REVIEW ARTICLE

The role of the renin-angiotensin-aldosterone system in the pathobiology of pulmonary arterial hypertension (2013 Grover Conference series) Bradley A. Maron,1,2 Jane A. Leopold1 1

Brigham and Women’s Hospital, Division of Cardiovascular Medicine, Boston, Massachusetts, USA; 2Veterans Affairs Boston Healthcare System, Department of Cardiology, 1400 VFW Parkway, Boston, Massachusetts, USA

Abstract: Pulmonary arterial hypertension (PAH) is associated with aberrant pulmonary vascular remodeling that leads to increased pulmonary artery pressure, pulmonary vascular resistance, and right ventricular dysfunction. There is now accumulating evidence that the renin-angiotensin-aldosterone system is activated and contributes to cardiopulmonary remodeling that occurs in PAH. Increased plasma and lung tissue levels of angiotensin and aldosterone have been detected in experimental models of PAH and shown to correlate with cardiopulmonary hemodynamics and pulmonary vascular remodeling. These processes are abrogated by treatment with angiotensin receptor or mineralocorticoid receptor antagonists. At a cellular level, angiotensin and aldosterone activate oxidant stress signaling pathways that decrease levels of bioavailable nitric oxide, increase inflammation, and promote cell proliferation, migration, extracellular matrix remodeling, and fibrosis. Clinically, enhanced renin-angiotensin activity and elevated levels of aldosterone have been detected in patients with PAH, which suggests a role for angiotensin and mineralocorticoid receptor antagonists in the treatment of PAH. This review will examine the current evidence linking renin-angiotensinaldosterone system activation to PAH with an emphasis on the cellular and molecular mechanisms that are modulated by aldosterone and may be of importance for the pathobiology of PAH. Keywords: aldosterone, angiotensin II, mineralocorticoid receptor, pulmonary arterial hypertension, spironolactone. Pulm Circ 2014;4(2):200-210. DOI: 10.1086/675984.

Pulmonary arterial hypertension (PAH) is an insidious disease with a poor long-term prognosis. In 2009, an epidemiological study estimated the incidence of PAH to be 1.1 cases per million per year and the prevalence to be 6.6 cases per million, with incident survival rates at 1, 2, and 3 years of 85.7%, 69.6%, and 54.9%, respectively, despite medical therapy.1,2 Although the etiology of PAH may vary between patients, the resultant cardiac and pulmonary vascular pathophenotypes are remarkably similar. The disease pathobiology is characterized by aberrant distal pulmonary vascular remodeling resulting from endothelial dysfunction, dysregulated vascular smooth muscle cell proliferation, and inflammation that promotes neointimal formation, thrombosis, and luminal obliteration.3 The formation of these plexiform lesions contributes to impaired vascular reactivity, elevated pulmonary artery

pressure, and increased pulmonary vascular resistance, which, in turn, initiates maladaptive right ventricular (RV) remodeling.4,5 The continued strain imposed on the RVpulmonary vascular circuit by these remodeling processes ultimately leads to RV failure and death.4,5 Although our understanding of the PAH disease process has advanced over the past few decades, therapeutic targets and evidencebased interventions remain limited, and novel approaches to modify the underlying disease process continue to be pursued.6 The renin-angiotensin-aldosterone system (RAAS) maintains blood pressure through its effects on the kidney to regulate sodium and water balance and on peripheral blood vessels to increase systemic vascular resistance.7 In 1963, 10 years after the discovery of aldosterone, investigators infused radioactive aldosterone into rats and demonstrated

Address correspondence to Dr. Jane A. Leopold, Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, NRB0630K, Boston, MA 02115, USA. E-mail: [email protected]. Submitted September 22, 2013; Accepted January 17, 2014; Electronically published May 15, 2014. © 2014 by the Pulmonary Vascular Research Institute. All rights reserved. 2045-8932/2014/0402-0008. $15.00.

Pulmonary Circulation

that, in addition to its localization to the kidney, it was also taken up and metabolized by the lungs, heart, and aorta. Preliminary histological assessment using autoradiography indicated that aldosterone was localized to cardiomyocytes in the heart and to the alveolar walls of the lungs; the methodology used could not distinguish the pulmonary vessels from the alveoli, and it remained unknown for decades whether the pulmonary vasculature participated in aldosterone metabolism.8 Since that time, there has been extensive investigation of the effects of aldosterone on the kidneys and systemic circulation, with recent interest in its role in modulating the RV-pulmonary vascular axis in PAH. This is not surprising, given that the cardiopulmonary remodeling that occurs with PAH recapitulates, in part, the effects of RAAS activation and, in particular, aldosterone, in the systemic circulation. Aldosterone has been linked to the development of systemic hypertension, left ventricular cardiac and systemic vascular remodeling, and left heart failure.9-11 Community-based longitudinal or cross-sectional studies have shown that higher levels of aldosterone are associated with an increased risk for developing arterial stiffness, incident hypertension, or left ventricular hypertrophy, which suggests that aldosterone is either causative or a disease modifier that facilitates adaptive cardiovascular remodeling.9,10 RE N I N -A N G I O T E N S I N SY S T E M The classical teaching is that the primary function of the RAAS is to regulate intravascular volume to maintain blood pressure (Fig. 1). In response to a real (blood loss, dehydration) or perceived (ventricular pump failure) decrease in circulating blood volume, the juxtaglomerular cells lining the afferent arterioles located proximal to the renal glomeruli release renin into the circulation. Renin cleaves angiotensinogen, which is synthesized and secreted by the liver, to angiotensin I. Circulating angiotensin I is then hydrolyzed to angiotensin II by angiotensinconverting enzyme (ACE), which is located primarily in the pulmonary and renal endothelium. Angiotensin II activates the angiotensin type 1 (AT1) receptor to initiate a vasoconstrictor response and stimulate aldosterone synthesis by the adrenal gland. Aldosterone then acts on the renal tubules to initiate sodium and water retention to increase circulating blood volume and, thereby, blood pressure (reviewed in Santos et al.12 and Briet and Schiffrin13). It is now recognized that the angiotensin arm of this hormonal pathway is far more complex than initially understood (Fig. 2). In fact, there are alternative pathways for angiotensin II generation, a number of vasoactive angiotensin cleavage peptides, and other angiotensin recep-

Volume 4

Number 2

June 2014

| 201

Figure 1. The renin-angiotensin-aldosterone system. In response to a drop in blood pressure or sodium intake (A), renin is released from the juxtaglomerular cells of the kidney to increase angiotensin II levels (B). Angiotensin II increases blood pressure by stimulating the production of aldosterone by the adrenal gland (C) to initiate salt and water retention and vasoconstriction of systemic resistance vessels (D). ACE ¼ angiotensin-converting enzyme.

tors that regulate cardiovascular hemodynamics and remodeling. In these nonclassical pathways, angiotensin II may be generated by the proteases chymase, which is present in mast cells and skeletal muscle, or cathepsin G, which exists in inflammatory cells.14 Angiotensin II exerts its effects primarily by binding to and activating the AT1 and angiotensin type 2 (AT2) receptors to modulate vascular tone; activation of the AT1 receptor initiates a vasoconstrictor response, whereas AT2 receptor signaling promotes vasodilation. A homologue of ACE, ACE2, hydrolyzes either angiotensin I or angiotensin II to generate the heptapeptide angiotensin-(1–7). Angiotensin-(1–7) may also be generated directly from angiotensin I through the actions of neutralendopeptidase (neprilysin) and prolyl-carboxypeptidase.12 Angiotensin-(1–7) binds to the G-protein coupled Mas receptor, opposes the vasoconstrictor effects of angiotensin II, and does not stimulate aldosterone secretion.12 By contrast, angiotensin III, another heptapeptide metabolite of angiotensin II, does stimulate aldosterone secretion through activation of the AT1 receptor.15 The complexities of the angiotensin pathway indicate that the pathophysiological consequences of reninangiotensin system activation in PAH may result from an imbalance between angiotensin II and other vasculopro-

202

| Renin-angiotensin-aldosterone system in pulmonary arterial hypertension

Maron and Leopold

Figure 2. Angiotensin signaling. Angiotensin II may be generated from angiotensin I by the actions of angiotensin-converting enzyme (ACE) or chymase. Angiotensin I may also be cleaved by angiotensin-converting enzyme 2 (ACE2) or neutral endopeptidase (neprilysin) to yield angiotensin-(1–7). Angiotensin II binds to the angiotensin type 1 (AT1) receptor and the angiotensin type 2 (AT2) receptor, which have opposing effects on vascular structure and function. In addition, angiotensin-(1–7), which binds to the Mas receptor, is considered vasculoprotective.

tective angiotensin cleavage peptides that favors vasoconstriction and impairs vascular reactivity. In addition, these angiotensin signaling peptides have opposing effects on modulating other vascular smooth muscle cell functions important for vessel remodeling in PAH. For example, angiotensin II increases intracellular free Ca2+ levels and activates the Na+/H+ exchanger, tyrosine kinase signaling, mitogenactivated protein kinase signaling and reactive oxygen species production to stimulate vascular smooth muscle cell proliferation and migration.16 By contrast, angiotensin-(1–7) inhibits these signaling pathways, limits proliferation and migration, and maintains vascular smooth muscle cell homeostatic processes.17 RE NI N -A NG I O TEN SI N ACT IV AT IO N I N P AH In PAH, there is clear evidence that the renin-angiotensin signaling system is activated and contributes to the disease pathology. Early preclinical studies performed in the rat hypobaric hypoxia model associated angiotensin with pulmonary hypertension by demonstrating an increase in pulmonary vascular ACE messenger RNA (mRNA) and protein expression with an approximately 50% increase in ACE activity in remodeled pulmonary vessels.18 Subsequent studies found that angiotensin signaling in remodeled vessels favored pulmonary artery smooth muscle cell proliferation and vasoconstriction with evidence of AT1

receptor upregulation and a concomitant downregulation of ACE2 and angiotensin-(1–7).19-22 The relevance of these findings for human disease was confirmed through pathological examination of lung sections from individuals with PAH. This revealed increased ACE and AT1receptor but not AT2-receptor expression in remodeled vessels, and pulmonary vascular smooth muscle cells isolated from these samples were found to be hyperproliferative under hypoxic conditions.19,23 Activation of the renin-angiotensin system was not limited to the pulmonary vasculature, and a 3.4-fold increase in ACE activity was detected in the RV after 14 days of hypoxia. Interestingly, increased ACE activity was localized to areas of myocardial fibrosis and occurred only in the RV, as there was a reduction in ACE expression in the free wall of the left ventricle in hypoxic rats.24 Evidence to support a causal role for angiotensin in PAH disease progression was provided by studies in hypoxic rats treated with the ACE inhibitor captopril or the AT1 receptor blocker losartan that demonstrated a decrease in the mean pulmonary artery pressure and RV hypertrophy as compared with saline-treated rats.25 Similarly, studies performed in the rat monocrotaline model of PAH found that losasrtan decreased vascular remodeling, improved RV performance by decreasing RV systolic pressure and end-diastolic diameter, but had no effect on

Pulmonary Circulation

RV hypertrophy or cardiac output. Losartan treatment did, however, decrease RV afterload, a marker of improved arterial elastance, to restore RV-arterial coupling, providing further evidence that inhibition of angiotensin signaling improved vascular remodeling.19 Investigators have also attempted to modulate the RVpulmonary vascular pathophenotype in PAH by augmenting ACE2 and angiotensin-(1–7) to overcome the deleterious effects of angiotensin II signaling. In a mouse model of pressure overload pulmonary hypertension, the administration of recombinant human ACE2 upregulated expression of the Mas receptor to increase angiotensin-(1–7) signaling, and it improved RV systolic and diastolic function and decreased RV hypertrophy without influencing left ventricular hemodynamics.26 These findings were supported by results from other studies that increased ACE2angiotensin-(1–7) signaling by gene transfer using a lentiviral packaged angiotensin-(1–7) fusion gene or ACE2 complementary DNA.27,28 Similarly, a pharmacological ACE2 activator XNT (1-[(2-dimethylamino) ethylamino]4-(hydroxymethyl)-7-[(4-methylphenyl) sulfonyl oxy]-9Hxanthene-9-one) diminished cardiopulmonary remodeling in PAH by increasing levels of the anti-inflammatory cytokine interleukin-10.29 The importance of neprilysin as a source of angiotensin-(1–7) was demonstrated in neprilysin null mice. Although there was no adverse effect of gene deletion at baseline, after exposure to chronic hypoxia, the mice developed exaggerated pulmonary pressures and distal arterial muscularization. Pulmonary artery smooth muscle cells isolated from these mice were hyperproliferative owing to Src kinase and platelet-derived growth factor receptor activation, suggesting that angiotensin-(1–7) plays a regulatory role in mediating tyrosine kinase receptor signaling.30 Findings from clinical studies indicate that RAAS activation occurs in patients with pulmonary hypertension. For example, there is an increased frequency of expression of an ACE gain-of-function polymorphism in patients with pulmonary hypertension compared with healthy individuals. Among 40 patients studied, however, the increase in ACE activity correlated with a higher cardiac output, and it was suggested that RAAS activation was merely a compensatory mechanism to prevent heart failure.31 More direct evidence for renin-angiotensin activation in patients with pulmonary hypertension was provided recently when investigators measured these components in serum samples obtained from 58 patients with idiopathic PAH (New York Heart Association class II, 60%; class III, 31%; class IV, 9%). In this cohort, serum renin and angiotensin I levels were increased above the upper limit of normal in the majority of patients, whereas angiotensin II levels were

Volume 4

Number 2

June 2014

| 203

elevated only in a smaller cohort. Follow-up studies (median duration, 39 months) indicated that persistently elevated levels of renin-angiotensin activity were associated with PAH disease progression assessed as a greater than 10% decrease in the 6-minute walk distance and an increased risk of lung transplantation or death.19 Despite the finding that renin-angiotensin levels were elevated in patients with PAH, early clinical studies with the ACE inhibitor captopril in this patient population yielded mixed results. Short-term (48-hour) administration of captopril had no effect on exercise-induced changes in pulmonary pressures, pulmonary vascular resistance, or cardiac output.32 Similarly, longer-term therapy was evaluated in a study involving 7 women treated with up to 300 mg/day of captopril for as long as 12 weeks. In these patients, captopril decreased blood pressure without influencing pulmonary artery pressure, resistance, cardiac output, or exercise tolerance.33 These outcomes contrast with findings from a pilot study involving 5 patients that enforced strict metabolic conditions, restricted sodium intake, and found that steady-state captopril dosing of 150 mg/day decreased mean pulmonary artery pressures and improved RV ejection fraction. Neurohormonal profiling in these patients identified a decrease in plasma angiotensin II and aldosterone levels with no effect of the drug on cortisol or norepinephrine levels.34 Conflicting results from these and other small studies led to a failure to reach a consensus opinion on the use of ACE inhibitors in patients with PAH, and the drugs were not evaluated in larger definitive clinical trials. Nonetheless, the observation that ACE inhibitors ameliorated disease only when aldosterone levels were decreased suggested that aldosterone may be the key component of the RAAS that mediates cardiopulmonary remodeling in PAH. Moreover, the mixed results from trials of ACE inhibition in PAH indicate that targeting the renin-angiotensin arm of the pathway alone may not be sufficient to limit disease progression and that the phenomenon of aldosterone breakthrough occurs in PAH. Aldosterone breakthrough was first described in patients with left heart failure who were treated optimally with β-blockers to inhibit renin and ACE inhibitors or AT1 receptor antagonists yet still had elevated levels of aldosterone.35 This may occur as a result of incomplete inhibition of ACE activity or angiotensin II– independent synthesis of aldosterone.36 Aldosterone breakthrough has also been proposed to account for the dramatic results of the Randomized Aldactone Evaluation Study (RALES), which demonstrated that aldosterone antagonism with spironolactone, when added to ACE inhibitors, decreased mortality and improved clinical outcomes.35

204

| Renin-angiotensin-aldosterone system in pulmonary arterial hypertension

ALDOSTERONE There are several lines of evidence that converge to identify aldosterone as an integral mediator of the cardiopulmonary vascular phenotype in PAH. Elevated levels of aldosterone have been detected in experimental models of PAH and humans with the disease.37-39 This occurs, in part, due to increased adrenal production of aldosterone as well as extra-adrenal synthesis of the hormone that has been identified in the pulmonary vasculature.37 These persistently elevated levels of aldosterone activate the mineralocorticoid receptor that is present in vascular and cardiac cells to initiate signaling pathways that promote vascular remodeling, impair vascular reactivity, and contribute to RV cardiac dysfunction associated with PAH. Aldosterone synthesis in the adrenal gland is stimulated by angiotensin II and endothelin-1, which are elevated in PAH, as well as adrenocorticotropic hormone, plasma potassium levels, and the relative level of serum sodium (reviewed in Beuschlein7 and Rossi35). Aldosterone is synthesized from cholesterol in a series of catalyzed reactions with two rate-limiting steps; the transport of cholesterol to the inner mitochondrial membrane by steroidogenic acute regulatory protein (StAR) and conversion of 11-deoxycorticosterone to aldosterone by the enzyme aldosterone synthase (CYP11B2). Transcription of the aldosterone synthase gene is calcium-dependent, and known aldosterone secretagogues increase calcium levels; angiotensin II releases calcium from intracellular stores, whereas changes in potassium levels increase calcium influx through voltage-dependent T- and L-type calcium.7 Although it is accepted that angiotensin II is generated by pulmonary vascular cells, the possibility that local extra-adrenal aldosterone synthesis occurs in the cardiopulmonary vascular system has been highly controversial.40 At present, there is compelling evidence to indicate that aldosterone synthesis in the heart is extremely limited or nonexistent, based on studies performed with adrenalectomized rats that were administered high- or low-salt diets. In these studies, CYP11B2 expression was examined by mRNA and found to be 1.05 × 10−6 of that measured for the adrenal gland. Aldosterone measured by enzyme-linked immunosorbent assay was detectable in the heart in only 30% of adrenalectomized animals, and the levels detected were less than 30 pmol/L, suggesting either extremely limited synthesis or the detection of residual bound aldosterone.40,41 Other studies that used sensitive mRNA detection methods found no evidence of CYP11B2 mRNA in hearts from rats subjected to a conditions that would stimulate aldosterone production, de-

Maron and Leopold

spite an assay sensitivity of less than 100 copies mRNA/μg total RNA.42 Aldosterone synthesis by vascular cells has been has been equally as controversial, with early studies detecting CYP11B2 expression in human umbilical vein endothelial cells but not pulmonary artery endothelial cells.43,44 The possibility of pulmonary vascular aldosterone synthesis was readdressed recently using more sensitive methodology, and CYP11B2 expression and aldosterone production were, in fact, detected in pulmonary artery endothelial cells stimulated with endothelin-1 or angiotensin II.37,41,43 These findings are in agreement with studies that analyzed and detected aldosterone production by mesenteric vessels using mass spectrometry and support the plausibility of pulmonary vascular extra-adrenal aldosterone synthesis.45 ALDOSTERONE IN PAH Hyperaldosteronism in PAH has been confirmed recently. Endothelin-1, which is upregulated in PAH and a known secretagogue of aldosterone, is likely an important determinant of pulmonary aldosterone synthesis.46 In the rat monocrotaline model of PAH, plasma and lung tissue levels of endothelin-1 were increased significantly, and this was associated with a 442% increase in plasma aldosterone levels and a 183% increase in lung tissue aldosterone. Endothelin-1 increased pulmonary endothelial expression of CYP11B2 by 2.73-fold, indicating that endothelin-1 stimulated extra-adrenal pulmonary aldosterone synthesis. Once present, aldosterone induced pulmonary artery endothelial dysfunction by increasing oxidant stress to oxidatively modify the endothelin B receptor and disrupt endothelin B receptor–stimulated activation of the endothelial isoform of nitric oxide synthase (eNOS), resulting in diminished nitric oxide production.37 Hyperaldosteronism was also associated with pulmonary arterial hypertension, pulmonary vascular remodeling, and RV hypertrophy in the rat monocrotaline and Sugen5416/hypoxia models of PAH.37 In these models, treatment with the mineralocorticoid receptor antagonists spironolactone or eplerenone decreased the number of muscularized vessels, increased cross-sectional luminal area, and limited vascular collagen deposition and fibrosis; decreased pulmonary artery systolic pressure; and decreased RV hypertrophy.37 Spironolactone has also been shown to prevent pulmonary vascular remodeling and RV dysfunction in the mouse chronic hypoxia model of PAH.47 Clinically, elevated levels of aldosterone are present in patients with PAH, which indicates that aldosterone has been an underrecognized contributor to the PAH cardiopulmonary vascular phenotype. In a pilot study of pa-

Pulmonary Circulation

tients with unexplained dyspnea who were referred for invasive hemodynamic evaluation, plasma aldosterone levels were found to be increased in patients with PAH, compared with control subjects (5,959.1  2,817.9 vs. 1,200.4  423.9 pg/mL; P < .02), and hyperaldosteronism was associated with a higher mean pulmonary artery pressure (45.5  10.4 vs. 21.4  5.0 mmHg; P < .002) and pulmonary vascular resistance. Furthermore, aldosterone levels correlated positively with pulmonary vascular resistance and inversely with cardiac output in a subset of patients with PAH who had severe disease.38 As suggested by the preclinical studies evaluating the effects of aldosterone on endothelin-1 signaling in PAH, clinical benefit was observed when spironolactone was added to the selective endothelin receptor A antagonist ambrisentan. This was identified in a retrospective study of patients enrolled in the Ambrisentan in Pulmonary Arterial Hypertension, Randomized, Double-Blind, PlaceboControlled, Multicenter, Efficacy Study 1 and 2 (ARIES-1 and ARIES-2). In this study, spironolactone use was identified in 21 (15.9%) and 10 (14.5%) of patients randomized to placebo or to ambrisentan (10 mg/kg), respectively.39,48 These trials demonstrated that patients treated with high-dose ambrisentan, compared with placebo, had a significantly improved 6-minute walk distance at 12 weeks.48 Although patients who had been treated with both ambrisentan and spironolactone were sicker at base-

Volume 4

Number 2

June 2014

| 205

line than patients treated with ambrisentan alone (higher pulmonary vascular resistance and increased levels of plasma B-type natriuretic peptide), patients treated with combination therapy had a 2.3-fold increase in their 6minute walk distance and a significant decrease in plasma B–type natriuretic peptide levels, compared with patients treated with ambrisentan alone.39 Although the rationale for the use of spironolactone among selected patients was also unknown in the ARIES studies, it is recognized that ambrisentan use is associated with peripheral edema, which was reported in 17% of patients treated with ambrisentan and only 11% of placebo-treated patients in the trials.48, 49 When the definition of edema was broadened, it was discovered that more patients with edema were classified as World Health Organization class III/IV than patients without this adverse effect, suggesting that these sicker patients with edema benefited more from coadministration of spironolactone.49 C E L L U L A R AN D M O L E C U L A R M E C H A N I S M S BY WHICH ALD OS TER ON E C O N T R I B U T E S T O P A H Aldosterone modulates the structure and function of the RV-pulmonary vascular circuit in PAH through several complementary cellular and molecular mechanisms, including increasing oxidant stress to decrease bioavailable nitric oxide, promote endothelial dysfunction, and impair vascular reactivity; enhancing inflammation; and stimu-

Figure 3. The effects of aldosterone on vascular function in pulmonary hypertension. Elevated levels of aldosterone have deleterious effects on vascular homeostasis, including increased oxidant stress resulting in a decrease in nitric oxide (NO) and endothelial dysfunction, cell swelling, and inflammation. Aldosterone also contributes to vascular remodeling by increasing cell proliferation and migration leading to vascular hypertrophy and by stimulating extracellular matrix remodeling and fibrosis. ACTH ¼ adrenocorticotropic hormone; eNOS ¼ endothelial isoform of nitric oxide synthase; ENaC ¼ epithelial sodium channel; EPC ¼ endothelial progenitor cell.

206

| Renin-angiotensin-aldosterone system in pulmonary arterial hypertension

lating vascular remodeling and fibrosis (Fig. 3).50,51 In the heart, aldosterone influences cardiomyocyte function through stimulation of the sympathetic nervous system, increased excretion of K+ and Mg+ to render the myocardium proarrhythmogenic, and initiation of cardiomyocyte apoptosis.52,53 Aldosterone-induced endothelial dysfunction has been confirmed by flow-mediated dilation studies in healthy volunteers infused with aldosterone, in patients with primary hyperaldosteronism, and in hypertensive patients with high levels of aldosterone.54,55 In vitro studies suggested that endothelial dysfunction was the result of increased reactive oxygen species generation and decreased nitric oxide bioavailability.56,57 In fact, aldosterone increases vascular endothelial and smooth muscle cell oxidant stress, which has been implicated in the pathogenesis of PAH, through several complementary mechanisms. Aldosterone increases oxidant stress by activating NADPH oxidase, uncoupling of eNOS to generate superoxide in preference to nitric oxide, and decreasing cellular antioxidant capacity by downregulating expression and activity of glucose-6-phosphate dehydrogenase.58-63 In pulmonary artery endothelial cells, aldosterone upregulates the NOX4 subunit of NADPH oxidase to increase enzymatic activity and superoxide generation. This, in turn, decreases levels of bioavailable nitric oxide through a reduction in synthesis owing to oxidative posttranslational modification of a cysteinyl thiol, Cys405, which resides in the eNOSactivating region of the endothelin B receptor.37 Other mechanisms by which aldosterone-stimulated oxidant stress decreases bioavailable nitric oxide include oxidation of nitric oxide to nitrite and nitrate as well as direct reaction with superoxide to form peroxynitrite.37 In vascular smooth muscle cells, the increase in oxidant stress following aldosterone exposure promotes cell proliferation and migration by activating Akt, mitogen-activated protein kinase, and p70S6kinase signaling and further disrupts nitric oxide signaling by oxidatively modifying Cys122 of the β-subunit of soluble guanylyl cyclase to render the enzyme nitric oxide insensitive.62,64,65 Within the vasculature, aldosterone has been associated with an enhanced inflammatory response, which is a recognized component of vascular remodeling in PAH. Aldosterone increases endothelial cell adhesion molecule expression and facilitates activation and vascular infiltration of monocytes, macrophages, and lymphocytes.66,67 Interestingly, circulating monocytic (early) endothelial progenitor cells are affected adversely by aldosterone through a mechanism that involves increased cellular oxidant stress. These early progenitor cells are functionally impaired with limited vascularization and repair capacity.68

Maron and Leopold

In PAH, there is increased pulmonary vascular stiffening that occurs as a result of the remodeling process. Aldosterone contributes to vascular stiffening by regulating the salt and water content of vascular cells as well as stimulating profibrotic remodeling. Using atomic force microscopy, it has been shown that aldosterone increases endothelial cell volume by activation of an amiloride-sensitive Na+/H+ exchanger. Aldosterone also directs insertion of the epithelial sodium channel ENaC, which is expressed by vascular cells, into the endothelial cell membrane to increase cell water retention, swelling, and stiffness and this phenomenon has been shown to contribute to a decrease in endothelial cell nitric oxide release.69-73 Aldosterone is known to stimulate perivascular fibrosis by increasing expression of profibrotic mediators, such as galectin-3; transforming growth factor-β, connective tissue growth factor, and type I and type III collagen; and decreasing expression of the anti-fibrotic peptides and bone morphogenetic protein-4 and brain natriuretic peptide.74,75 In the heart, aldosterone increases cardiac NADPH oxidase activity, which, through endothelial-cardiomyocyte crosstalk, leads to myocardial endothelial dysfunction.76 Aldosterone has also been shown to initiate changes in cardiomyocyte calcium handling and ion channel remodeling by modulating expression of the T-type calcium channel, L-type calcium channel, and ryanodine receptor activities.77-79 Similar to what is observed in the vasculature, aldosterone enhances myocardial stiffness and remodeling by increasing type I and type III collagen deposition, leading to perivascular and interstitial fibrosis.80 Aldosterone also contributes to cardiac matrix remodeling through a mechanism that involves oxidation of Ca2+/calmodulindependent protein kinase II and increased matrix metalloproteinase-9 activity.81 THE RAPE UT IC IN TE RV EN TI ON S T O TARGET MINERALOCORTIC OID RECE PTOR AC TIVA TION At present, there are no specific recommendations for the use of mineralocorticoid receptor antagonists in the treatment of PAH, and the use of diuretics is suggested as background therapy in symptomatic patients with signs of RV failure and volume overload (Table 1).82,83 For patients with a clinical indication for a mineralocorticoid receptor antagonist, treatment with the drugs should be started only if the patient’s serum potassium is less than 5.0 mEq/L and the serum creatinine is less than 2.0 mg/ dL for women and less than 2.5 mg/dL for men. Typically, serum potassium and renal function are measured 2 weeks after the initiation of the drug, with the frequency of measurements increased up to 6 months with longer-

Pulmonary Circulation

Volume 4

Number 2

June 2014

| 207

Table 1. Mineralocorticoid receptor antagonists Variable Potency and selectivity MR IC50, nM PR IC50/EC50, nM AR IC50, nM Excretion Dose Adverse effects

Spironolactone

Eplerenone

BAY 94-8862

24.2 740 77.1 Renal and bile Hypertension: 50–100 mg daily; CHF: 25–200 mg daily Hyperkalemia, gynecomastia

990 31,240 21,240 Renal and GI Hypertension: 50–100 mg daily; CHF: 25–200 mg daily Hyperkalemia, gynecomastia

17.8 10,000 10,000 Renal CHF: 5–10 mg daily Hyperkalemia

Note: Currently available mineralocorticoid receptor (MR) antagonists in the United States include spironolactone and eplerenone; BAY 94-8862 is a nonsteroidal MR that has been trialed recently in patients with congestive heart failure (CHF). There is variability in the binding of these drugs to other sex steroid hormone receptors, including the progesterone receptor (PR) and androgen receptor (AR), which explains some of the observed adverse effects. EC50 ¼ half-maximal effective concentration; GI ¼ gastrointestinal; IC50 ¼ half-maximal inhibitory concentration.

term use.53 Concomitant use of supplements that contain potassium or use of other drugs, such as ACE inhibitors or nonsteroidal anti-inflammatory agents, necessitates increased surveillance of electrolytes and renal function. It should also be noted that, for patients with PAH who receive sildenafil, the addition of a potassium-sparing diuretic increases the area under the curve of the active metabolite N-desmethyl sildenafil by 62% (http://www.drugs .com/sildenafil); however, the clinical implications of this interaction are not known. Spironolactone is a widely available, inexpensive mineralocorticoid receptor antagonist that is nonselective and requires hepatic metabolism to generate its active metabolites. The maximal drug response is observed approximately 48 hours after the first dose. Spironolactone is structurally similar to progesterone, and there is some sex-steroid receptor cross reactivity. This phenomenon accounts for the anti-progesterone and anti-androgen effects observed in some patients treated with the drug. Spironolactone is dosed between 25 and 200 mg daily for congestive heart failure and between 50 and 100 mg daily for hypertension, with dose titration recommended at 4- to 6-week intervals until the desired clinical effect is achieved.53 Optimal dosing of spironolactone in PAH is not known. Eplerenone is a selective mineralocorticoid receptor antagonist that was derived from spironolactone but has limited affinity for the progesterone and androgen receptors and therefore lacks the sex-related adverse effects attributed to spironolactone. Eplerenone is metabolized by cytochrome P450 isoenzyme CYP3A4 and, therefore, is subject to drug-drug interactions with inhibitors of this isoenzyme, including ketoconazole, itraconazole, ribonavir, and

clarithromycin. Eplerenone metabolism is also influenced by inducers of CYP3A4, such as St John’s Wort, which has been shown to decrease levels by 30%. Steady-state drug levels of eplerenone are usually achieved 48 hours after the first dose is administered. When eplerenone treatment is started, it is dosed at 25 mg daily, and the dose is titrated until a clinical response is observed or a maximum dose of 50 mg twice daily is reached.53 There is now a third-generation nonsteroidal mineralocorticoid receptor antagonist, BAY 94-8662, which is reported to have improved selectivity for the mineralocorticoid receptor compared with spironolactone or eplerenone in preclinical studies.84,85 In models of hypertensionmediated left heart failure and renal dysfunction, BAY 94-8862 provided better cardiorenal end-organ protection than other mineralocorticoid receptor antagonists.84 In humans, the plasma half-life of BAY 94-8862 is approximately 2 hours in healthy individuals, and the drug is administered once daily. Recently, results from the first randomized clinical trial of this compound in patients with left heart failure, a reduced ejection fraction, and mild or moderate chronic kidney disease were reported.86 In this pilot study, BAY 94-8862 reduced indices of heart failure to a degree similar to that observed with spironolactone but with fewer episodes of hyperkalemia. This suggests that this agent may broaden the population of patients who can be treated with a mineralocorticoid receptor antagonist.86 CONCLUSION There is clear evidence that the RAAS is activated in PAH and modulates cardiopulmonary remodeling and function, which contributes to progression of the disease. Re-

208

| Renin-angiotensin-aldosterone system in pulmonary arterial hypertension

sults from preclinical and clinical studies indicate that angiotensin II and, in particular, aldosterone, regulate cellular signaling processes that increase proliferation, migration, and vascular hypertrophy as well as initiate cardiovascular fibrosis and interrupt repair processes. Although the prevailing evidence suggests that mineralocorticoid receptor antagonists will benefit patients with PAH, currently available drugs remain to be studied in a systematic randomized clinical trial. Toward this end, a planned clinical trial of early initiation of spironolactone therapy in 70 patients with PAH without clinical evidence of right heart failure has been announced. In this placebo-controlled study, the effect of spironolactone on disease modification will be examined by evaluating the 6-minute walk distance and clinical progression of PAH at 24 weeks.87 In addition to oral agents, recent advances with inhaled therapies, including gene transfer using aerosolized adeno-associated virus, suggest that other methodologies to deliver mineralocorticoid antagonists or selectively decrease pulmonary mineralocorticoid receptor expression may also be available as future pharmacotherapies.88 Source of Support: This work was supported by funds from the National Institutes of Health (NIH)/National Heart, Lung, and Blood Institute (NHLBI) K08111207 and the Klarman Foundation (to BAM) and NIH/NHLBI HL105301 and the Thomas W. Smith MD Foundation (to JAL). Conflict of Interest: B.A.M. receives funding from Gilead Sciences to study experimental pulmonary hypertension.

RE FER ENCES 1. Ling Y, Johnson MK, Kiely DG, et al. Changing demographics, epidemiology, and survival of incident pulmonary arterial hypertension: results from the pulmonary hypertension registry of the United Kingdom and Ireland. Am J Respir Crit Care Med 2012;186:790–796. 2. Humbert M, Sitbon O, Chaouat A, et al. Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern management era. Circulation 2010;122:156–163. 3. Voelkel NF, Cool C. Pathology of pulmonary hypertension. Cardiol Clin 2004;22:343–351, v. 4. McLaughlin VV. Looking to the future: a new decade of pulmonary arterial hypertension therapy. Eur Respir Rev 2011; 20:262–269. 5. Davies RJ, Morrell NW. Molecular mechanisms of pulmonary arterial hypertension: role of mutations in the bone morphogenetic protein type II receptor. Chest 2008;134:1271–1277. 6. Rubin LJ, Simonneau G, Badesch D, et al. The study of risk in pulmonary arterial hypertension. Eur Respir Rev 2012; 21:234–238. 7. Beuschlein F. Regulation of aldosterone secretion: from physiology to disease. Eur J Endocrinol 2013;168:R85–R93. 8. Sulya LL, McCaa CS, Read VH, Bomer D. Uptake of tritiated aldosterone by rat tissues. Nature 1963;200:788–789.

Maron and Leopold

9. Vasan RS, Evans JC, Larson MG, et al. Serum aldosterone and the incidence of hypertension in nonhypertensive persons. N Engl J Med 2004;351:33–41. 10. Lieb W, Xanthakis V, Sullivan LM, et al. Longitudinal tracking of left ventricular mass over the adult life course: clinical correlates of short- and long-term change in the framingham offspring study. Circulation 2009;119:3085– 3092. 11. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 1999;341:709–717. 12. Santos RA, Ferreira AJ, Simoes ESAC. Recent advances in the angiotensin-converting enzyme 2-angiotensin(1–7)-Mas axis. Exp Physiol 2008;93:519–527. 13. Briet M, Schiffrin EL. Aldosterone: effects on the kidney and cardiovascular system. Nat Rev Nephrol 2010;6:261–273. 14. Batt J, Shadly Ahmed S, Correa J, Bain A, Granton J. Skeletal muscle dysfunction in idiopathic pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2014;50:74–86. 15. Oki K, Kopf PG, Campbell WB, et al. Angiotensin II and III metabolism and effects on steroid production in the HAC15 human adrenocortical cell line. Endocrinology 2013;154: 214–221. 16. Touyz RM, Schiffrin EL. Signal transduction mechanisms mediating the physiological and pathophysiological actions of angiotensin II in vascular smooth muscle cells. Pharmacol Rev 2000;52:639–672. 17. Zhang F, Hu Y, Xu Q, Ye S. Different effects of angiotensin II and angiotensin-(1–7) on vascular smooth muscle cell proliferation and migration. PLoS One 2013;5:e12323. 18. Morrell NW, Atochina EN, Morris KG, Danilov SM, Stenmark KR. Angiotensin converting enzyme expression is increased in small pulmonary arteries of rats with hypoxiainduced pulmonary hypertension. J Clin Invest 1995;96: 1823–1833. 19. de Man FS, Tu L, Handoko ML, et al. Dysregulated reninangiotensin-aldosterone system contributes to pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;186: 780–789. 20. Li G, Liu Y, Zhu Y, et al. ACE2 activation confers endothelial protection and attenuates neointimal lesions in prevention of severe pulmonary arterial hypertension in rats. Lung 2013;191:327–336. 21. Morrell NW, Upton PD, Higham MA, Yacoub MH, Polak JM, Wharton J. Angiotensin II stimulates proliferation of human pulmonary artery smooth muscle cells via the AT1 receptor. Chest 1998;114:90S–91S. 22. Morrell NW, Upton PD, Kotecha S, et al. Angiotensin II activates MAPK and stimulates growth of human pulmonary artery smooth muscle via AT1 receptors. Am J Physiol 1999;277:L440–L448. 23. Orte C, Polak JM, Haworth SG, Yacoub MH, Morrell NW. Expression of pulmonary vascular angiotensin-converting enzyme in primary and secondary plexiform pulmonary hypertension. J Pathol 2000;192:379–384. 24. Morrell NW, Danilov SM, Satyan KB, Morris KG, Stenmark KR. Right ventricular angiotensin converting enzyme activity and expression is increased during hypoxic pulmonary hypertension. Cardiovasc Res 1997;34:393–403.

Pulmonary Circulation

25. Morrell NW, Morris KG, Stenmark KR. Role of angiotensinconverting enzyme and angiotensin II in development of hypoxic pulmonary hypertension. Am J Physiol 1995;269: H1186–H1194. 26. Johnson JA, West J, Maynard KB, Hemnes AR. ACE2 improves right ventricular function in a pressure overload model. PLoS One 2011;6:e20828. 27. Shenoy V, Ferreira AJ, Qi Y, et al. The angiotensinconverting enzyme 2/angiogenesis-(1–7)/Mas axis confers cardiopulmonary protection against lung fibrosis and pulmonary hypertension. Am J Respir Crit Care Med 2010;182: 1065–1072. 28. Yamamoto K, Ohishi M, Katsuya T, et al. Deletion of angiotensinconverting enzyme 2 accelerates pressure overload-induced cardiac dysfunction by increasing local angiotensin II. Hypertension 2006;47:718–726. 29. Ferreira AJ, Shenoy V, Yamazato Y, 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. 30. Dempsey EC, Wick MJ, Karoor V, et al. Neprilysin null mice develop exaggerated pulmonary vascular remodeling in response to chronic hypoxia. Am J Pathol 2009;174:782–796. 31. Abraham WT, Raynolds MV, Badesch DB, et al. Angiotensinconverting enzyme DD genotype in patients with primary pulmonary hypertension: increased frequency and association with preserved haemodynamics. J Renin Angiotensin Aldosterone Syst 2003;4:27–30. 32. Rich S, Martinez J, Lam W, Rosen KM. Captopril as treatment for patients with pulmonary hypertension: problem of variability in assessing chronic drug treatment. Br Heart J 1982;48:272–277. 33. Leier CV, Bambach D, Nelson S, et al. Captopril in primary pulmonary hypertension. Circulation 1983;67:155–161. 34. Ikram H, Maslowski AH, Nicholls MG, Espiner EA, Hull FT. Haemodynamic and hormonal effects of captopril in primary pulmonary hypertension. Br Heart J 1982;48:541–545. 35. Rossi GP. Aldosterone breakthrough during RAS blockade: a role for endothelins and their antagonists? Curr Hypertens Rep 2006;8:262–268. 36. Jorde UP, Vittorio T, Katz SD, Colombo PC, Latif F, Le Jemtel TH. Elevated plasma aldosterone levels despite complete inhibition of the vascular angiotensin-converting enzyme in chronic heart failure. Circulation 2002;106:1055–1057. 37. Maron BA, Zhang YY, White K, et al. Aldosterone inactivates the endothelin-B receptor via a cysteinyl thiol redox switch to decrease pulmonary endothelial nitric oxide levels and modulate pulmonary arterial hypertension. Circulation 2012;126:963–974. 38. Maron BA, Opotowsky AR, Landzberg MJ, Loscalzo J, Waxman AB, Leopold JA. Plasma aldosterone levels are elevated in patients with pulmonary arterial hypertension in the absence of left ventricular heart failure: a pilot study. Eur J Heart Fail 2013;15:277–283. 39. Maron BA, Waxman AB, Opotowsky AR, et al. Effectiveness of spironolactone plus ambrisentan for treatment of pulmonary arterial hypertension (from the [ARIES] study 1 and 2 trials). Am J Cardiol 2013;112:720–725. 40. Funder JW. Cardiac synthesis of aldosterone: going, going, gone..? Endocrinology 2004;145:4793–4795.

Volume 4

Number 2

June 2014

| 209

41. Gomez-Sanchez EP, Ahmad N, Romero DG, GomezSanchez CE. Origin of aldosterone in the rat heart. Endocrinology 2004;145:4796–4802. 42. Ye P, Kenyon CJ, MacKenzie SM, et al. The aldosterone synthase (CYP11B2) and 11beta-hydroxylase (CYP11B1) genes are not expressed in the rat heart. Endocrinology 2005;146: 5287–5293. 43. Takeda Y, Miyamori I, Yoneda T, et al. Regulation of aldosterone synthase in human vascular endothelial cells by angiotensin II and adrenocorticotropin. J Clin Endocrinol Metab 1996;81:2797–2800. 44. Ahmad N, Romero DG, Gomez-Sanchez EP, Gomez-Sanchez CE. Do human vascular endothelial cells produce aldosterone? Endocrinology 2004;145:3626–3629. 45. Takeda R, Hatakeyama H, Takeda Y, et al. Aldosterone biosynthesis and action in vascular cells. Steroids 1995;60:120– 124. 46. Belloni AS, Rossi GP, Andreis PG, et al. Endothelin adrenocortical secretagogue effect is mediated by the B receptor in rats. Hypertension 1996;27:1153–1159. 47. Preston IR, Sagliani KD, Warburton RR, Hill NS, Fanburg BL, Jaffe IZ. Mineralocorticoid receptor antagonism attenuates experimental pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2013;304:L678–L688. 48. Galie` N, Olschewski H, Oudiz RJ, et al. Ambrisentan for the treatment of pulmonary arterial hypertension: results of the ambrisentan in pulmonary arterial hypertension, randomized, double-blind, placebo-controlled, multicenter, efficacy (ARIES) study 1 and 2. Circulation 2008;117:3010– 3019. 49. Shapiro S, Pollock DM, Gillies H, et al. Frequency of edema in patients with pulmonary arterial hypertension receiving ambrisentan. Am J Cardiol 2012;110:1373–1377. 50. Briet M, Schiffrin EL. Vascular actions of aldosterone. J Vasc Res 2013;50:89–99. 51. Nguyen Dinh Cat A, Jaisser F. Extrarenal effects of aldosterone. Curr Opin Nephrol Hypertens 2012;21:147–156. 52. Maron BA, Leopold JA. Mineralocorticoid receptor antagonists and endothelial function. Curr Opin Investig Drugs 2008;9:963–969. 53. Maron BA, Leopold JA. Aldosterone receptor antagonists: effective but often forgotten. Circulation 2010;121:934–939. 54. Farquharson CA, Struthers AD. Aldosterone induces acute endothelial dysfunction in vivo in humans: evidence for an aldosterone-induced vasculopathy. Clin Sci (Lond) 2002; 103:425–431. 55. Nishizaka MK, Zaman MA, Green SA, Renfroe KY, Calhoun DA. Impaired endothelium-dependent flow-mediated vasodilation in hypertensive subjects with hyperaldosteronism. Circulation 2004;109:2857–2861. 56. Nagata D, Takahashi M, Sawai K, et al. Molecular mechanism of the inhibitory effect of aldosterone on endothelial NO synthase activity. Hypertension 2006;48:165–171. 57. Griol-Charhbili V, Fassot C, Messaoudi S, Perret C, Agrapart V, Jaisser F. Epidermal growth factor receptor mediates the vascular dysfunction but not the remodeling induced by aldosterone/salt. Hypertension 2011;57:238–244. 58. Leopold JA, Dam A, Maron BA, et al. Aldosterone impairs vascular reactivity by decreasing glucose-6-phosphate dehydrogenase activity. Nat Med 2007;13:189–197.

210

| Renin-angiotensin-aldosterone system in pulmonary arterial hypertension

59. Wilkinson-Berka JL, Tan G, Jaworski K, Miller AG. Identification of a retinal aldosterone system and the protective effects of mineralocorticoid receptor antagonism on retinal vascular pathology. Circ Res 2009;104:124–133. 60. Pessoa BS, Peixoto EB, Papadimitriou A, Lopes de Faria JM, Lopes de Faria JB. Spironolactone improves nephropathy by enhancing glucose-6-phosphate dehydrogenase activity and reducing oxidative stress in diabetic hypertensive rat. J Renin Angiotensin Aldosterone Syst 2012;13:56–66. 61. Bowers R, Cool C, Murphy RC, et al. Oxidative stress in severe pulmonary hypertension. Am J Respir Crit Care Med 2004;169:764–769. 62. Maron BA, Zhang YY, Handy DE, et al. Aldosterone increases oxidant stress to impair guanylyl cyclase activity by cysteinyl thiol oxidation in vascular smooth muscle cells. J Biol Chem 2009;284:7665–7672. 63. Shi G, Fu Y, Jiang W, et al. Activation of Src-ATF1 pathway is involved in upregulation of Nox1, a catalytic subunit of NADPH oxidase, by aldosterone. Endocr J 2011;58:491–499. 64. Fan C, Kawai Y, Inaba S, et al. Synergy of aldosterone and high salt induces vascular smooth muscle hypertrophy through up-regulation of NOX1. J Steroid Biochem Mol Biol 2008;111:29–36. 65. Cascella T, Radhakrishnan Y, Maile LA, et al. Aldosterone enhances IGF-I-mediated signaling and biological function in vascular smooth muscle cells. Endocrinology 2010;151: 5851–5864. 66. Kasal DA, Barhoumi T, Li MW, et al. T regulatory lymphocytes prevent aldosterone-induced vascular injury. Hypertension 2012;59:324–330. 67. Caprio M, Newfell BG, la Sala A, et al. Functional mineralocorticoid receptors in human vascular endothelial cells regulate intercellular adhesion molecule-1 expression and promote leukocyte adhesion. Circ Res 2008;102:1359–1367. 68. Thum T, Schmitter K, Fleissner F, et al. Impairment of endothelial progenitor cell function and vascularization capacity by aldosterone in mice and humans. Eur Heart J 2011; 32:1275–1286. 69. Oberleithner H, Ludwig T, Riethmuller C, et al. Human endothelium: target for aldosterone. Hypertension 2004;43: 952–956. 70. Oberleithner H, Riethmuller C, Ludwig T, Hausberg M, Schillers H. Aldosterone remodels human endothelium. Acta Physiol (Oxf) 2006;187:305–312. 71. Kusche-Vihrog K, Callies C, Fels J, Oberleithner H. The epithelial sodium channel (ENaC): mediator of the aldosterone response in the vascular endothelium? Steroids 2010; 75:544–549. 72. Kusche-Vihrog K, Sobczak K, Bangel N, et al. Aldosterone and amiloride alter ENaC abundance in vascular endothelium. Pfluegers Arch 2008;455:849–857. 73. Fels J, Oberleithner H, Kusche-Vihrog K. Menage´ a` trois: aldosterone, sodium and nitric oxide in vascular endothelium. Biochim Biophys Acta 2010;1802:1193–1202. 74. Azibani F, Benard L, Schlossarek S, et al. Aldosterone inhibits antifibrotic factors in mouse hypertensive heart. Hypertension 2012;59:1179–1187. 75. Nakano S, Kobayashi N, Yoshida K, Ohno T, Matsuoka H. Cardioprotective mechanisms of spironolactone associated

76.

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

Maron and Leopold

with the angiotensin-converting enzyme/epidermal growth factor receptor/extracellular signal-regulated kinases, NAD (P)H oxidase/lectin-like oxidized low-density lipoprotein receptor-1, and Rho-kinase pathways in aldosterone/saltinduced hypertensive rats. Hypertens Res 2005;28:925–936. Favre J, Gao J, Zhang AD, et al. Coronary endothelial dysfunction after cardiomyocyte-specific mineralocorticoid receptor overexpression. Am J Physiol Heart Circ Physiol 2011; 300:H2035–H2043. Ouvrard-Pascaud A, Sainte-Marie Y, Benitah JP, et al. Conditional mineralocorticoid receptor expression in the heart leads to life-threatening arrhythmias. Circulation 2005;111: 3025–3033. Perrier R, Richard S, Sainte-Marie Y, et al. A direct relationship between plasma aldosterone and cardiac L-type Ca2+ current in mice. J Physiol 2005;569:153–162. Gomez AM, Rueda A, Sainte-Marie Y, et al. Mineralocorticoid modulation of cardiac ryanodine receptor activity is associated with downregulation of FK506-binding proteins. Circulation 2009;119:2179–2187. Weber KT, Brilla CG, Janicki JS. Myocardial fibrosis: functional significance and regulatory factors. Cardiovasc Res 1993;27:341–348. He BJ, Joiner ML, Singh MV, et al. Oxidation of CaMKII determines the cardiotoxic effects of aldosterone. Nat Med 2011;17:1610–1618. McLaughlin VV, Archer SL, Badesch DB, 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 2009;119:2250–2294. Galie` N, Hoeper MM, Humbert M, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J 2009;34:1219–1263. Delbeck M, Kretschmer A, Kast R, et al. Cardiorenal protection by BAY 94-8862, a novel non-steroidal mineralocorticoid receptor antagonist in a preclinical model of hypertension and diastolic heart failure. Eur Heart J 2012; 33:772–773. Barfacker L, Kuhl A, Hillisch A, et al. Discovery of BAY 94-8862: a nonsteroidal antagonist of the mineralocorticoid receptor for the treatment of cardiorenal diseases. ChemMedChem 2012;7:1385–1403. Pitt B, Kober L, Ponikowski P, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J 2013;34:2453–2463. Elinoff JM, Rame JE, Forfia PR, et al. A pilot study of the effect of spironolactone therapy on exercise capacity and endothelial dysfunction in pulmonary arterial hypertension: study protocol for a randomized controlled trial. Trials 2013; 14:91. Hadri L, Kratlian RG, Benard L, et al. Therapeutic efficacy of AAV1.SERCA2a in monocrotaline-induced pulmonary arterial hypertension. Circulation 2013;128:512–523.

The role of the renin-angiotensin-aldosterone system in the pathobiology of pulmonary arterial hypertension (2013 Grover Conference series).

Pulmonary arterial hypertension (PAH) is associated with aberrant pulmonary vascular remodeling that leads to increased pulmonary artery pressure, pul...
2MB Sizes 2 Downloads 3 Views