.

PDE 1 and PDE 3 (and splice-variants thereof) have been implicated in pulmonary vascular homeostasis and PH (Bender and Beavo, 2006). These enzymes hydrolyse cGMP and cAMP, although the PDE1A/1B splice variants have a higher affinity for cGMP (Bender and Beavo, 2006). PDE1A and PDE1C expression and activity are up-regulated in animal models of PH and in tissues from patients with the disease (Evgenov et al., 2006; Murray et al., 2007; Schermuly et al., 2007). Moreover, the selective PDE1 inhibitor, 8-methoxymethyl-isobutyl-1-methyl xanthine, reduces proliferation of human vascular smooth muscle cells (Rybalkin et al., 2002) and reverses the haemodynamic and morphological aberrations associated with monocrotaline and hypoxia-induced PH (Schermuly et al., 2007).

PDE 3A/3B expression and activity are also enhanced in PH (Murray et al., 2002), and the presence of this ‘cGMP-inhibited’ PDE might underlie the synergistic cytoprotective activity of NO and prostacyclin in PH, and explain the benefit of co-administration of therapies promoting these pathways concomitantly [i.e. sildenafil and iloprost (Wilkens et al., 2001)]. Indeed, a dual PDE3/4 inhibitor reverses monocrotaline-induced PH and synergizes with iloprost (Schermuly et al., 2004; Dony et al., 2008). The PDE3 inhibitor milrinone is currently being investigated for safety and efficacy in treatment of PPHN, but despite this potential, the increased mortality associated with the use of PDE3 inhibitors in (left) heart failure (Amsallem et al., 2005) has limited the therapeutic enthusiasm for this approach in PH.

Anti-proliferative pathways

PAH is characterised by a shift in the proliferative/apoptotic balance and enhanced glycolytic metabolism (Mandegar et al., 2004). Several growth factors, including platelet derived growth factor (PDGF), fibroblast growth factor 2, epidermal growth factor, vascular endothelial growth factor (VEGF) and, more recently, the non-canonical Wnt pathway have been implicated in the abnormal proliferation in PH (Oka et al., 2007b; Hassoun, 2009; Izikki et al., 2009). Levels of PDGF and its tyrosine kinase receptor PDGFR, are elevated in PAH patient lung samples (Perros et al., 2008) and HIV-associated PH samples (Humbert et al., 1998). VEGF levels are also increased in plexiform lesions in PAH patients (Cool et al., 1999). These growth factors act as potent mitogens and chemoattractants, and through their transmembrane tyrosine kinase receptor pathways activate major proliferative signalling pathways such as the ras-mitogen activated protein kinase (MAPK) cascade, resulting in proliferation, migration and resistance to apoptosis (Hassoun, 2009). Consequently, this has led to increased interest in translation of anti-proliferative strategies, often originally developed for cancer therapy, to PAH patients.

Tyrosine kinase inhibitors

Imatinib (Gleevac) was initially developed as an anti-cancer therapy, predominantly chronic myelogenous leukemia, via inhibition of the oncogenic tyrosine kinase Bcr-Alb, but was later found to block the PDGFR and improve experimental PH (Schermuly et al., 2005; Klein et al., 2008). Several case studies of end-stage PH patients also suggest that treatment with imatinib can improve clinical conditions (Ghofrani et al., 2005; Patterson et al., 2006; Souza et al., 2006; Tapper et al., 2009; Ten et al., 2009; Chhina et al., 2010). This has led to a Phase III randomized, placebo-controlled clinical trial of imatinib in PAH (IMPRES), from which results are eagerly awaited. Nonetheless, there is some concern that long-term of imatinib could be associated with left ventricular dysfunction and heart failure (Kerkela et al., 2006). Accordingly, other tyrosine kinase inhibitors have been developed and evaluated. Two such molecules are sunitinib and sorafenib, multi-kinase inhibitors, blocking PDGF, VEGF and other pro-proliferative signalling pathways. These molecules are currently being evaluated for safety and tolerability in Phase I, and are undoubtedly efficacious in animal models of PH (Klein et al., 2008; Gomberg-Maitland et al., 2010). However, it remains to be seen if such molecules are also associated with cardiotoxicity.

Several further molecules, often originally developed as anti-cancer agents, have also been investigated in animal models of PH, with positive outcomes, and are likely to lead to clinical evaluation in patients with the disease, particularly those molecules that are already licensed medicines. These include cell cycle inhibitors [e.g. rapamycin (Paddenberg et al., 2007)], anti-apoptotic drugs [e.g. survivin inhibitors (McMurtry et al., 2005)] and elastase inhibitors (Merklinger et al., 2005).

Rho kinase inhibitors

The Rho kinase pathway participates in vasoconstriction elicited by numerous agents involved in PAH, including 5-HT, ET-1 and TXA2 (Oka et al., 2008). Rho is a small monomeric GTPase which activates Rho-associated kinase (ROCK) which in turn phosphorylates and inhibits myosin light chain phosphatase, which leads to prolonged, refractory vasoconstriction. Rho and ROCK also mediate smooth muscle cell proliferation, in a 5-HT-BMPR dependent pathway, and have been found to be elevated in smooth muscle cells from PAH patients (Do e Z et al., 2009). Rho-kinase inhibitors have been shown to reduce PH in many animal models, including the monocrotaline rat, fawn hooded rats and chronic hypoxia/SUGEN exposure (Oka et al., 2007a; Mouchaers et al., 2010). In humans, Rho-kinase inhibition with fasudil shows modest, immediate reductions in PVR, but this inhibitor of Rho-kinase has to be administered by nebulization, that is, directly into the lungs, to avoid systemic hypotension (Ishikura et al., 2006; Fujita et al., 2010).

Bone morphogenetic protein signalling pathway

The discovery of the association between mutations in BMPR2 and PAH has led to increased interest in the BMP signalling pathway as a therapeutic target (Lane et al., 2000). BMPR2 is a constitutively active serine-threonine kinase and a member of the TGFβ superfamily. In response to ligand, BMPR2 heterodimerises with one of four BMPR1 receptors (BMPR1A, BMPR1B, Alk1, Alk2), and phosphorylates the internal domain, triggering the cytosolic Smad protein signalling cascade (Yang et al., 2005). Activation of the MAPK system [i.e. p38, extracellular signal regulated kinase 1/2 (ERK 1 /2) or Jun – N-terminal kinase] may also be an underlying mechanism. While BMPR2 mutations are relatively rare in non-familial PAH, dysfunctional BMPR signalling is often seen in PAH. For example, the expression of BMPR2 protein is markedly reduced in the lungs of patients with idiopathic PAH with no detectable mutation in BMPR2 (Atkinson et al., 2002), and cells isolated from PAH patients show altered response to BMP signalling (Morrell et al., 2001). Reduced expression of BMPR2 is also found in the lungs of rats with monocrotaline-induced pulmonary hypertension (Morty et al., 2007)

Results of BMPR2-targetted therapy in animal models have been mixed. Adenoviral gene delivery of BMPR2 failed to reverse monocrotaline-induced PH (McMurtry et al., 2007), but intravascular administration of BMPR2 with an endothelial targeted vector in hypoxic rats produced better results (Reynolds et al., 2007). In humans, BMPR2 mutations are thought to result in direct inactivation of the receptor or impaired trafficking of the receptor to the cell surface. Rescue strategies using viral vectors or chemical chaperones to overcome these aberrations are currently being investigated (Sobolewski et al., 2008). Moreover, as pulmonary artery smooth muscle cells from familial PAH patients demonstrate increased sensitivity to TGFβ signalling, molecules aimed at blocking this pro-proliferative transduction system may be of therapeutic utility (Morrell et al., 2001).

Peroxisome proliferator activated receptors

Recent observations suggest peroxisome proliferator activated receptors (PPARs) as another potential therapeutic target in PH. PPARγ is a downstream target of BMPR signalling and mediates the inhibitory effect of BMP on PGDF-induced smooth muscle cell proliferation (Hansmann and Zamanian, 2009) and PPARγ null mice develop PAH (Guignabert et al., 2009). Moreover, PPARγ agonists have direct anti-inflammatory, anti-proliferative and pro-apoptotic effects (Hansmann et al., 2007; 2008;). Rosiglitazone, a PPARγ agonist, is effective in reducing the PH produced in ApoE−/− mice and reduces right ventricular hypertrophy and vascular remodelling in hypoxia-induced PH (Crossno et al., 2007; Nisbet et al., 2007). PPARs may also underpin some of the beneficial effects of prostacyclin analogues in PH (Ali et al., 2006; Falcetti et al., 2010; Harrington et al., 2010).

5-HT signalling blockers

The 5-HT (serotonin) is a potent pulmonary vasoconstrictor. It was first implicated in the pathogenesis of PAH after outbreaks of the disease in patients using the anorexigenic drugs, aminorex and dexfenfluramine, appetite suppressants that inhibit 5-HT uptake (Dempsie et al., 2008). 5-HT is synthesized in pulmonary artery endothelial cells by the enzyme tryptophan hydroxylase 1 (TPH1) and then then acts at one of several 5-HT receptor subtypes (primarily 5-HT1B, 5-HT2A and 5-HT2B) and through the 5-HT transporter (SERT), to mediate constriction and proliferation of pulmonary artery smooth muscle cells and fibroblasts (Welsh et al., 2004). This results in a thickening of the medial layer and a narrowing of the lumen of the pulmonary artery and contributes to the pulmonary vascular remodelling associated with PAH. Downstream signalling molecules which play a role in 5-HT-signalling include ROCK, p38 and ERK1/2. Plasma 5-HT levels are elevated in PAH, as are SERT, 5-HT1B receptor and TPH1 expression in pulmonary artery smooth muscle and endothelial cells from PAH patients (MacLean and Dempsie, 2009). In addition, endothelial cells from PAH patients generate more 5-HT and proliferate more in response to 5-HT than control cells (Eddahibi et al., 2001). Experimentally, the inhibition of SERT prevents 5-HT-dependent proliferation in cells, and reduces hypoxic PH in rodent models (Guignabert et al., 2005; Song et al., 2005; Zhai et al., 2009; Zhu et al., 2009). There is also evidence to suggest that 5-HT may interact with BMPR2 to provide a ‘second hit’ risk factor for PAH (Long et al., 2006; Willers et al., 2006). A single nucleotide polymorphism in the SERT gene has been identified in PAH patients that appears to associate with higher SERT expression and higher mean PAP, though this link has not been corroborated in subsequent studies (Eddahibi et al., 2003; Machado et al., 2006; Willers et al., 2006; Roberts et al., 2009). At present, a number of drugs modifying 5-HT signalling are under clinical evaluation for the treatment of PH, including terguride (5-HT2A and 5-HT2B receptor antagonist), PRX-08066 (selective 5-HT2B receptor antagonist) and escilatorpram (selective 5-HT re-uptake inhibitor, SSRI).

Renin-angiotensin-aldosterone axis

The renin-angiotensin-aldosterone system (RAAS) is up-regulated in PAH (Cargill and Lipworth, 1995) and steps in the RAAS system cascade appear to be viable therapeutic targets in PH. Indeed, the ACE inhibitor captopril was evaluated almost 20 years ago in PAH patients with some success (Alpert et al., 1992). However, the development of more selective PAH therapies (i.e. specific to the pulmonary vasculature) has diverted attention from the RAAS as a viable target. Recently, the discovery that angiotensin-converting enzyme 2 (ACE2), a member of the vasoprotective arm of the RAAS, is up-regulated in both experimental models of PH and human PAH has refocused attention on this system (Ferreira et al., 2009). ACE2 plays a regulatory role in the lung and activation of endogenous ACE2 shifts the balance from the vasoconstrictor, proliferative path (ACE/Angiotensin II/AT1 receptor) to the vasoprotective anti-mitogenic path (ACE2/Angiotensin1-7/Mas) of the RAAS. Over-expression of ACE2 (by lentiviral gene delivery) or an ACE2 activator, XNT, reverses experimental PH (Ferreira et al., 2009; Shenoy et al., 2010).

Statins

Statins offer a novel approach to the treatment of PAH. This class of drugs have long been known to suppress vascular inflammation and vascular smooth muscle cell proliferation through a variety of mechanisms. In addition to lowering cholesterol via inhibition of 3-hydroxyl-3-methyl glutaryl CoA reductase, statins have been shown to have anti-proliferative, anti-thrombotic, anti-inflammatory and antioxidant effects, some of which may be secondary to cholesterol lowering. Statins, in particular simvastatin, have been reported to attenuate the development of PH in a number of experimental animal models (Nishimura et al., 2002; Girgis et al., 2003). Very recently, results from a double-blind, randomized, placebo-controlled study of the effects of simvastatin added to optimized conventional care produced a small and transient early reduction in right ventricular mass and NT-proBNP levels in patients with PAH, but this was not sustained over 12 months (Wilkins et al., 2010).

Ion channels

The haemodynamic dysfunction in PH patients also stems from abnormalities in the activity of ion channels that physiologically regulate local blood flow in the pulmonary circulation. For example, down-regulation of voltage-gated potassium channels, principally Kv1.5, appears to be a common feature of animal models of PH and in humans with the disease (Yuan et al., 1998; Pozeg et al., 2003). Targeting this potassium channel in PH is attractive because the facilitation of K+ flux through this pore causes hyperpolarization, vasodilatation, and is also thought to promote apoptosis. The expression of Kv1.5 is inversely related to pulmonary vessel size, suggesting that therapy would concentrate on the small pulmonary arteries and thereby exert the greatest effect on pulmonary vascular resistance (Pozeg et al., 2003). More recently, interest has arisen in the transient receptor potential (TRP) channel family. Experimental evidence suggests that TRPC6 expression and activity is up-regulated in PH and this leads to excessive Ca2+ entry into (pulmonary) vascular smooth muscle cells and vasoconstriction (Yu et al., 2004), in addition to PDGF-mediated proliferation (Schermuly et al., 2005). Moreover, an SNP in the TRPC6 promoter appears to associate with PH (Yu et al., 2009). Indeed, reversal of TRPC6 up-regulation may represent an added benefit of sildenafil therapy in PH (Lu et al., 2010). Finally, KATP channel activators such as iptakalim may have therapeutic utility in PH by producing pulmonary vasodilatation and preventing hypoxia- and ET-1-mediated pulmonary vascular smooth muscle cell proliferation (Xie et al., 2004; Zhu et al., 2008).

Cardiac-targeted therapy: β-adrenoceptor blockade

The major cause of death in PAH patients remains right ventricular failure, and perhaps one of the most-overlooked approaches in the treatment of the disease is cardiac-targeted therapies. Such strategies may have little or no direct effects on the pulmonary vasculature but prevent or reverse right heart dysfunction; it is reasonable to predict that such a tactic might make a major contribution to survival (Voelkel et al., 2006).

The antagonism of β-adrenoceptors is a commonly used strategy in patients with left-sided systolic heart failure, in which mortality is reduced by approximately 30%, but is not used clinically in right heart failure (i.e. PAH). The α112-adrenoceptor blocker carvedilol and the selective β1-adrenoceptor blockers, bisoprolol and metoprolol, reduce mortality in patients with left-sided systolic heart failure with a reversal of maladaptive cardiac remodelling, improved cardiac function and prevention of arrhythmias (Bristow et al., 1996; Fowler et al., 2007; MacGregor et al., 2009). β-Adrenoceptor tachyphylaxis has also been demonstrated in PAH and may contribute to maladaptive right ventricular remodelling and the development of arrhythmias (Velez-Roa et al., 2004). Carvedilol and metoprolol have been shown to reverse right ventricular remodelling and improve right ventricular function in experimental PH (Bogaard et al., 2010), and the β-blocker arotinolol decreases both PAP and right ventricular hypertrophy, without altering systemic blood pressure, in a rat model of monocrotaline-induced PAH (Ishikawa et al., 2009). Use of β-blockers in PAH has possible detrimental effects on haemodynamics and exercise capacity. While no specific clinical trial has been conducted to evaluate the efficacy and safety of β-blockers in PAH, a small cohort of porto-pulmonary hypertension patients were found to experience significant functional improvement following cessation of β-blocker therapy (Provencher et al., 2006), suggesting a detrimental rather than beneficial outcome. Nonetheless, further investigation of this class of anti-hypertensive medicines may bring forth promising results in PAH patients.

Combination therapies

Since PH has a complex, multi-factorial aetiology, and the fact that current treatments (and the vast majority of the emerging therapies described previously) only target one aspect of the disease, modern approaches have focused on combining existing and newer therapies to bring about a significant improvement in outcome. This is a logical approach (based on the need for a combinatorial approach to adequately control systemic hypertension) and many studies suggest additive, if not synergistic, effects of combination therapy in PH (Schermuly et al., 2001; Baliga et al., 2008). Indeed, in clinical practice, combination therapy has become the default position even though trial evidence to support this strategy is limited. Small scale clinical evaluation of combinations of prostanoids, ERAs and PDE5 inhibitors have been tried with some success (Ghofrani et al., 2002; Stiebellehner et al., 2003; Stocker et al., 2003; Hoeper et al., 2004; Humbert et al., 2004), with additional studies currently recruiting [e.g. COMPASS-2 (sildenafil plus bosentan), STEP (iloprost plus bosentan)]; however, validation of these combination therapies will require further larger scale trials. Moreover, these dual approaches have, to date, been restricted to combinations of existing therapies which are largely centred on the haemodynamic dysfunction. Newer therapies, targeting cell proliferation rather than vasodilatation, will necessarily entail novel combinations (as future trials will be on a background of existing treatment).

Combination therapy, however, has important implications for the cost of treating PH patients, which at present is approximately £45 000 per annum in the UK (National Institute for Health and Clinical Excellence). The partnership between academia, the pharmaceutical industry and healthcare providers has been successful in developing treatments for PH, but these drug costs pose a real challenge to healthcare systems. Exploring the potential of drug combination in PH that include generic medicines, such as simvastatin and racecadotril, has real potential for affordable drug development.

Conclusions

Advances in the treatment of PH over the past decade have enabled physicians to substantially improve the prognosis, yet the mortality rate remains high. Existing treatments are based predominantly on vasodilatation, whereas many emerging therapies are aimed at cell proliferation and re-modelling (Figure 2). There is great optimism that this alternative strategy will yield superior results, either alone or in combination.

Acknowledgments

The authors are supported by the British Heart Foundation, The Wellcome Trust and The Medical Research Council.

Glossary

Abbreviations

AC

adenylate cyclase

ACE

angiotensin converting enzyme

BH4

tetrahydrobiopterin

BMPR

bone morphogenetic protein receptor

CCB

calcium channel blockers

CTEPH

chronic thromboembolic PH

eNOS

endothelial NO synthase

EPC

endothelial progenitor cell

ERA

endothelin receptor antagonist

ERK1/2

extra-cellular signal regulated kinase

ET-1

endothelin-1

FGF

fibroblast growth factor

IP

prostacyclin receptor

JAK/STAT

Janus kinase/signal transducer and activator of transcription

Kv

voltage-sensitive potassium channel

MAPK

mitogen-activated protein kinase

NEP

neutral endopeptidase

NP

natriuretic peptide

NPR

natriuretic peptide receptor

PAH

pulmonary arterial hypertension

PDGF

platelet derived growth factor

pGC

particulate guanylate cyclase

PGI2

prostacyclin

PH

pulmonary hypertension

PI3K

phosphoinositide-3-kinase

PKC

protein kinase C

PPAR

peroxisome proliferator activated receptor

PPHN

persistent pulmonary hypertension of the newborn

PVR

pulmonary vascular resistance

ROCK

Rho-associated kinase

SERT

5-HT transporter

sGC

soluble guanylate cyclase

SNP

single nucleotide polymorphism

Src

Src kinase

SSRI

selective 5-HT reuptake inhibitor

TGFβ

transforming growth factor β

TRK

tyrosine kinase receptor

TRPC6

transient receptor potential C6

TXA2

thromboxane A2

VEGF

vascular endothelial growth factor

VOCC

voltage operated calcium channel

XNT

1-[(2-dimethylamino) ethylamino]-4-(hydroxymethyl)-7-[(4-methylphenyl) sulphonyl oxy]-9H-xanthene-9-one)

Conflicts of interest

None.

Supporting Information

Teaching Materials; Figs 1–2 as PowerPoint slide.

bph0163-0125-SD1.pptx (366.5KB, pptx)

References

  1. Ahluwalia A, Hobbs AJ. Endothelium-derived C-type natriuretic peptide: more than just a hyperpolarizing factor. Trends Pharmacol Sci. 2005;26:162–167. doi: 10.1016/j.tips.2005.01.005. [DOI] [PubMed] [Google Scholar]
  2. Ahluwalia A, MacAllister RJ, Hobbs AJ. Vascular actions of natriuretic peptides. Cyclic GMP-dependent and -independent mechanisms. Basic Res Cardiol. 2004;99:83–89. doi: 10.1007/s00395-004-0459-6. [DOI] [PubMed] [Google Scholar]
  3. Alexander SPH, Mathie A, Peters JA. Guide to receptors and channels (GRAC) Br J Pharmacol. (4th edn.) 2009;158(Suppl. 1):S1–S254. doi: 10.1111/j.1476-5381.2011.01649_1.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ali FY, Egan K, FitzGerald GA, Desvergne B, Wahli W, Bishop-Bailey D, et al. Role of prostacyclin versus peroxisome proliferator-activated receptor beta receptors in prostacyclin sensing by lung fibroblasts. Am J Respir Cell Mol Biol. 2006;34:242–246. doi: 10.1165/rcmb.2005-0289OC. [DOI] [PubMed] [Google Scholar]
  5. Alpert MA, Pressly TA, Mukerji V, Lambert CR, Mukerji B. Short- and long-term hemodynamic effects of captopril in patients with pulmonary hypertension and selected connective tissue disease. Chest. 1992;102:1407–1412. doi: 10.1378/chest.102.5.1407. [DOI] [PubMed] [Google Scholar]
  6. Amsallem E, Kasparian C, Haddour G, Boissel JP, Nony P. Phosphodiesterase III inhibitors for heart failure. Cochrane Database Syst Rev. 2005;(1):CD002230. doi: 10.1002/14651858.CD002230.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Archer SL, Djaballah K, Humbert M, Weir KE, Fartoukh M, I'ava-Santucci J, et al. Nitric oxide deficiency in fenfluramine- and dexfenfluramine-induced pulmonary hypertension. Am J Respir Crit Care Med. 1998;158:1061–1067. doi: 10.1164/ajrccm.158.4.9802113. [DOI] [PubMed] [Google Scholar]
  8. Atkinson C, Stewart S, Upton PD, Machado R, Thomson JR, Trembath RC, et al. Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor. Circulation. 2002;105:1672–1678. doi: 10.1161/01.cir.0000012754.72951.3d. [DOI] [PubMed] [Google Scholar]
  9. Badesch DB, McGoon MD, Barst RJ, Tapson VF, Rubin LJ, Wigley FM, et al. Longterm survival among patients with scleroderma-associated pulmonary arterial hypertension treated with intravenous epoprostenol. J Rheumatol. 2009;36:2244–2249. doi: 10.3899/jrheum.081277. [DOI] [PubMed] [Google Scholar]
  10. Baliga RS, Zhao L, Madhani M, Lopez-Torondel B, Visintin C, Selwood D, et al. Synergy between natriuretic peptides and phosphodiesterase 5 inhibitors ameliorates pulmonary arterial hypertension. Am J Respir Crit Care Med. 2008;178:861–869. doi: 10.1164/rccm.200801-121OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Barst RJ, Rubin LJ, Long WA, McGoon MD, Rich S, Badesch DB, et al. A comparison of continuous intravenous epoprostenol (prostacyclin) with conventional therapy for primary pulmonary hypertension. The Primary Pulmonary Hypertension Study Group. N Engl J Med. 1996;334:296–302. doi: 10.1056/NEJM199602013340504. [DOI] [PubMed] [Google Scholar]
  12. Barst RJ, Langleben D, Frost A, Horn EM, Oudiz R, Shapiro S, et al. Sitaxsentan therapy for pulmonary arterial hypertension. Am J Respir Crit Care Med. 2004;169:441–447. doi: 10.1164/rccm.200307-957OC. [DOI] [PubMed] [Google Scholar]
  13. Belik J. Riociguat, an oral soluble guanylate cyclase stimulator for the treatment of pulmonary hypertension. Curr Opin Investig Drugs. 2009;10:971–979. [PubMed] [Google Scholar]
  14. Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev. 2006;58:488–520. doi: 10.1124/pr.58.3.5. [DOI] [PubMed] [Google Scholar]
  15. Black SM, Sanchez LS, Mata-Greenwood E, Bekker JM, Steinhorn RH, Fineman JR. sGC and PDE5 are elevated in lambs with increased pulmonary blood flow and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2001;281:L1051–L1057. doi: 10.1152/ajplung.2001.281.5.L1051. [DOI] [PubMed] [Google Scholar]
  16. Bogaard HJ, Natarajan R, Mizuno S, Abbate A, Chang PJ, Chau VQ, et al. Adrenergic receptor blockade reverses right heart remodeling and dysfunction in pulmonary hypertensive rats. Am J Respir Crit Care Med. 2010;182:652–660. doi: 10.1164/rccm.201003-0335OC. [DOI] [PubMed] [Google Scholar]
  17. Bristow MR, Gilbert EM, Abraham WT, Adams KF, Fowler MB, Hershberger RE, et al. Carvedilol produces dose-related improvements in left ventricular function and survival in subjects with chronic heart failure. MOCHA Investigators. Circulation. 1996;94:2807–2816. doi: 10.1161/01.cir.94.11.2807. [DOI] [PubMed] [Google Scholar]
  18. Budhiraja R, Tuder RM, Hassoun PM. Endothelial dysfunction in pulmonary hypertension. Circulation. 2004;109:159–165. doi: 10.1161/01.CIR.0000102381.57477.50. [DOI] [PubMed] [Google Scholar]
  19. Cargill RI, Lipworth BJ. The role of the renin-angiotensin and natriuretic peptide systems in the pulmonary vasculature. Br J Clin Pharmacol. 1995;40:11–18. doi: 10.1111/j.1365-2125.1995.tb04528.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Channick RN, Simonneau G, Sitbon O, Robbins IM, Frost A, Tapson VF, et al. Effects of the dual endothelin-receptor antagonist bosentan in patients with pulmonary hypertension: a randomised placebo-controlled study. Lancet. 2001;358:1119–1123. doi: 10.1016/S0140-6736(01)06250-X. [DOI] [PubMed] [Google Scholar]
  21. Chen YF, Feng JA, Li P, Xing D, Ambalavanan N, Oparil S. Atrial natriuretic peptide-dependent modulation of hypoxia-induced pulmonary vascular remodeling. Life Sci. 2006;79:1357–1365. doi: 10.1016/j.lfs.2006.03.051. [DOI] [PubMed] [Google Scholar]
  22. Chester M, Tourneux P, Seedorf G, Grover TR, Gien J, Abman SH. Cinaciguat, a soluble guanylate cyclase activator, causes potent and sustained pulmonary vasodilation in the ovine fetus. Am J Physiol Lung Cell Mol Physiol. 2009;297:L318–L325. doi: 10.1152/ajplung.00062.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chhina MK, Nargues W, Grant GM, Nathan SD. Evaluation of imatinib mesylate in the treatment of pulmonary arterial hypertension. Future Cardiol. 2010;6:19–35. doi: 10.2217/fca.09.54. [DOI] [PubMed] [Google Scholar]
  24. Christman BW, McPherson CD, Newman JH, King GA, Bernard GR, Groves BM, et al. 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. [DOI] [PubMed] [Google Scholar]
  25. Cool CD, Stewart JS, Werahera P, Miller GJ, Williams RL, Voelkel NF, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Crawley DE, Zhao L, Giembycz MA, Liu S, Barnes PJ, Winter RJ, et al. Chronic hypoxia impairs soluble guanylyl cyclase-mediated pulmonary arterial relaxation in the rat. Am J Physiol. 1992;263:L325–L332. doi: 10.1152/ajplung.1992.263.3.L325. [DOI] [PubMed] [Google Scholar]
  27. Creagh-Brown BC, Griffiths MJ, Evans TW. Bench-to-bedside review: Inhaled nitric oxide therapy in adults. Crit Care. 2009;13:221. doi: 10.1186/cc7734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Crossno JT, Jr, Garat CV, Reusch JE, Morris KG, Dempsey EC, McMurtry IF, et al. Rosiglitazone attenuates hypoxia-induced pulmonary arterial remodeling. Am J Physiol Lung Cell Mol Physiol. 2007;292:L885–L897. doi: 10.1152/ajplung.00258.2006. [DOI] [PubMed] [Google Scholar]
  29. Dempsie Y, Morecroft I, Welsh DJ, MacRitchie NA, Herold N, Loughlin L, et al. Converging evidence in support of the 5-HT hypothesis of dexfenfluramine-induced pulmonary hypertension with novel transgenic mice. Circulation. 2008;117:2928–2937. doi: 10.1161/CIRCULATIONAHA.108.767558. [DOI] [PubMed] [Google Scholar]
  30. Denton CP, Pope JE, Peter HH, Gabrielli A, Boonstra A, van den Hoogen FH, et al. Long-term effects of bosentan on quality of life, survival, safety and tolerability in pulmonary arterial hypertension related to connective tissue diseases. Ann Rheum Dis. 2008;67:1222–1228. doi: 10.1136/ard.2007.079921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Do e Z. Fukumoto Y, Takaki A, Tawara S, Ohashi J, Nakano M, et al. Evidence for Rho-kinase activation in patients with pulmonary arterial hypertension. Circ J. 2009;73:1731–1739. doi: 10.1253/circj.cj-09-0135. [DOI] [PubMed] [Google Scholar]
  32. Dony E, Lai YJ, Dumitrascu R, Pullamsetti SS, Savai R, Ghofrani HA, et al. Partial reversal of experimental pulmonary hypertension by phosphodiesterase-3/4 inhibition. Eur Respir J. 2008;31:599–610. doi: 10.1183/09031936.00002007. [DOI] [PubMed] [Google Scholar]
  33. Dumitrascu R, Weissmann N, Ghofrani HA, Dony E, Beuerlein K, Schmidt H, et al. Activation of soluble guanylate cyclase reverses experimental pulmonary hypertension and vascular remodeling. Circulation. 2006;113:286–295. doi: 10.1161/CIRCULATIONAHA.105.581405. [DOI] [PubMed] [Google Scholar]
  34. Eddahibi S, Humbert M, Fadel E, Raffestin B, Darmon M, Capron F, et al. 5-HT 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] [PMC free article] [PubMed] [Google Scholar]
  35. Eddahibi S, Chaouat A, Morrell N, Fadel E, Fuhrman C, Bugnet AS, et al. Polymorphism of the 5-HT transporter gene and pulmonary hypertension in chronic obstructive pulmonary disease. Circulation. 2003;108:1839–1844. doi: 10.1161/01.CIR.0000091409.53101.E8. [DOI] [PubMed] [Google Scholar]
  36. Evgenov OV, Busch CJ, Evgenov NV, Liu R, Petersen B, Falkowski GE, et al. Inhibition of phosphodiesterase 1 augments the pulmonary vasodilator response to inhaled nitric oxide in awake lambs with acute pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2006;290:L723–L729. doi: 10.1152/ajplung.00485.2004. [DOI] [PubMed] [Google Scholar]
  37. Evgenov OV, Kohane DS, Bloch KD, Stasch JP, Volpato GP, Bellas E, et al. Inhaled agonists of soluble guanylate cyclase induce selective pulmonary vasodilation. Am J Respir Crit Care Med. 2007;176:1138–1145. doi: 10.1164/rccm.200707-1121OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Fadini GP, Avogaro A, Ferraccioli G, Agostini C. Endothelial progenitors in pulmonary hypertension: new pathophysiology and therapeutic implications. Eur Respir J. 2010;35:418–425. doi: 10.1183/09031936.00112809. [DOI] [PubMed] [Google Scholar]
  39. Fagan KA, Fouty BW, Tyler RC, Morris KG, Jr, Hepler LK, Sato K, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Falcetti E, Hall SM, Phillips PG, Patel J, Morrell NW, Haworth SG, et al. Smooth muscle proliferation and role of the prostacyclin (IP) receptor in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2010;182:1161–1170. doi: 10.1164/rccm.201001-0011OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Ferreira AJ, Shenoy V, Yamazato Y, Sriramula S, Francis J, Yuan L, et al. Evidence for angiotensin-converting enzyme 2 as a therapeutic target for the prevention of pulmonary hypertension. Am J Respir Crit Care Med. 2009;179:1048–1054. doi: 10.1164/rccm.200811-1678OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Fowler MB, Lottes SR, Nelson JJ, Lukas MA, Gilbert EM, Greenberg B, et al. Beta-blocker dosing in community-based treatment of heart failure. Am Heart J. 2007;153:1029–1036. doi: 10.1016/j.ahj.2007.03.010. [DOI] [PubMed] [Google Scholar]
  43. Frantz S, Adamek A, Fraccarollo D, Tillmanns J, Widder JD, Dienesch C, et al. The eNOS enhancer AVE 9488: a novel cardioprotectant against ischemia reperfusion injury. Basic Res Cardiol. 2009;104:773–779. doi: 10.1007/s00395-009-0041-3. [DOI] [PubMed] [Google Scholar]
  44. de Frutos S, Nitta CH, Caldwell E, Friedman J, Gonzalez Bosc LV. Regulation of soluble guanylyl cyclase-alpha1 expression in chronic hypoxia-induced pulmonary hypertension: role of NFATc3 and HuR. Am J Physiol Lung Cell Mol Physiol. 2009;297:L475–L486. doi: 10.1152/ajplung.00060.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fujita H, Fukumoto Y, Saji K, Sugimura K, Demachi J, Nawata J, et al. Acute vasodilator effects of inhaled fasudil, a specific Rho-kinase inhibitor, in patients with pulmonary arterial hypertension. Heart Vessels. 2010;25:144–149. doi: 10.1007/s00380-009-1176-8. [DOI] [PubMed] [Google Scholar]
  46. Galie N, Badesch D, Oudiz R, Simonneau G, McGoon MD, Keogh AM, et al. Ambrisentan therapy for pulmonary arterial hypertension. J Am Coll Cardiol. 2005a;46:529–535. doi: 10.1016/j.jacc.2005.04.050. [DOI] [PubMed] [Google Scholar]
  47. Galie N, Ghofrani HA, Torbicki A, Barst RJ, Rubin LJ, Badesch D, et al. Sildenafil citrate therapy for pulmonary arterial hypertension. N Engl J Med. 2005b;353:2148–2157. doi: 10.1056/NEJMoa050010. [DOI] [PubMed] [Google Scholar]
  48. Galie N, Brundage BH, Ghofrani HA, Oudiz RJ, Simonneau G, Safdar Z, et al. Tadalafil therapy for pulmonary arterial hypertension. Circulation. 2009a;119:2894–2903. doi: 10.1161/CIRCULATIONAHA.108.839274. [DOI] [PubMed] [Google Scholar]
  49. Galie N, Manes A, Negro L, Palazzini M, Bacchi-Reggiani ML, Branzi A. A meta-analysis of randomized controlled trials in pulmonary arterial hypertension. Eur Heart J. 2009b;30:394–403. doi: 10.1093/eurheartj/ehp022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Ghofrani HA, Wiedemann R, Rose F, Olschewski H, Schermuly RT, Weissmann N, et al. Combination therapy with oral sildenafil and inhaled iloprost for severe pulmonary hypertension. Ann Intern Med. 2002;136:515–522. doi: 10.7326/0003-4819-136-7-200204020-00008. [DOI] [PubMed] [Google Scholar]
  51. Ghofrani HA, Seeger W, Grimminger F. Imatinib for the treatment of pulmonary arterial hypertension. N Engl J Med. 2005;353:1412–1413. doi: 10.1056/NEJMc051946. [DOI] [PubMed] [Google Scholar]
  52. Ghofrani HA, Hoeper MM, Halank M, Meyer FJ, Staehler G, Behr J, et al. Riociguat for chronic thromboembolic pulmonary hypertension and pulmonary arterial hypertension: a phase II study. Eur Respir J. 2010;36:792–799. doi: 10.1183/09031936.00182909. [DOI] [PubMed] [Google Scholar]
  53. 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. [DOI] [PubMed] [Google Scholar]
  54. Girgis RE, Li D, Zhan X, Garcia JG, Tuder RM, Hassoun PM, et al. Attenuation of chronic hypoxic pulmonary hypertension by simvastatin. Am J Physiol Heart Circ Physiol. 2003;285:H938–H945. doi: 10.1152/ajpheart.01097.2002. [DOI] [PubMed] [Google Scholar]
  55. Gomberg-Maitland M, Maitland ML, Barst RJ, Sugeng L, Coslet S, Perrino TJ, et al. A dosing/cross-development study of the multikinase inhibitor sorafenib in patients with pulmonary arterial hypertension. Clin Pharmacol Ther. 2010;87:303–310. doi: 10.1038/clpt.2009.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Goyal P, Kiran U, Chauhan S, Juneja R, Choudhary M. Efficacy of nitroglycerin inhalation in reducing pulmonary arterial hypertension in children with congenital heart disease. Br J Anaesth. 2006;97:208–214. doi: 10.1093/bja/ael112. [DOI] [PubMed] [Google Scholar]
  57. Grimminger F, Weimann G, Frey R, Voswinckel R, Thamm M, Bolkow D, et al. BAY 63-2521, an oral soluble guanylate cyclase stimulator, has a favourable safety profile, improves cardiopulmonary haemodynamics and has therapeutic potential in pulmonary hypertension. 2009.
  58. Guignabert C, Raffestin B, Benferhat R, Raoul W, Zadigue P, Rideau D, et al. 5-HT transporter inhibition prevents and reverses monocrotaline-induced pulmonary hypertension in rats. Circulation. 2005;111:2812–2819. doi: 10.1161/CIRCULATIONAHA.104.524926. [DOI] [PubMed] [Google Scholar]
  59. Guignabert C, Alvira CM, Alastalo TP, Sawada H, Hansmann G, Zhao M, et al. Tie2-mediated loss of peroxisome proliferator-activated receptor-gamma in mice causes PDGF receptor-beta-dependent pulmonary arterial muscularization. Am J Physiol Lung Cell Mol Physiol. 2009;297:L1082–L1090. doi: 10.1152/ajplung.00199.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Gunaydin S, Imai Y, Takanashi Y, Seo K, Hagino I, Chang D, et al. 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] [PubMed] [Google Scholar]
  61. Hampl V, Tristani-Firouzi M, Hutsell TC, Archer SL. Nebulized nitric oxide/nucleophile adduct reduces chronic pulmonary hypertension. Cardiovasc Res. 1996;31:55–62. [PubMed] [Google Scholar]
  62. Hansmann G, Zamanian RT. PPARgamma activation: a potential treatment for pulmonary hypertension. Sci Transl Med. 2009;1:12ps14. doi: 10.1126/scitranslmed.3000267. [DOI] [PubMed] [Google Scholar]
  63. Hansmann G, Wagner RA, Schellong S, Perez VA, Urashima T, Wang L, et al. 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] [PubMed] [Google Scholar]
  64. Hansmann G, de Jesus Perez VA, Alastalo TP, Alvira CM, Guignabert C, Bekker JM, et al. An antiproliferative BMP-2/PPARgamma/apoE axis in human and murine SMCs and its role in pulmonary hypertension. J Clin Invest. 2008;118:1846–1857. doi: 10.1172/JCI32503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Harrington LS, Moreno L, Reed A, Wort SJ, Desvergne B, Garland C, et al. The PPARbeta/delta agonist GW0742 relaxes pulmonary vessels and limits right heart hypertrophy in rats with hypoxia-induced pulmonary hypertension. PloS One. 2010;5:e9526. doi: 10.1371/journal.pone.0009526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hassoun PM. Pulmonary arterial hypertension complicating connective tissue diseases. Semin Respir Crit Care Med. 2009;30:429–439. doi: 10.1055/s-0029-1233312. [DOI] [PubMed] [Google Scholar]
  67. Hobbs AJ. Soluble guanylate cyclase: the forgotten sibling. Trends Pharmacol Sci. 1997;18:484–491. doi: 10.1016/s0165-6147(97)01137-1. [DOI] [PubMed] [Google Scholar]
  68. Hoeper MM, Faulenbach C, Golpon H, Winkler J, Welte T, Niedermeyer J. Combination therapy with bosentan and sildenafil in idiopathic pulmonary arterial hypertension. Eur Respir J. 2004;24:1007–1010. doi: 10.1183/09031936.04.00051104. [DOI] [PubMed] [Google Scholar]
  69. Humbert M, Monti G, Fartoukh M, Magnan A, Brenot F, Rain B, et al. Platelet-derived growth factor expression in primary pulmonary hypertension: comparison of HIV seropositive and HIV seronegative patients. Eur Respir J. 1998;11:554–559. [PubMed] [Google Scholar]
  70. Humbert M, Barst RJ, Robbins IM, Channick RN, Galie N, Boonstra A, et al. Combination of bosentan with epoprostenol in pulmonary arterial hypertension: BREATHE-2. Eur Respir J. 2004;24:353–359. doi: 10.1183/09031936.04.00028404. [DOI] [PubMed] [Google Scholar]
  71. Ichinose F, Roberts JD, Jr, Zapol WM. Inhaled nitric oxide: a selective pulmonary vasodilator: current uses and therapeutic potential. Circulation. 2004;109:3106–3111. doi: 10.1161/01.CIR.0000134595.80170.62. [DOI] [PubMed] [Google Scholar]
  72. Ishikawa M, Sato N, Asai K, Takano T, Mizuno K. Effects of a pure alpha/beta-adrenergic receptor blocker on monocrotaline-induced pulmonary arterial hypertension with right ventricular hypertrophy in rats. Circ J. 2009;73:2337–2341. doi: 10.1253/circj.cj-09-0213. [DOI] [PubMed] [Google Scholar]
  73. Ishikura K, Yamada N, Ito M, Ota S, Nakamura M, Isaka N, et al. 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] [PubMed] [Google Scholar]
  74. Izikki M, Guignabert C, Fadel E, Humbert M, Tu L, Zadigue P, et al. Endothelial-derived FGF2 contributes to the progression of pulmonary hypertension in humans and rodents. J Clin Invest. 2009;119:512–523. doi: 10.1172/JCI35070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Jackson G, Benjamin N, Jackson N, Allen MJ. Effects of sildenafil citrate on human hemodynamics. Am J Cardiol. 1999;83:13C–20C. doi: 10.1016/s0002-9149(99)00043-0. [DOI] [PubMed] [Google Scholar]
  76. Jin H, Yang RH, Chen YF, Jackson RM, Oparil S. Atrial natriuretic peptide attenuates the development of pulmonary hypertension in rats adapted to chronic hypoxia. J Clin Invest. 1990;85:115–120. doi: 10.1172/JCI114400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Jin H, Yang RH, Oparil S. Cicletanine blunts the pulmonary pressor response to acute hypoxia in rats. Am J Med Sci. 1992;304:14–19. doi: 10.1097/00000441-199207000-00005. [DOI] [PubMed] [Google Scholar]
  78. Johnson SR, Mehta S, Granton JT. Anticoagulation in pulmonary arterial hypertension: a qualitative systematic review. Eur Respir J. 2006;28:999–1004. doi: 10.1183/09031936.06.00015206. [DOI] [PubMed] [Google Scholar]
  79. Kerkela R, Grazette L, Yacobi R, Iliescu C, Patten R, Beahm C, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med. 2006;12:908–916. doi: 10.1038/nm1446. [DOI] [PubMed] [Google Scholar]
  80. Khoo JP, Zhao L, Alp NJ, Bendall JK, Nicoli T, Rockett K, et al. Pivotal role for endothelial tetrahydrobiopterin in pulmonary hypertension. Circulation. 2005;111:2126–2133. doi: 10.1161/01.CIR.0000162470.26840.89. [DOI] [PubMed] [Google Scholar]
  81. Klein M, Schermuly RT, Ellinghaus P, Milting H, Riedl B, Nikolova S, et al. Combined tyrosine and serine/threonine kinase inhibition by sorafenib prevents progression of experimental pulmonary hypertension and myocardial remodeling. Circulation. 2008;118:2081–2090. doi: 10.1161/CIRCULATIONAHA.108.779751. [DOI] [PubMed] [Google Scholar]
  82. Klinger JR, Petit RD, Warburton RR, Wrenn DS, Arnal F, Hill NS. Neutral endopeptidase inhibition attenuates development of hypoxic pulmonary hypertension in rats. J Appl Physiol. 1993;75:1615–1623. doi: 10.1152/jappl.1993.75.4.1615. [DOI] [PubMed] [Google Scholar]
  83. Klinger JR, Warburton RR, Pietras L, Hill NS. Brain natriuretic peptide inhibits hypoxic pulmonary hypertension in rats. J Appl Physiol. 1998;84:1646–1652. doi: 10.1152/jappl.1998.84.5.1646. [DOI] [PubMed] [Google Scholar]
  84. Klinger JR, Warburton RR, Pietras LA, Smithies O, Swift R, Hill NS. Genetic disruption of atrial natriuretic peptide causes pulmonary hypertension in normoxic and hypoxic mice. Am J Physiol. 1999;276:L868–L874. doi: 10.1152/ajplung.1999.276.5.L868. [DOI] [PubMed] [Google Scholar]
  85. Klinger JR, Thaker S, Houtchens J, Preston IR, Hill NS, Farber HW. Pulmonary hemodynamic responses to brain natriuretic peptide and sildenafil in patients with pulmonary arterial hypertension. Chest. 2006;129:417–425. doi: 10.1378/chest.129.2.417. [DOI] [PubMed] [Google Scholar]
  86. Kuhn M. Molecular physiology of natriuretic peptide signalling. Basic Res Cardiol. 2004;99:76–82. doi: 10.1007/s00395-004-0460-0. [DOI] [PubMed] [Google Scholar]
  87. Landmesser U, Dikalov S, Price SR, McCann L, Fukai T, Holland SM, et al. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest. 2003;111:1201–1209. doi: 10.1172/JCI14172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Lane KB, Machado RD, Pauciulo MW, Thomson JR, Phillips JA, III, Loyd JE, et al. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat Genet. 2000;26:81–84. doi: 10.1038/79226. [DOI] [PubMed] [Google Scholar]
  89. Lapp H, Mitrovic V, Franz N, Heuer H, Buerke M, Wolfertz J, et al. Cinaciguat (BAY 58-2667) improves cardiopulmonary hemodynamics in patients with acute decompensated heart failure. Circulation. 2009;119:2781–2788. doi: 10.1161/CIRCULATIONAHA.108.800292. [DOI] [PubMed] [Google Scholar]
  90. Le Cras TD, Tyler RC, Horan MP, Morris KG, Tuder RM, McMurtry IF, et al. Effects of chronic hypoxia and altered hemodynamics on endothelial nitric oxide synthase expression in the adult rat lung. J Clin Invest. 1998;101:795–801. doi: 10.1172/JCI786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Leiper J, Nandi M, Torondel B, Murray-Rust J, Malaki M, O'Hara B, et al. Disruption of methylarginine metabolism impairs vascular homeostasis. Nat Med. 2007;13:198–203. doi: 10.1038/nm1543. [DOI] [PubMed] [Google Scholar]
  92. Li P, Oparil S, Novak L, Cao X, Shi W, Lucas J, et al. ANP signaling inhibits TGF-beta-induced Smad2 and Smad3 nuclear translocation and extracellular matrix expression in rat pulmonary arterial smooth muscle cells. J Appl Physiol. 2007;102:390–398. doi: 10.1152/japplphysiol.00468.2006. [DOI] [PubMed] [Google Scholar]
  93. Long L, MacLean MR, Jeffery TK, Morecroft I, Yang X, Rudarakanchana N, et al. 5-HT increases susceptibility to pulmonary hypertension in BMPR2-deficient mice. Circ Res. 2006;98:818–827. doi: 10.1161/01.RES.0000215809.47923.fd. [DOI] [PubMed] [Google Scholar]
  94. Lu W, Ran P, Zhang D, Peng G, Li B, Zhong N, et al. Sildenafil inhibits chronically hypoxic upregulation of canonical transient receptor potential expression in rat pulmonary arterial smooth muscle. Am J Physiol Cell Physiol. 2010;298:C114–C123. doi: 10.1152/ajpcell.00629.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. MacGregor JF, Wachter SB, Munger M, Stoddard G, Bristow MR, Gilbert EM. Carvedilol produces sustained long-term benefits: follow-up at 12 years. Congest Heart Fail. 2009;15:5–8. doi: 10.1111/j.1751-7133.2008.00038.x. [DOI] [PubMed] [Google Scholar]
  96. McLaughlin VV, Archer SL, Badesch DB, Barst RJ, Farber HW, Lindner JR, et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association developed in collaboration with the American College of Chest Physicians; American Thoracic Society, Inc.; and the Pulmonary Hypertension Association. J Am Coll Cardiol. 2009a;53:1573–1619. doi: 10.1016/j.jacc.2009.01.004. [DOI] [PubMed] [Google Scholar]
  97. McLaughlin VV, Archer SL, Badesch DB, Barst RJ, Farber HW, Lindner JR, et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension: a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association: developed in collaboration with the American College of Chest Physicians, American Thoracic Society, Inc., and the Pulmonary Hypertension Association. Circulation. 2009b;119:2250–2294. doi: 10.1161/CIRCULATIONAHA.109.192230. [DOI] [PubMed] [Google Scholar]
  98. MacLean MR, Dempsie Y. 5-HT and pulmonary hypertension – from bench to bedside? Curr Opin Pharmacol. 2009;9:281–286. doi: 10.1016/j.coph.2009.02.005. [DOI] [PubMed] [Google Scholar]
  99. McMurtry MS, Archer SL, Altieri DC, Bonnet S, Haromy A, Harry G, et al. 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] [PMC free article] [PubMed] [Google Scholar]
  100. McMurtry MS, Moudgil R, Hashimoto K, Bonnet S, Michelakis ED, Archer SL. Overexpression of human bone morphogenetic protein receptor 2 does not ameliorate monocrotaline pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 2007;292:L872–L878. doi: 10.1152/ajplung.00309.2006. [DOI] [PubMed] [Google Scholar]
  101. Machado RD, Koehler R, Glissmeyer E, Veal C, Suntharalingam J, Kim M, et al. Genetic association of the 5-HT transporter in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;173:793–797. doi: 10.1164/rccm.200509-1365OC. [DOI] [PubMed] [Google Scholar]
  102. Macrae DJ, Field D, Mercier JC, Moller J, Stiris T, Biban P, et al. Inhaled nitric oxide therapy in neonates and children: reaching a European consensus. Intensive Care Med. 2004;30:372–380. doi: 10.1007/s00134-003-2122-3. [DOI] [PubMed] [Google Scholar]
  103. Madhani M, Okorie M, Hobbs AJ, MacAllister RJ. Reciprocal regulation of human soluble and particulate guanylate cyclases in vivo. Br J Pharmacol. 2006;149:797–801. doi: 10.1038/sj.bjp.0706920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Mandegar M, Fung YC, Huang W, Remillard CV, Rubin LJ, Yuan JX. Cellular and molecular mechanisms of pulmonary vascular remodeling: role in the development of pulmonary hypertension. Microvasc Res. 2004;68:75–103. doi: 10.1016/j.mvr.2004.06.001. [DOI] [PubMed] [Google Scholar]
  105. Matsui H, Shimosawa T, Itakura K, Guanqun X, Ando K, Fujita T. Adrenomedullin can protect against pulmonary vascular remodeling induced by hypoxia. Circulation. 2004;109:2246–2251. doi: 10.1161/01.CIR.0000127950.13380.FD. [DOI] [PubMed] [Google Scholar]
  106. Merklinger SL, Jones PL, Martinez EC, Rabinovitch M. Epidermal growth factor receptor blockade mediates smooth muscle cell apoptosis and improves survival in rats with pulmonary hypertension. Circulation. 2005;112:423–431. doi: 10.1161/CIRCULATIONAHA.105.540542. [DOI] [PubMed] [Google Scholar]
  107. Morrell NW, Yang X, Upton PD, Jourdan KB, Morgan N, Sheares KK, et al. 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] [PubMed] [Google Scholar]
  108. Morty RE, Nejman B, Kwapiszewska G, Hecker M, Zakrzewicz A, Kouri FM, et al. Dysregulated bone morphogenetic protein signaling in monocrotaline-induced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol. 2007;27:1072–1078. doi: 10.1161/ATVBAHA.107.141200. [DOI] [PubMed] [Google Scholar]
  109. Mouchaers KT, Schalij I, de Boer MA, Postmus PE, van Hinsbergh V, Van Nieuw Amerongen GP, et al. Effective reduction of MCT-PAH by Fasudil. Comparison with bosentan and sildenafil. Eur Respir J. 2010;36:800–807. doi: 10.1183/09031936.00130209. [DOI] [PubMed] [Google Scholar]
  110. Murray F, MacLean MR, Pyne NJ. Increased expression of the cGMP-inhibited cAMP-specific (PDE3) and cGMP binding cGMP-specific (PDE5) phosphodiesterases in models of pulmonary hypertension. Br J Pharmacol. 2002;137:1187–1194. doi: 10.1038/sj.bjp.0704984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Murray F, Patel HH, Suda RY, Zhang S, Thistlethwaite PA, Yuan JX, et al. Expression and activity of cAMP phosphodiesterase isoforms in pulmonary artery smooth muscle cells from patients with pulmonary hypertension: role for PDE1. Am J Physiol Lung Cell Mol Physiol. 2007;292:L294–L303. doi: 10.1152/ajplung.00190.2006. [DOI] [PubMed] [Google Scholar]
  112. Nandi M, Miller A, Stidwill R, Jacques TS, Lam AA, Haworth S, et al. Pulmonary hypertension in a GTP-cyclohydrolase 1-deficient mouse. Circulation. 2005;111:2086–2090. doi: 10.1161/01.CIR.0000163268.32638.F4. [DOI] [PubMed] [Google Scholar]
  113. Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: structures, properties, and functions. Physiol Rev. 1999;79:1193–1226. doi: 10.1152/physrev.1999.79.4.1193. [DOI] [PubMed] [Google Scholar]
  114. National Pulmonary Hypertension Centres of the UK and Ireland. Consensus statement on the management of pulmonary hypertension in clinical practice in the UK and Ireland. Thorax. 2008;63:ii1–ii41. doi: 10.1136/thx.2007.090480. [DOI] [PubMed] [Google Scholar]
  115. Newman JH, Trembath RC, Morse JA, Grunig E, Loyd JE, Adnot S, et al. Genetic basis of pulmonary arterial hypertension: current understanding and future directions. J Am Coll Cardiol. 2004;43:33S–39S. doi: 10.1016/j.jacc.2004.02.028. [DOI] [PubMed] [Google Scholar]
  116. Newman JH, Phillips JA, III, Loyd JE. Narrative review: the enigma of pulmonary arterial hypertension: new insights from genetic studies. Ann Intern Med. 2008;148:278–283. doi: 10.7326/0003-4819-148-4-200802190-00006. [DOI] [PubMed] [Google Scholar]
  117. Nisbet RE, Sutliff RL, Hart CM. The role of peroxisome proliferator-activated receptors in pulmonary vascular disease. PPAR Res. 2007;2007:18797. doi: 10.1155/2007/18797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Nishimura T, Faul JL, Berry GJ, Vaszar LT, Qiu D, Pearl RG, et al. 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] [PubMed] [Google Scholar]
  119. Oka M, Homma N, Taraseviciene-Stewart L, Morris KG, Kraskauskas D, Burns N, et al. Rho kinase-mediated vasoconstriction is important in severe occlusive pulmonary arterial hypertension in rats. Circ Res. 2007a;100:923–929. doi: 10.1161/01.RES.0000261658.12024.18. [DOI] [PubMed] [Google Scholar]
  120. Oka M, Karoor V, Homma N, Nagaoka T, Sakao E, Golembeski SM, et al. Dehydroepiandrosterone upregulates soluble guanylate cyclase and inhibits hypoxic pulmonary hypertension. Cardiovasc Res. 2007b;74:377–387. doi: 10.1016/j.cardiores.2007.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Oka M, Fagan KA, Jones PL, McMurtry IF. Therapeutic potential of RhoA/Rho kinase inhibitors in pulmonary hypertension. Br J Pharmacol. 2008;155:444–454. doi: 10.1038/bjp.2008.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Okolicany J, McEnroe GA, Koh GY, Lewicki JA, Maack T. Clearance receptor and neutral endopeptidase-mediated metabolism of atrial natriuretic factor. Am J Physiol Renal Physiol. 1992;263:F546–F553. doi: 10.1152/ajprenal.1992.263.3.F546. [DOI] [PubMed] [Google Scholar]
  123. Oliver PM, Fox JE, Kim R, Rockman HA, Kim HS, Reddick RL, et al. Hypertension, cardiac hypertrophy, and sudden death in mice lacking natriuretic peptide receptor A. Proc Natl Acad Sci USA. 1997;94:14730–14735. doi: 10.1073/pnas.94.26.14730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Paddenberg R, Stieger P, von Lilien AL, Faulhammer P, Goldenberg A, Tillmanns HH, et al. Rapamycin attenuates hypoxia-induced pulmonary vascular remodeling and right ventricular hypertrophy in mice. Respir Res. 2007;8:15. doi: 10.1186/1465-9921-8-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Patterson KC, Weissmann A, Ahmadi T, Farber HW. Imatinib mesylate in the treatment of refractory idiopathic pulmonary arterial hypertension. Ann Intern Med. 2006;145:152–153. doi: 10.7326/0003-4819-145-2-200607180-00020. [DOI] [PubMed] [Google Scholar]
  126. Perros F, Montani D, Dorfmuller P, Durand-Gasselin I, Tcherakian C, Le PJ, et al. Platelet-derived growth factor expression and function in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2008;178:81–88. doi: 10.1164/rccm.200707-1037OC. [DOI] [PubMed] [Google Scholar]
  127. Potter LR, Abbey-Hosch S, Dickey DM. Natriuretic peptides, their receptors, and cyclic guanosine monophosphate-dependent signaling functions. Endocr Rev. 2006;27:47–72. doi: 10.1210/er.2005-0014. [DOI] [PubMed] [Google Scholar]
  128. Pozeg ZI, Michelakis ED, McMurtry MS, Thebaud B, Wu XC, Dyck JR, et al. 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] [PubMed] [Google Scholar]
  129. Preston IR, Hill NS, Gambardella LS, Warburton RR, Klinger JR. Synergistic effects of ANP and sildenafil on cGMP levels and amelioration of acute hypoxic pulmonary hypertension. Exp Biol Med. 2004;229:920–925. doi: 10.1177/153537020422900908. [DOI] [PubMed] [Google Scholar]
  130. Provencher S, Herve P, Jais X, Lebrec D, Humbert M, Simonneau G, et al. Deleterious effects of beta-blockers on exercise capacity and hemodynamics in patients with portopulmonary hypertension. Gastroenterology. 2006;130:120–126. doi: 10.1053/j.gastro.2005.10.013. [DOI] [PubMed] [Google Scholar]
  131. Qi JG, Ding YG, Tang CS, Du JB. Chronic administration of adrenomedullin attenuates hypoxic pulmonary vascular structural remodeling and inhibits proadrenomedullin N-terminal 20-peptide production in rats. Peptides. 2007;28:910–919. doi: 10.1016/j.peptides.2006.12.008. [DOI] [PubMed] [Google Scholar]
  132. Raja SG. Macitentan, a tissue-targeting endothelin receptor antagonist for the potential oral treatment of pulmonary arterial hypertension and idiopathic pulmonary fibrosis. Curr Opin Investig Drugs. 2010;11:1066–1073. [PubMed] [Google Scholar]
  133. Reynolds AM, Xia W, Holmes MD, Hodge SJ, Danilov S, Curiel DT, et al. Bone morphogenetic protein type 2 receptor gene therapy attenuates hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1182–L1192. doi: 10.1152/ajplung.00020.2006. [DOI] [PubMed] [Google Scholar]
  134. Rich S, Brundage BH. High-dose calcium channel-blocking therapy for primary pulmonary hypertension: evidence for long-term reduction in pulmonary arterial pressure and regression of right ventricular hypertrophy. Circulation. 1987;76:135–141. doi: 10.1161/01.cir.76.1.135. [DOI] [PubMed] [Google Scholar]
  135. Roberts JD, Polaner DM, Lang P, Zapol WM. Inhaled nitric oxide in persistent pulmonary hypertension of the newborn. Lancet. 1992;340:818–819. doi: 10.1016/0140-6736(92)92686-a. [DOI] [PubMed] [Google Scholar]
  136. Roberts KE, Fallon MB, Krowka MJ, Benza RL, Knowles JA, Badesch DB, et al. 5-HT transporter polymorphisms in patients with portopulmonary hypertension. Chest. 2009;135:1470–1475. doi: 10.1378/chest.08-1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Rubens C, Ewert R, Halank M, Wensel R, Orzechowski HD, Schultheiss HP, et al. Big endothelin-1 and endothelin-1 plasma levels are correlated with the severity of primary pulmonary hypertension. Chest. 2001;120:1562–1569. doi: 10.1378/chest.120.5.1562. [DOI] [PubMed] [Google Scholar]
  138. Rubin LJ, Mendoza J, Hood M, McGoon M, Barst R, Williams WB, et al. Treatment of primary pulmonary hypertension with continuous intravenous prostacyclin (epoprostenol). Results of a randomized trial. Ann Intern Med. 1990;112:485–491. doi: 10.7326/0003-4819-112-7-485. [DOI] [PubMed] [Google Scholar]
  139. Rybalkin SD, Rybalkina I, Beavo JA, Bornfeldt KE. Cyclic nucleotide phosphodiesterase 1C promotes human arterial smooth muscle cell proliferation. Circ Res. 2002;90:151–157. doi: 10.1161/hh0202.104108. [DOI] [PubMed] [Google Scholar]
  140. Saadjian A, Philip-Joet F, Paganelli F, Arnaud A, Levy S. Long-term effects of cicletanine on secondary pulmonary hypertension. J Cardiovasc Pharmacol. 1998;31:364–371. doi: 10.1097/00005344-199803000-00006. [DOI] [PubMed] [Google Scholar]
  141. Said SI, Hamidi SA, Dickman KG, Szema AM, Lyubsky S, Lin RZ, et al. 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] [PubMed] [Google Scholar]
  142. Sasaki K, Heeschen C, Aicher A, Ziebart T, Honold J, Urbich C, et al. Ex vivo pretreatment of bone marrow mononuclear cells with endothelial NO synthase enhancer AVE9488 enhances their functional activity for cell therapy. Proc Natl Acad Sci USA. 2006;103:14537–14541. doi: 10.1073/pnas.0604144103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Schermuly RT, Weissmann N, Enke B, Ghofrani HA, Forssmann WG, Grimminger F, et al. Urodilatin, a natriuretic peptide stimulating particulate guanylate cyclase, and the phosphodiesterase 5 inhibitor dipyridamole attenuate experimental pulmonary hypertension: synergism upon coapplication. Am J Respir Cell Mol Biol. 2001;25:219–225. doi: 10.1165/ajrcmb.25.2.4256. [DOI] [PubMed] [Google Scholar]
  144. Schermuly RT, Kreisselmeier KP, Ghofrani HA, Samidurai A, Pullamsetti S, Weissmann N, et al. Antiremodeling effects of iloprost and the dual-selective phosphodiesterase 3/4 inhibitor tolafentrine in chronic experimental pulmonary hypertension. Circ Res. 2004;94:1101–1108. doi: 10.1161/01.RES.0000126050.41296.8E. [DOI] [PubMed] [Google Scholar]
  145. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R, Roth M, et al. Reversal of experimental pulmonary hypertension by PDGF inhibition. J Clin Invest. 2005;115:2811–2821. doi: 10.1172/JCI24838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Schermuly RT, Pullamsetti SS, Kwapiszewska G, Dumitrascu R, Tian X, Weissmann N, et al. Phosphodiesterase 1 upregulation in pulmonary arterial hypertension: target for reverse-remodeling therapy. Circulation. 2007;115:2331–2339. doi: 10.1161/CIRCULATIONAHA.106.676809. [DOI] [PubMed] [Google Scholar]
  147. Schermuly RT, Stasch JP, Pullamsetti SS, Middendorff R, Muller D, Schluter KD, et al. Expression and function of soluble guanylate cyclase in pulmonary arterial hypertension. Eur Respir J. 2008;32:881–891. doi: 10.1183/09031936.00114407. [DOI] [PubMed] [Google Scholar]
  148. Schmidt HH, Schmidt PM, Stasch JP. NO- and Haem-independent soluble guanylate cyclase activators. Handb Exp Pharmacol. 2009;191:309–339. doi: 10.1007/978-3-540-68964-5_14. [DOI] [PubMed] [Google Scholar]
  149. Sebkhi A, Strange JW, Phillips SC, Wharton J, Wilkins MR. Phosphodiesterase type 5 as a target for the treatment of hypoxia-induced pulmonary hypertension. Circulation. 2003;107:3230–3235. doi: 10.1161/01.CIR.0000074226.20466.B1. [DOI] [PubMed] [Google Scholar]
  150. Shaul PW, Yuhanna IS, German Z, Chen Z, Steinhorn RH, Morin FC., III Pulmonary endothelial NO synthase gene expression is decreased in fetal lambs with pulmonary hypertension. Am J Physiol. 1997;272:L1005–L1012. doi: 10.1152/ajplung.1997.272.5.L1005. [DOI] [PubMed] [Google Scholar]
  151. Shenoy V, Ferreira AJ, Qi Y, Fraga-Silva RA, Díez-Freire C, Dooies A, et al. The ACE2/Ang-(1-7)/Mas axis confers cardiopulmonary protection against lung fibrosis and pulmonary hypertension. Am J Respir Crit Care Med. 2010;182:1065–L1072. doi: 10.1164/rccm.200912-1840OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. 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] [PubMed] [Google Scholar]
  153. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix M, Denton CP, et al. Updated clinical classification of pulmonary hypertension. J Am Coll Cardiol. 2009;54:S43–S54. doi: 10.1016/j.jacc.2009.04.012. [DOI] [PubMed] [Google Scholar]
  154. Sitbon O, Humbert M, Jais X, Ioos V, Hamid AM, Provencher S, et al. Long-term response to calcium channel blockers in idiopathic pulmonary arterial hypertension. Circulation. 2005;111:3105–3111. doi: 10.1161/CIRCULATIONAHA.104.488486. [DOI] [PubMed] [Google Scholar]
  155. Sobolewski A, Rudarakanchana N, Upton PD, Yang J, Crilley TK, Trembath RC, et al. Failure of bone morphogenetic protein receptor trafficking in pulmonary arterial hypertension: potential for rescue. Hum Mol Genet. 2008;17:3180–3190. doi: 10.1093/hmg/ddn214. [DOI] [PubMed] [Google Scholar]
  156. Song Y, Jones JE, Beppu H, Keaney JF, Jr, Loscalzo J, Zhang YY. Increased susceptibility to pulmonary hypertension in heterozygous BMPR2-mutant mice. Circulation. 2005;112:553–562. doi: 10.1161/CIRCULATIONAHA.104.492488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Souza R, Sitbon O, Parent F, Simonneau G, Humbert M. Long term imatinib treatment in pulmonary arterial hypertension. Thorax. 2006;61:736. doi: 10.1136/thx.2006.064097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Stasch JP, Hobbs AJ. NO-independent, haem-dependent soluble guanylate cyclase stimulators. Handb Exp Pharmacol. 2009;191:277–308. doi: 10.1007/978-3-540-68964-5_13. [DOI] [PubMed] [Google Scholar]
  159. Stasch JP, Alonso-Alija C, Apeler H, Dembowsky K, Feurer A, Minuth T, et al. Pharmacological actions of a novel NO-independent guanylyl cyclase stimulator, BAY 41-8543: in vitro studies. Br J Pharmacol. 2002a;135:333–343. doi: 10.1038/sj.bjp.0704484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Stasch JP, Dembowsky K, Perzborn E, Stahl E, Schramm M. Cardiovascular actions of a novel NO-independent guanylyl cyclase stimulator, BAY 41-8543: in vivo studies. Br J Pharmacol. 2002b;135:344–355. doi: 10.1038/sj.bjp.0704483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Steudel W, Ichinose F, Huang PL, Hurford WE, Jones RC, Bevan JA, et al. Pulmonary vasoconstriction and hypertension in mice with targeted disruption of the endothelial nitric oxide synthase (NOS 3) gene. Circ Res. 1997;81:34–41. doi: 10.1161/01.res.81.1.34. [DOI] [PubMed] [Google Scholar]
  162. Stiebellehner L, Petkov V, Vonbank K, Funk G, Schenk P, Ziesche R, et al. Long-term treatment with oral sildenafil in addition to continuous IV epoprostenol in patients with pulmonary arterial hypertension. Chest. 2003;123:1293–1295. doi: 10.1378/chest.123.4.1293. [DOI] [PubMed] [Google Scholar]
  163. Stocker C, Penny DJ, Brizard CP, Cochrane AD, Soto R, Shekerdemian LS. Intravenous sildenafil and inhaled nitric oxide: a randomised trial in infants after cardiac surgery. Intensive Care Med. 2003;29:1996–2003. doi: 10.1007/s00134-003-2016-4. [DOI] [PubMed] [Google Scholar]
  164. Sun CK, Lee FY, Sheu JJ, Yuen CM, Chua S, Chung SY, et al. Early combined treatment with cilostazol and bone marrow-derived endothelial progenitor cells markedly attenuates pulmonary arterial hypertension in rats. J Pharmacol Exp Ther. 2009;330:718–726. doi: 10.1124/jpet.109.154328. [DOI] [PubMed] [Google Scholar]
  165. Tapper EB, Knowles D, Heffron T, Lawrence EC, Csete M. Portopulmonary hypertension: imatinib as a novel treatment and the Emory experience with this condition. Transplant Proc. 2009;41:1969–1971. doi: 10.1016/j.transproceed.2009.02.100. [DOI] [PubMed] [Google Scholar]
  166. Ten FH, Dumitrescu D, Bovenschulte H, Erdmann E, Rosenkranz S. Significant improvement of right ventricular function by imatinib mesylate in scleroderma-associated pulmonary arterial hypertension. Clin Res Cardiol. 2009;98:265–267. doi: 10.1007/s00392-009-0752-3. [DOI] [PubMed] [Google Scholar]
  167. Thenappan T, Shah SJ, Rich S, Gomberg-Maitland M. A USA-based registry for pulmonary arterial hypertension: 1982–2006. Eur Respir J. 2007;30:1103–1110. doi: 10.1183/09031936.00042107. [DOI] [PubMed] [Google Scholar]
  168. Toshner M, Voswinckel R, Southwood M, Al-Lamki R, Howard LS, Marchesan D, et al. Evidence of dysfunction of endothelial progenitors in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2009;180:780–787. doi: 10.1164/rccm.200810-1662OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Tuder RM, Cool CD, Geraci MW, Wang J, Abman SH, Wright L, et al. 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. [DOI] [PubMed] [Google Scholar]
  170. Vanderford PA, Wong J, Chang R, Keefer LK, Soifer SJ, Fineman JR. Diethylamine/nitric oxide (NO) adduct, an NO donor, produces potent pulmonary and systemic vasodilation in intact newborn lambs. J Cardiovasc Pharmacol. 1994;23:113–119. doi: 10.1097/00005344-199401000-00016. [DOI] [PubMed] [Google Scholar]
  171. Velez-Roa S, Ciarka A, Najem B, Vachiery JL, Naeije R, van de Borne P. Increased sympathetic nerve activity in pulmonary artery hypertension. Circulation. 2004;110:1308–1312. doi: 10.1161/01.CIR.0000140724.90898.D3. [DOI] [PubMed] [Google Scholar]
  172. Vermeersch P, Buys E, Pokreisz P, Marsboom G, Ichinose F, Sips P, et al. Soluble guanylate cyclase-alpha1 deficiency selectively inhibits the pulmonary vasodilator response to nitric oxide and increases the pulmonary vascular remodeling response to chronic hypoxia. Circulation. 2007;116:936–943. doi: 10.1161/CIRCULATIONAHA.106.677245. [DOI] [PubMed] [Google Scholar]
  173. Voelkel NF, Quaife RA, Leinwand LA, Barst RJ, McGoon MD, Meldrum DR, et al. Right ventricular function and failure: report of a National Heart, Lung, and Blood Institute working group on cellular and molecular mechanisms of right heart failure. Circulation. 2006;114:1883–1891. doi: 10.1161/CIRCULATIONAHA.106.632208. [DOI] [PubMed] [Google Scholar]
  174. Weissmann N, Hackemack S, Dahal BK, Pullamsetti SS, Savai R, Mittal M, et al. The soluble guanylate cyclase activator HMR1766 reverses hypoxia-induced experimental pulmonary hypertension in mice. Am J Physiol Lung Cell Mol Physiol. 2009;297:L658–L665. doi: 10.1152/ajplung.00189.2009. [DOI] [PubMed] [Google Scholar]
  175. Welsh DJ, Harnett M, MacLean M, Peacock AJ. Proliferation and signaling in fibroblasts: role of 5-hydroxytryptamine2A receptor and transporter. Am J Respir Crit Care Med. 2004;170:252–259. doi: 10.1164/rccm.200302-264OC. [DOI] [PubMed] [Google Scholar]
  176. Westermann D, Riad A, Richter U, Jager S, Savvatis K, Schuchardt M, et al. Enhancement of the endothelial NO synthase attenuates experimental diastolic heart failure. Basic Res Cardiol. 2009;104:499–509. doi: 10.1007/s00395-009-0014-6. [DOI] [PubMed] [Google Scholar]
  177. Wharton J, Strange JW, Moller GM, Growcott EJ, Ren X, Franklyn AP, et al. Antiproliferative effects of phosphodiesterase type 5 inhibition in human pulmonary artery cells. Am J Respir Crit Care Med. 2005;172:105–113. doi: 10.1164/rccm.200411-1587OC. [DOI] [PubMed] [Google Scholar]
  178. Wilkens H, Guth A, Konig J, Forestier N, Cremers B, Hennen B, et al. Effect of inhaled iloprost plus oral sildenafil in patients with primary pulmonary hypertension. Circulation. 2001;104:1218–1222. doi: 10.1161/hc3601.096826. [DOI] [PubMed] [Google Scholar]
  179. Wilkins MR, Ali O, Bradlow W, Wharton J, Taegtmeyer A, Rhodes CJ, et al. Simvastatin as a treatment for pulmonary hypertension trial. Am J Respir Crit Care Med. 2010;181:1106–1113. doi: 10.1164/rccm.2009111-699OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Willers ED, Newman JH, Loyd JE, Robbins IM, Wheeler LA, Prince MA, III, et al. 5-HT transporter polymorphisms in familial and idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;173:798–802. doi: 10.1164/rccm.200509-1361OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Williamson DJ, Wallman LL, Jones R, Keogh AM, Scroope F, Penny R, et al. Hemodynamic effects of Bosentan, an endothelin receptor antagonist, in patients with pulmonary hypertension. Circulation. 2000;102:411–418. doi: 10.1161/01.cir.102.4.411. [DOI] [PubMed] [Google Scholar]
  182. Wohlfart P, Xu H, Endlich A, Habermeier A, Closs EI, Hubschle T, et al. Antiatherosclerotic effects of small-molecular-weight compounds enhancing endothelial nitric-oxide synthase (eNOS) expression and preventing eNOS uncoupling. J Pharmacol Exp Ther. 2008;325:370–379. doi: 10.1124/jpet.107.128009. [DOI] [PubMed] [Google Scholar]
  183. Xie W, Wang H, Wang H, Hu G. Effects of iptakalim hydrochloride, a novel KATP channel opener, on pulmonary vascular remodeling in hypoxic rats. Life Sci. 2004;75:2065–2076. doi: 10.1016/j.lfs.2004.03.031. [DOI] [PubMed] [Google Scholar]
  184. Yang X, Long L, Southwood M, Rudarakanchana N, Upton PD, Jeffery TK, et al. 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] [PubMed] [Google Scholar]
  185. Yip HK, Chang LT, Sun CK, Sheu JJ, Chiang CH, Youssef AA, et al. Autologous transplantation of bone marrow-derived endothelial progenitor cells attenuates monocrotaline-induced pulmonary arterial hypertension in rats. Crit Care Med. 2008;36:873–880. doi: 10.1097/CCM.0B013E318165B7EA. [DOI] [PubMed] [Google Scholar]
  186. Yu Y, Fantozzi I, Remillard CV, Landsberg JW, Kunichika N, Platoshyn O, et al. 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] [PMC free article] [PubMed] [Google Scholar]
  187. Yu Y, Keller SH, Remillard CV, Safrina O, Nicholson A, Zhang SL, et al. A functional single-nucleotide polymorphism in the TRPC6 gene promoter associated with idiopathic pulmonary arterial hypertension. Circulation. 2009;119:2313–2322. doi: 10.1161/CIRCULATIONAHA.108.782458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Yuan JX, Rubin LJ. Pathogenesis of pulmonary arterial hypertension: the need for multiple hits. Circulation. 2005;111:534–538. doi: 10.1161/01.CIR.0000156326.48823.55. [DOI] [PubMed] [Google Scholar]
  189. Yuan XJ, Wang J, Juhaszova M, Gaine SP, Rubin LJ. Attenuated K+ channel gene transcription in primary pulmonary hypertension. Lancet. 1998;351:726–727. doi: 10.1016/S0140-6736(05)78495-6. [DOI] [PubMed] [Google Scholar]
  190. Zhai FG, Zhang XH, Wang HL. Fluoxetine protects against monocrotaline-induced pulmonary arterial hypertension: potential roles of induction of apoptosis and upregulation of Kv1.5 channels in rats. Clin Exp Pharmacol Physiol. 2009;36:850–856. doi: 10.1111/j.1440-1681.2009.05168.x. [DOI] [PubMed] [Google Scholar]
  191. Zhao L, Hughes JM, Winter RJ. Effects of natriuretic peptides and neutral endopeptidase 24.11 inhibition in isolated perfused rat lung. Am Rev Respir Dis. 1992;146:1198–1201. doi: 10.1164/ajrccm/146.5_Pt_1.1198. [DOI] [PubMed] [Google Scholar]
  192. Zhao L, Long L, Morrell NW, Wilkins MR. NPR-A-Deficient mice show increased susceptibility to hypoxia-induced pulmonary hypertension. Circulation. 1999;99:605–607. doi: 10.1161/01.cir.99.5.605. [DOI] [PubMed] [Google Scholar]
  193. Zhao L, Mason NA, Strange JW, Walker H, Wilkins MR. Beneficial effects of phosphodiesterase 5 inhibition in pulmonary hypertension are influenced by natriuretic Peptide activity. Circulation. 2003;107:234–237. doi: 10.1161/01.cir.0000050653.10758.6b. [DOI] [PubMed] [Google Scholar]
  194. Zhao YY, Zhao YD, Mirza MK, Huang JH, Potula HH, Vogel SM, et al. Persistent eNOS activation secondary to caveolin-1 deficiency induces pulmonary hypertension in mice and humans through PKG nitration. J Clin Invest. 2009;119:2009–2018. doi: 10.1172/JCI33338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Zhu Y, Zhang S, Xie W, Li Q, Zhou Y, Wang H. Iptakalim inhibited endothelin-1-induced proliferation of human pulmonary arterial smooth muscle cells through the activation of K(ATP) channel. Vascul Pharmacol. 2008;48:92–99. doi: 10.1016/j.vph.2008.01.001. [DOI] [PubMed] [Google Scholar]
  196. Zhu SP, Mao ZF, Huang J, Wang JY. Continuous fluoxetine administration prevents recurrence of pulmonary arterial hypertension and prolongs survival in rats. Clin Exp Pharmacol Physiol. 2009;36:e1–e5. doi: 10.1111/j.1440-1681.2009.05181.x. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

bph0163-0125-SD1.pptx (366.5KB, pptx)

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES