Aldosterone exerts direct effects on the vascular system by inducing oxidative stress, inflammation, hypertrophic remodeling, fibrosis, and endothelial dysfunction. Aldosterone exerts its effects through genomic and nongenomic pathways in a mineralocorticoid receptor (MR)-dependent or independent manner. Other aldosterone receptors such as GPR30 have been identified. A tight relation exists between the aldosterone and angiotensin II pathways, as well as with the endothelin-1 system. There is a correlation between plasma levels of aldosterone and cardiovascular risk. Recently, an increasing body of evidence has underlined the importance of aldosterone in cardiovascular complications associated with the metabolic syndrome, such as arterial remodeling and endothelial dysfunction. Blockade of MR is an increasingly used evidence-based therapy for many forms of cardiovascular disease, including hypertension, heart failure, chronic kidney disease, and diabetes mellitus.

Aldosterone is a steroid hormone synthesized by the adrenal glomerulosa in response to various stimuli including angiotensin II, high potassium levels, and adrenocorticotropic hormone. Aldosterone classically acts on sodium reabsorption at the level of the distal nephron through its interaction with the mineralocorticoid receptor (MR) and activation of the apical epithelial sodium channel [1]. The link between aldosterone and hypertension was clearly established many years ago by Conn [2] in secondary forms of hypertension but is also present in essential hypertension, since mild elevations of aldosterone levels have been shown in patients with essential hypertension [3].

Beside its effect on sodium reabsorption and hypertension, aldosterone is involved in various biological processes at the level of the vasculature, where it induces inflammation, oxidative stress, endothelial dysfunction, and vascular remodeling. In this review we will detail the molecular mechanisms involved in the vascular effects of aldosterone, the cross talk with angiotensin II and endothelin-1 (ET-1) pathways, and the genomic versus nongenomic effects of aldosterone (fig. 1).

Fig. 1

Effects of aldosterone on the vascular system. Aldosterone is mainly synthesized by the adrenal gland in response to angiotensin II, potassium (K+), and adrenocorticotropic hormone (ACTH). At the level of the vessel, aldosterone induces inflammation and oxidative stress, hypertrophic remodeling, and endothelial dysfunction. Aldosterone acts through its effect on MR and another type of receptor, the G protein-coupled receptor 30 (GPR30), via genomic and non genomic pathways. Aldosterone induces endothelial dysfunction through a mechanism involving reduction of nitrite oxide bioavailability. At the level of endothelial cells, aldosterone exerts a direct effect on glucose 6 dehydrogenase (G6PD), and epidermal growth factor (EGFR), increasing their volume and stiffness. Finally, aldosterone acts on endothelial progenitor cells (EPCs) reducing vascular homing, migration, differentiation, and proliferation. T regulatory cells reduce the vascular effects of aldosterone. Perivascular fat synthesizes aldosterone and influences the vascular wall in an MR-dependent manner.

Fig. 1

Effects of aldosterone on the vascular system. Aldosterone is mainly synthesized by the adrenal gland in response to angiotensin II, potassium (K+), and adrenocorticotropic hormone (ACTH). At the level of the vessel, aldosterone induces inflammation and oxidative stress, hypertrophic remodeling, and endothelial dysfunction. Aldosterone acts through its effect on MR and another type of receptor, the G protein-coupled receptor 30 (GPR30), via genomic and non genomic pathways. Aldosterone induces endothelial dysfunction through a mechanism involving reduction of nitrite oxide bioavailability. At the level of endothelial cells, aldosterone exerts a direct effect on glucose 6 dehydrogenase (G6PD), and epidermal growth factor (EGFR), increasing their volume and stiffness. Finally, aldosterone acts on endothelial progenitor cells (EPCs) reducing vascular homing, migration, differentiation, and proliferation. T regulatory cells reduce the vascular effects of aldosterone. Perivascular fat synthesizes aldosterone and influences the vascular wall in an MR-dependent manner.

Close modal

Role of Aldosterone in Cardiovascular Remodeling Induced by Angiotensin II

Aldosterone induces vascular fibrosis and hypertrophic remodeling particularly in the presence of salt [4,5]. In animal models of hypertension, such as spontaneously hypertensive rats, vascular fibrosis has been prevented by administration of spironolactone, an MR antagonist, independently of blood pressure reduction [6]. Cardiovascular remodeling effects usually attributed to angiotensin II are significantly reduced by the use of MR blockers. Indeed, in Sprague-Dawley rats infused with angiotensin II, spironolactone reduced systolic blood pressure and blunted the hypertrophic remodeling characterized by an increase in the wall-to-lumen ratio in small resistance arteries [7].

Aldosterone Signaling Pathways in Vascular Smooth Muscle Cells

In vitro, aldosterone (10–7m) exerts a direct effect on signaling in vascular smooth muscle cells (VSMCs) involving extracellular signal-regulated kinase (ERK) [8], mitogen-activated protein kinase (MAPK) [8], the tyrosine kinase c-Src [9], and c-jun N terminal kinase (JNK) and participates in epidermal growth factor receptor (EGFR) transactivation [8]. Aldosterone also significantly increased phosphorylation of myosin phosphate target subunit-1 (MYPY1), a marker of Rho-kinase activity and the amount of GTP-Rho. As a consequence, aldosterone induced VSMC stress fiber formation and migration. All of these effects were blocked by eplerenone, a selective MR blocker, and Y27632, a specific Rho-kinase inhibitor [10].

Effect of Aldosterone on Arterial Mechanical Properties

Aldosterone infusion (1 µg/h) into uninephrectomized Sprague-Dawley rats receiving a high-salt diet increased large artery stiffness with an increase in pulse pressure, and the carotid elastic modulus. These functional changes were associated with increased aortic fibronectin expression with no change in elastin or collagen density. This phenotype was completely blunted by eplerenone, an MR antagonist [11]. In hypertensive patients, an inverse correlation was found between the plasma aldosterone concentration (PAC) (mean ± SD: 13.6 ± 4.5 ng/100 ml in normotensive patients and 11.5 ± 6.6 in hypertensive patients) and large vessel compliance, independently of age and blood pressure [12]. In addition, a polymorphism of CYP11B2 was associated with higher aortic stiffness in hypertensive patients [13].

Role of Salt in Aldosterone-Induced Vascular Disease

The effects of aldosterone on the cardiovascular system are dependent on salt co-administration or pre-existing vascular injury. Indeed, in uninephrectomized rats fed a low-salt diet, aldosterone infusion (0.75 µg/h) did not induce cardiac fibrosis [14]. Vascular function and remodeling in patients with Gitelman or Bartter syndrome characterized by a renal loss of sodium were similar to those measured in normotensive controls despite a high level of aldosterone associated with these diseases (PAC, mean ± SD: 0.18 ± 0.02 nm in controls and 0.77 ± 0.09 nm in patients with Gitelman syndrome) [15]. These studies underlined the importance of salt in the sensitization of vascular tissues to aldosterone effects.

It is well demonstrated now that aldosterone induces oxidative stress and inflammation in the vascular wall, in endothelial cells, and in VSMCs [16]. Part of the effects of angiotensin II on the vasculature is due to aldosterone since selective aldosterone blockade by an MR antagonist blunted perivascular inflammation characterized by macrophage infiltration, and reduced the expression of inflammatory markers, cyclooxygenase-2 and osteopontin, in coronary arteries of rats infused with angiotensin II [17].

In rats, aldosterone and salt infusion induced an increase in NADPH oxidase subunit expression in an MR-dependent manner. Aldosterone infusion in Nox2-deficient mice was not associated with an increase in NADPH oxidase activity as was observed in wild-type controls, which underlines the role of Nox2 in aldosterone-induced oxidative stress in the vascular system [18].

Signaling Pathways Involved in Aldosterone-Induced Oxidative Stress

In vitro, in VSMCs, aldosterone (10–7m) induced a time-dependent increase in NADPH-oxidase activity through c-Src activation since this effect was significantly reduced in VSMCs from c-Src+/– mice [19]. Recently, Callera et al. [20] demonstrated that aldosterone-induced c-Src activation involved lipid rafts/caveolae via the platelet-derived growth factor receptor. In endothelial cells, aldosterone (10–8–10–6m) induced superoxide generation through Rac1 activation of NADPH oxidase in an MR-dependent manner [21].

Impact of Aldosterone-Induced Oxidative Stress on Vascular Remodeling

Increased oxidative stress secondary to aldosterone infusion has consequences on vascular remodeling. Indeed, in Wistar rats receiving aldosterone infusion (0.75 µg/h) and 1% NaCl in the drinking water, administration of apocynin, an NADPH oxidase inhibitor, prevented hypertrophic remodeling and vascular fibrosis and suppressed the expression of transforming growth factor-β1, type I and III collagen, and monocyte chemoattractant protein-1 [16]. In addition, cardiac fibrosis induced by aldosterone in wild-type mice was blunted in Nox2-deficient mice [18]. Oxidative stress induced by aldosterone also upregulated the expression of oxidized low-density lipoprotein receptor-1 (Olr-1). Olr-1 is a member of the C-type lectin family known to acts as a cell-surface receptor for oxidized lipoprotein. This observation provides a link between aldosterone and the development of atherosclerosis [16].

Aldosterone Effect of Vascular Inflammation

Aldosterone infusion (600 µg/kg/day) into experimental animals resulted in vascular infiltration of monocytes, macrophages, and lymphocytes [22]. In human coronary endothelial cells, aldosterone (1–100 nm) promoted ICAM-1 transcription and leukocyte adhesion in an MR-dependent manner [23]. In response to aldosterone (0.75 µg/h), vascular inflammation is partly dependent on oxidative stress [24]. Interestingly, increased oxidative stress and vascular inflammation are the first effects of aldosterone in vessels. Indeed, in uninephrectomized rats receiving aldosterone and salt 1%, a vascular increase in oxidative stress and inflammation, characterized by expression of intracellular adhesion modecule-1, monocyte chemoattractant protein-1, and tumor necrosis factor-α, preceded coronary artery fibrosis [24], which could be prevented by treatment with pyrrolidine dithiocarbamate or N-acetylcysteine [24].

Recently, we showed that adaptive immunity plays a pivotal role in aldosterone-induced vascular injury. In mice, adoptive transfer of CD4+CD25+ (T regulatory) lymphocytes prevented the vascular effects of aldosterone infusion characterized by endothelial dysfunction, hypertrophic remodeling, aortic superoxide production, and inflammation [22].

Aldosterone affects endothelial function in humans with primary hyperaldosteronism and in various animal models of hypertension through mechanisms involving inflammation and oxidative stress and through direct effects on the electrolyte composition of endothelial cells and on endothelial progenitor cell (EPC) function.

Effect of Aldosterone on Endothelial Function

In a clinical study, endothelial function, evaluated by measurement of flow-mediated dilation, was significantly impaired in 36 patients with primary hyperaldosteronism (PAC, mean ± SD: 20.4 ± 12.8 ng/dl) compared to 44 patients with essential hypertension (9.5 ± 6.6 ng/dl). A 3-month treatment with spironolactone was associated with a significant improvement in endothelial function [25].

Aldosterone infusion in various models of mice [22] and rats [26] induced a significant decrease in endothelium-dependent relaxation in large [26] and small arteries [22]. In addition, part of the effects of angiotensin II on endothelial function can be attributed to aldosterone. Indeed, in Sprague-Dawley rats infused with angiotensin II, treatment with the MR antagonist spironolactone partially reversed endothelial dysfunction and abolished angiotensin II-increased oxidative stress [7].

Mechanisms Involved in Aldosterone-Induced Endothelial Dysfunction

Aldosterone induces an increase in oxidative stress in VSMCs, which may play a role in endothelial dysfunction through a reduction in nitric oxide (NO) bioavailability. Generation of oxygen species and the activity of NADPH oxidase were increased in mice [22] and rats [16] infused with aldosterone in an MR-dependent manner. In patients with chronic heart failure, treatment with spironolactone improved endothelial function measured at the level of the forearm by plethysmography, in association with increased NO bioavailability [27]. Interestingly, in a model of mice with cardiomyocyte-specific overexpression of aldosterone synthase (1.7-fold increase in PAC in the heart, no change in plasma), despite the absence of structural or functional alterations at the level of the heart, impairment in coronary endothelial-dependent relaxation was observed [28]. The alteration of endothelial function was mostly due to a decrease in NO availability [28]. In this model, coronary endothelial dysfunction was associated with an increase in cardiac reactive oxygen species production and Nox expression and activity. Treatment with the antioxidant vitamins E and C and by the Nox inhibitor apocynin (also considered a superoxide scavenger) prevented endothelial dysfunction in this model [29]. Another mechanism of aldosterone- induced endothelial dysfunction implicates glucose-6-phosphate dehydrogenase (G6PD) activity, a critical regulator of the intracellular redox state. Indeed, it has been shown that aldosterone (10–9–10–7m) decreases endothelial G6PD expression and activity in vitro. In vivo, aldosterone infusion also decreased vascular expression of G6PD in an MR-dependent manner. Vascular gene transfer of G6PD restored endothelial dysfunction induced by aldosterone [30]. Altogether, these studies underline the deleterious effect of aldosterone on endothelial function through a mechanism involving oxidative stress generation.

Recently, Griol-Charhbili et al. [31] underlined the role of EGFR in aldosterone-mediated endothelial dysfunction. In the waved 2 mouse, a model characterized by a mutation in the EGFR gene altering the receptor kinase domain and reducing the functional activity of EGFR, functional consequences of aldosterone infusion (60 µg/kg/day) on endothelial function were attenuated [31].

The participation of prostacyclin in aldosterone-induced endothelial dysfunction at the level of the aorta was shown in normotensive (Wistar Kyoto) and hypertensive (spontaneously hypertensive) rats. More precisely, aldosterone induced endothelial dysfunction through cyclooxygenase-2 activation in normotensive and hypertensive conditions. Prostacyclin was the main factor implicated in endothelial dysfunction in hypertensive rats [26].

Effect of Aldosterone on Mechanical Properties of Endothelial Cells

Using atomic force microscopy, Oberleithner’s group have demonstrated that aldosterone exerts a direct effect on endothelial cells, increasing the volume through an MR-dependent mechanism that implicated the amiloride-sensitive sodium-proton exchanger [32,33], resulting in increased stiffness of endothelial cells [34], MR-dependent cell growth [35], and reduced NO release by endothelial cells [36]. Altogether, these results show that aldosterone exerts a direct effect on the electrolyte composition of endothelial cells that influences their mechanical and functional properties.

Effect of Aldosterone on EPCs

Recently, Thum et al. [37] showed that treatment of EPCs with aldosterone induced nuclear translocation of MR and impaired EPC functions such as differentiation, migration, and proliferation. In mice, aldosterone infusion impaired EPC homing to vascular structures and vascularization capacity in an MR-dependent manner [37]. Interestingly, they also showed that in patients with primary hyperaldosteronism, EPCs had a reduced migratory potential. Treatment with spironolactone for 4–6 weeks significantly improved EPC migratory function [37]. In another clinical study comparing 113 patients with primary hyperaldosteronism (PAC, mean ± SD: 47.4 ± 36.8 ng/dl) to 55 patients with essential hypertension (PAC, mean ± SD: 24.2.4 ± 16.4 ng/dl), a reduction of circulating EPCs and endothelial colony-forming units was seen in patients with primary hyperaldosteronism. The preoperative number of EPCs predicted the curability of hypertension after adrenalectomy [38].

An increasing body of evidence suggests that cross talk between aldosterone and angiotensin II pathways takes place in VSMCs. Aldosterone and angiotensin II exert a synergistic effect on proliferation, senescence, and migration.

Effect of Angiotensin II on Aldosterone Secretion

Aldosterone increases the expression of angiotensin receptors in vivo [39,40] and in vitro [40]. On the contrary, angiotensin II stimulates aldosterone synthesis by the adrenal gland. There is also controversial data suggesting the existence of local synthesis of aldosterone in the vasculature after stimulation by angiotensin II. Hatakeyama et al. [41] showed that VSMCs and endothelial cells express CYP11B2, the key enzyme involved in aldosterone synthesis, as well as MR. In vitro, angiotensin II administration to VSMCs had a proliferative effect which was blunted by ZK 91587, an MR antagonist, suggesting local synthesis of aldosterone by VSMCs [41]. Direct evidence of aldosterone synthesis in rat mesenteric arteries has been shown using a reverse-phase high-performance liquid chromatographic system [42]. More recent studies have failed to show aldosterone synthase mRNA and aldosterone synthesis in human endothelial cells in response to angiotensin II, ACTH, and potassium, and in the presence of deoxycorticosterone [43]. In adrenalectomized rats, Fiebeler et al. [44] showed a strong reduction in cardiac aldosterone suggesting that more than 99% of cardiac aldosterone was not produced locally. Thus, local synthesis of aldosterone in the vascular system has not been demonstrated in a rigorous way, or the amount produced is very low and its physiological local effect remains to be demonstrated.

Aldosterone and Angiotensin II Cross Talk on VSMC Function

Aldosterone exerts a synergistic effect with angiotensin II on VSMC proliferation [45,46]. Low doses of aldosterone (10–12m) and angiotensin II (10–10m) alone had no effect on VSMC proliferation, whereas the combination significantly increased DNA synthesis. This synergistic effect on proliferation implicates the angiotensin II type 1 receptor, MR, MAPK/ERK1/2, and EGF receptor tyrosine kinase, since proliferation was blunted by specific inhibitors of each of these components. Interestingly, treatment of VSMCs with aldosterone and angiotensin II at a low dose induced two peaks of ERK phosphorylation: the first one within 5 min, compatible with a nongenomic action, and the second one between 2 and 4 h later, compatible with a genomic pathway. The first peak of ERK phosphorylation was angiotensin II type 1 receptor and EGF receptor dependent, whereas the second one also implicated MR. The late peak of ERK phosphorylation was dependent on Ki-ras2A activity.

Min et al. [47] also showed that angiotensin II and aldosterone exert a synergistic effect on senescence through a mechanism involving oxidative stress and Ki-ras2A. Low doses of angiotensin II (100 pm) and aldosterone (1 pm) separately had no effect on VSMC senescence, whereas in combination an increase in SA-β-gal-positive cells and expressions of p21, p53, p16, and p27, which are markers of senescence, were observed. This effect was attenuated by knocking down the expression of Ki-ras2A and by the inhibition of oxidative stress by an antioxidant, N-acetyl-l-cysteine (NAC), or superoxide dismutase.

Synergistic effects of aldosterone and angiotensin II were also involved in migration [48]. Montezano et al. [48] showed that VSMC migration was stimulated by co-administration of a low dose of aldosterone (0.1 nm) and angiotensin II (0.1 nm), whereas angiotensin II or aldosterone alone at this low dose had no effect. This process implicated c-Src pathways that regulated RhoA/Rhokinase, and was dependent on transactivation of EGFR and PDGFR.

Recently, Batenbourg et al. [49] demonstrated that aldosterone potentiated the constrictor effect of angiotensin II on human coronary microarteries at nanomolar levels depending on G protein-coupled receptor 30 (GPR30) and EGFR transactivation. Interestingly, at micromolar levels, aldosterone abolished the angiotensin II-induced contraction through a mechanism involving endothelial NOS phosphorylation [49].

Mazak et al. [8] showed that aldosterone (10–7m) and angiotensin II (10–7m) had synergistic effects on ERK1/2 and JNK phosphorylation, which was dependent on ROS generation.

In VSMCs, we have provided direct evidence of the involvement of angiotensin II type 1 receptor in aldosterone signaling pathways. Aldosterone-induced activation of ERK1/2, JNK, and NF-ĸB was reduced in VSMCs from AT1a-deficient mice or in VSMCs which had undergone knock-down for AT1a receptor [50].

These experimental data emphasize the cross talk between aldosterone and angiotensin II pathways, and provide physiological arguments for the use of MR antagonists in addition to angiotensin converting enzyme inhibitors or angiotensin II receptor blockers in hypertensive patients.

An increase in ET-1 expression has been shown in the deoxycorticosterone (DOCA)-salt hypertension rat model [51,52]. It is well established that ET-1 is implicated in the vascular remodeling process. Endothelium-restricted overexpression of human prepro-ET-1 mouse exhibited inward hypertrophic remodeling of resistance arteries, and vascular inflammation and endothelial dysfunction, in the absence of blood pressure rise [53]. Accordingly, ETA receptor antagonists prevented blood pressure rise and vascular remodeling in aldosterone-infused rats [4,5] and in DOCA-salt-treated spontaneously hypertensive rats [54,55]. An ETA blocker also blunted aldosterone-induced vascular inflammation [56]. These studies, however, do not establish if there is a direct cross talk between ET-1 and aldosterone or a physiological antagonism induced by the ETA blocker. The report from Stow et al. [57] that aldosterone modulates steroid receptor binding to the ET-1 gene (edn1) are in agreement with a direct link. These experiments underline the importance of the endothelin system in aldosterone-induced vascular injury.

Aldosterone exerts some effects on the vascular system with a time course which is not compatible with transcriptional mechanisms and may thus involve nongenomic mechanisms. These short-term actions involve variations in intracellular electrolyte concentration, the activity of the sodium-proton exchanger, inositol triphosphate (IP3) production, and vascular reactivity. Genomic effects are all mediated via the activation of the MR whereas nongenomic effects may be MR dependent or MR independent involving other receptors such as GPR30 (fig. 2).

Fig. 2

Genomic and nongenomic action of aldosterone. Aldosterone diffuses across the cytoplasmic membrane of vascular smooth cells and binds to the MR, inducing dimerization and translocation to the nucleus, where the complex binds to the glucocorticoid response element, and induces the expression of Sgk-1. Sgk-1 increases Ki-Ras2A, which in turn activates ERK1/2 (extracellular stimulus regulating kinase). Nongenomic pathways are MR dependent or independent. Aldosterone, in an MR-dependent pathway, induces rapid activation of JNK, p38 MAPK, and ERK1/2 through the transactivation of EGFR and c-Src (a nonreceptor tyrosine kinase). c-Src induces ROS (reactive oxygen species) production, which activates ROCK and MAPK. Aldosterone, through its interaction with GPR30, activates ERK1/2 nongenomically without intervention of MR.

Fig. 2

Genomic and nongenomic action of aldosterone. Aldosterone diffuses across the cytoplasmic membrane of vascular smooth cells and binds to the MR, inducing dimerization and translocation to the nucleus, where the complex binds to the glucocorticoid response element, and induces the expression of Sgk-1. Sgk-1 increases Ki-Ras2A, which in turn activates ERK1/2 (extracellular stimulus regulating kinase). Nongenomic pathways are MR dependent or independent. Aldosterone, in an MR-dependent pathway, induces rapid activation of JNK, p38 MAPK, and ERK1/2 through the transactivation of EGFR and c-Src (a nonreceptor tyrosine kinase). c-Src induces ROS (reactive oxygen species) production, which activates ROCK and MAPK. Aldosterone, through its interaction with GPR30, activates ERK1/2 nongenomically without intervention of MR.

Close modal

Nongenomic Effects of Aldosterone on the Vascular Tone

In clinical studies, aldosterone injection in healthy subjects was associated with an increase in vascular resistance within the first 10 min [58,59], with a rapid decrease in forearm blood flow between 4 and 8 min [60]. A rapid decrease in forearm blood flow was also observed in treated chronic heart failure after aldosterone infusion [61]. In healthy subjects, aldosterone infusion (0.5 mg) did not significantly affect renal plasma flow, whereas co-infusion with N(G)monomethyl-l-arginine (l-NMMA) that inhibits NO synthesis unmasked the effect of aldosterone on the renal circulation and induced a decrease in renal plasma flow and an increase in renal vascular resistance which was significantly higher than with l-NMMA alone (1,588 ± 237 vs. 614 ± 240 dyn·s·cm–5; p = 0.014, respectively) within 30 min after injection. This is compatible with a nongenomic effect [62]. In accordance with the observations made in humans, Liu et al. [63] described a rapid and opposite effect of aldosterone on the vascular reactivity of aortic rings isolated from Wistar rats, which could be dependent or not on the presence of an intact endothelium. On aortic rings with intact endothelium, aldosterone (0.01 nm) induced a 25% reduction of phenylephrine-mediated constriction compared to controls in 10 min. This effect was MR dependent since it was attenuated by spironolactone. On the contrary, in denuded aortic rings, aldosterone induced a monophasic dose-dependent enhancement of vasoconstrictor responses. In conclusion, aldosterone induces a constrictor effect, which is attenuated by intact endothelial function. This protective effect of endothelium is partly due to an increase in NO synthase activity which is phosphatidylinositol 3-kinase (PI3K) dependent [63].

Receptors Involved in Nongenomic Effects of Aldosterone

In intact mesenteric vessels isolated from male Sprague-Dawley rats, aldosterone (10 nm, 10 min) potentiated the constrictor response to norepinephrine and elicited a direct constrictor effect within the first 10 min [64]. A direct constrictor action of aldosterone (10–13– 10–16m, <10 min) was also observed in mouse mesenteric vessels [65]. Interestingly, contradictory effects of MR antagonists spironolactone and eplerenone on the direct constrictor action of aldosterone have been described, from complete inhibition [64] to the absence of blockade [65], suggesting that different receptors might be involved.

Yamada et al. [65] showed that the rapid constrictor effect of aldosterone on mesenteric vessels was unaffected by MR antagonists but was blunted by the AT1 blockers candesartan or valsartan and by the transglutaminase inhibitors cystamine and monodansyl cadaverine. These results emphasize the potential role of intracellular transglutaminase activity and AT1 dimer formation on the direct and rapid vascular action of aldosterone [65].

Recently, Gros et al. [66] identified a new receptor involved in nongenomic effects of aldosterone, i.e. the 7-transmembrane-spanning, GPR30. They demonstrated that aldosterone induced a rapid (15 min) and concentration-dependent increase in phospho-ERK in endothelium-denuded aortic rings, which was partly attenuated by the MR antagonist eplerenone and by the GPR30-selective antagonist G15 [66].

Mechanisms Involved in Aldosterone-Induced Rapid Vascular Constriction

Aldosterone-induced rapid vascular constriction involves various mechanisms. In intact mesenteric vessels, aldosterone induced a rise in intracellular pH, and the direct constrictor effect was blunted in the presence of amiloride, an Na+-H+ exchanger inhibitor [64]. In vitro on VSMCs, aldosterone (1 nm) stimulated the activity of the sodium-proton exchanger since 22Na influx increased after 4 min, an effect blocked by amiloride, a specific inhibitor of the Na+-H+ exchanger [67]. The activation of the Na+-H+ exchanger by aldosterone in VSMCs was associated with an increase in IP3 levels and was inhibited by the specific inhibitors of phospholipase C neomycin and U-73122 [67]. Interestingly, in this study, MR antagonists did not block this effect of aldosterone [67]. These experiments suggest that in VSMCs, aldosterone induces an increase in the activity of the Na+-H+ exchanger through a mechanism involving phospholipase C and IP3 generation through MR-independent pathways [67,68]. In addition to increased IP3 levels and Na+-H+ exchanger activation, aldosterone induced an immediate rise in intracellular calcium, which was also MR independent since it was not blunted by spironolactone [68]. Finally, aldosterone induced myosin light chain phosphorylation. Myosin light chain phosphorylation started at 10 min with 1 nm of aldosterone and was MR [69] and GPR dependent [66].

Aldosterone exerts also a direct and rapid effect on p38 MAPK and NADPH oxidase activity through a c-Src-dependent pathway in VSMCs. This effect was abrogated by the MR blocker eplerenone, by PP2, a c-Src inhibitor, and in c-Src-deficient mice [19]. Interestingly, in VSMCs from hypertensive rats, upregulation of the c-Src signaling pathway was observed in response to exogenous aldosterone [9]. These processes may play a role in the fibrotic response to aldosterone.

Nongenomic pathways have also been involved in aldosterone-induced VSMC proliferation. Indeed, Ishizawa et al. [70] demonstrated that aldosterone induced a dose-dependent activation of big MAP kinase 1, an MAP kinase implicated in cell proliferation, differentiation, and survival, in rat aortic VSMCs. This effect was blunted by eplerenone but not by cycloheximide, suggesting a nongenomic process [70].

Aldosterone is increased in metabolic syndrome and obese patients and is also a predictor of incident metabolic syndrome as shown in the longitudinal follow-up of 2,292 participants of the Framingham Offspring Study [71]. An independent association between body weight and PAC has been shown in overweight patients with [72] or without hypertension [73]. Interestingly, plasma aldosterone decreases after weight loss [74]. Increased aldosterone levels in obese patients could be attributed to circulating factors which stimulate aldosterone synthesis by the adrenal gland such as insulin [75], C1q TNF-related protein [76], exogenous fatty acid oxidation products [77], and adipokines [78]. Local synthesis of aldosterone by adipocytes could also participate in the hyperaldosteronism of the metabolic syndrome and/or in obese patients. Indeed, aldosterone can now be considered a novel adipocyte-derived factor since direct evidence of aldosterone synthesis by adipocytes has been shown. Adipocytes possess aldosterone synthase and produce aldosterone in an AT1 receptor-dependent manner [79].

Aldosterone synthesis by adipocytes influences vascular function. Briones et al. [79] showed that mesenteric arteries from db/db mice had an impairment of acetylcholine-induced relaxation when perivascular fat was preserved, and it was blunted by the MR antagonist eplerenone.

Adipocyte-conditioned medium obtained from differentiated 3T3-L1 adipocytes activated MAPK signaling in VSMCs leading to inflammatory and profibrotic responses. MR antagonism blunted adipocyte-conditioned medium stimulation of p38 MAPK and ERK1/2 phosphorylation [80]. Aldosterone induces insulin resistance in rat VSMCs through a mechanism involving insulin growth factor-1 receptor (IGF1R) [81]. Aldosterone increased insulin-induced ERK1/2 phosphorylation and α-smooth muscle actin expression, which was significantly attenuated by eplerenone or picropodophyllin, an IGF1R inhibitor [81]. In addition, aldosterone decreased insulin receptor substrate-1 expression via c-Src and reactive oxygen species stimulation by proteasome-dependent degradation in VSMCs [82].

The advantage of aldosterone blockade in patients has been shown with the two drugs that block MR, i.e. spironolactone and eplerenone, in congestive heart failure. Low doses of these drugs (25–50 mg/day) in addition to renin-angiotensin system blockers (RAS) and β-blockers improved the cardiovascular morbidity and mortality of patients with class II–IV chronic heart failure (RALES trial in chronic heart failure and EPHESUS in heart failure after myocardial infarction) [83,84]. In hypertensive patients, eplerenone has been shown to significantly improve the stiffness of small resistance arteries [85]. Small clinical studies have shown a beneficial effect of the use of eplerenone or spironolactone on proteinuria and chronic kidney disease progression [1]. Though useful in resistant hypertension, in the presence of renal failure there is the problem of handling hyperkalemia, which becomes a limitation for the use of these agents. The use of spironoclactone is limited by its lack of specificity for MR. Spironolactone also binds to the progesterone and androgen receptors and long-term use is associated with gynecomastia and menstrual irregularities. In addition, the use of MR blockers leads to an increase in aldosterone concentration, which may exert deleterious effects on the cardiovascular system through non-MR-dependent pathways. New drugs targeting aldosterone synthase have been developed more recently, such as LCI699. The main limitation of this agent is that a partial inhibition of the glucocorticoid axis was also observed in 20% of treated patients with primary hyperaldosteronism [86] or in essential hypertension [87]. Second generation aldosterone synthase inhibitors with higher specificity for the enzyme should limit this adverse effect.

Aldosterone exerts direct effects on the vascular system by inducing oxidative stress, inflammation, hypertrophic remodeling, and fibrosis. Aldosterone exerts its effects through genomic and nongenomic pathways in an MR-dependent or independent manner. Other aldosterone receptors such as GPR30 have been identified. A tight relation exists between aldosterone and angiotensin II pathways underlying the pharmacological interest of blockade of both pathways. Recently, an increasing body of evidence has underlined the importance of aldosterone in cardiovascular complications associated with the metabolic syndrome, such as arterial remodeling and endothelial dysfunction. Blockade of MR is an increasingly used evidence-based therapy for many forms of cardiovascular disease. However, the absence of specificity of some of the agents and the potential for development of hyperkalemia when renal function is impaired results in side effects that somewhat limit their use.

The work of E.L.S. was supported by Canadian Institutes of Health Research (CIHR) grants 37917, 82790, and 102606, a Canada Research Chair on Hypertension and Vascular Research from CIHR/Government of Canada, and the Canada Fund for Innovation. M.B. was supported by a fellowship from the Heart and Stroke Foundation of Canada.

1.
Briet M, Schiffrin EL: Aldosterone: effects on the kidney and cardiovascular system. Nat Rev Nephrol 2010;6:261–273.
2.
Conn JW: Primary aldosteronism, a new clinical syndrome. J Lab Clin Med 1955;45:3–17.
3.
Genest J, Lemieux G, Davignon A, Koiw E, Nowaczynski W, Steyermark P: Human arterial hypertension: a state of mild chronic hyperaldosteronism? Science 1956;123:503–505.
4.
Park JB, Schiffrin EL: ET(A) receptor antagonist prevents blood pressure elevation and vascular remodeling in aldosterone-infused rats. Hypertension 2001;37:1444–1449.
5.
Park JB, Schiffrin EL: Cardiac and vascular fibrosis and hypertrophy in aldosterone-infused rats: role of endothelin-1. Am J Hypertens 2002;15:164–169.
6.
Benetos A, Lacolley P, Safar ME: Prevention of aortic fibrosis by spironolactone in spontaneously hypertensive rats. Arterioscler Thromb Vasc Biol 1997;17:1152–1156.
7.
Virdis A, Neves MF, Amiri F, Viel E, Touyz RM, Schiffrin EL: Spironolactone improves angiotensin-induced vascular changes and oxidative stress. Hypertension 2002;40:504–510.
8.
Mazak I, Fiebeler A, Muller DN, Park JK, Shagdarsuren E, Lindschau C, Dechend R, Viedt C, Pilz B, Haller H, et al: Aldosterone potentiates angiotensin II-induced signaling in vascular smooth muscle cells. Circulation 2004;109:2792–2800.
9.
Callera GE, Montezano AC, Yogi A, Tostes RC, He Y, Schiffrin EL, Touyz RM: c-Src-dependent nongenomic signaling responses to aldosterone are increased in vascular myocytes from spontaneously hypertensive rats. Hypertension 2005;46:1032–1038.
10.
Miyata K, Hitomi H, Guo P, Zhang GX, Kimura S, Kiyomoto H, Hosomi N, Kagami S, Kohno M, Nishiyama A: Possible involvement of Rho-kinase in aldosterone-induced vascular smooth muscle cell remodeling. Hypertens Res 2008;31:1407–1413.
11.
Lacolley P, Labat C, Pujol A, Delcayre C, Benetos A, Safar M: Increased carotid wall elastic modulus and fibronectin in aldosterone-salt-treated rats: effects of eplerenone. Circulation 2002;106:2848–2853.
12.
Blacher J, Amah G, Girerd X, Kheder A, Ben Mais H, London GM, Safar ME: Association between increased plasma levels of aldosterone and decreased systemic arterial compliance in subjects with essential hypertension. Am J Hypertens 1997;10:1326–1334.
13.
Safar ME, Cattan V, Lacolley P, Nzietchueng R, Labat C, Lajemi M, de Luca N, Benetos A: Aldosterone synthase gene polymorphism, stroke volume and age-related changes in aortic pulse wave velocity in subjects with hypertension. J Hypertens 2005;23:1159–1166.
14.
Brilla CG, Weber KT: Mineralocorticoid excess, dietary sodium, and myocardial fibrosis. J Lab Clin Med 1992;120:893–901.
15.
Calo LA, Puato M, Schiavo S, Zanardo M, Tirrito C, Pagnin E, Balbi G, Davis PA, Palatini P, Pauletto P: Absence of vascular remodelling in a high angiotensin-II state (Bartter’s and Gitelman’s syndromes): implications for angiotensin II signalling pathways. Nephrol Dial Transplant 2008;23:2804–2809.
16.
Nakano S, Kobayashi N, Yoshida K, Ohno T, Matsuoka H: Cardioprotective mechanisms of spironolactone associated 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/salt-induced hypertensive rats. Hypertens Res 2005;28:925–936.
17.
Rocha R, Martin-Berger CL, Yang P, Scherrer R, Delyani J, McMahon E: Selective aldosterone blockade prevents angiotensin II/salt-induced vascular inflammation in the rat heart. Endocrinology 2002;143:4828–4836.
18.
Johar S, Cave AC, Narayanapanicker A, Grieve DJ, Shah AM: Aldosterone mediates angiotensin II-induced interstitial cardiac fibrosis via a Nox2-containing NADPH oxidase. FASEB J 2006;20:1546–1548.
19.
Callera GE, Touyz RM, Tostes RC, Yogi A, He Y, Malkinson S, Schiffrin EL: Aldosterone activates vascular p38MAP kinase and NADPH oxidase via c-Src. Hypertension 2005;45:773–779.
20.
Callera GE, Yogi A, Briones AM, Montezano AC, He Y, Tostes RC, Schiffrin EL, Touyz RM: Vascular proinflammatory responses by aldosterone are mediated via c-Src trafficking to cholesterol-rich microdomains: role of PDGFR. Cardiovasc Res 2011;91:720–731.
21.
Iwashima F, Yoshimoto T, Minami I, Sakurada M, Hirono Y, Hirata Y: Aldosterone induces superoxide generation via Rac1 activation in endothelial cells. Endocrinology 2008;149:1009–1014.
22.
Kasal DA, Barhoumi T, Li MW, Yamamoto N, Zdanovich E, Rehman A, Neves MF, Laurant P, Paradis P, Schiffrin EL: T regulatory lymphocytes prevent aldosterone-induced vascular injury. Hypertension 2012;59:324–330.
23.
Caprio M, Newfell BG, la Sala A, Baur W, Fabbri A, Rosano G, Mendelsohn ME, Jaffe IZ: Functional mineralocorticoid receptors in human vascular endothelial cells regulate intercellular adhesion molecule-1 expression and promote leukocyte adhesion. Circ Res 2008;102:1359–1367.
24.
Sun Y, Zhang J, Lu L, Chen SS, Quinn MT, Weber KT: Aldosterone-induced inflammation in the rat heart: role of oxidative stress. Am J Pathol 2002;161:1773–1781.
25.
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.
26.
Blanco-Rivero J, Cachofeiro V, Lahera V, Aras-Lopez R, Marquez-Rodas I, Salaices M, Xavier FE, Ferrer M, Balfagon G: Participation of prostacyclin in endothelial dysfunction induced by aldosterone in normotensive and hypertensive rats. Hypertension 2005;46:107–112.
27.
Farquharson CA, Struthers AD: Spironolactone increases nitric oxide bioactivity, improves endothelial vasodilator dysfunction, and suppresses vascular angiotensin I/angiotensin II conversion in patients with chronic heart failure. Circulation 2000;101:594–597.
28.
Garnier A, Bendall JK, Fuchs S, Escoubet B, Rochais F, Hoerter J, Nehme J, Ambroisine ML, De Angelis N, Morineau G, et al: Cardiac specific increase in aldosterone production induces coronary dysfunction in aldosterone synthase-transgenic mice. Circulation 2004;110:1819–1825.
29.
Favre J, Gao J, Zhang AD, Remy-Jouet I, Ouvrard-Pascaud A, Dautreaux B, Escoubet B, Thuillez C, Jaisser F, Richard V: Coronary endothelial dysfunction after cardiomyocyte-specific mineralocorticoid receptor overexpression. Am J Physiol Heart Circ Physiol 2011;300:H2035–H2043.
30.
Leopold JA, Dam A, Maron BA, Scribner AW, Liao R, Handy DE, Stanton RC, Pitt B, Loscalzo J: Aldosterone impairs vascular reactivity by decreasing glucose-6-phosphate dehydrogenase activity. Nat Med 2007;13:189–197.
31.
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.
32.
Schneider SW, Yano Y, Sumpio BE, Jena BP, Geibel JP, Gekle M, Oberleithner H: Rapid aldosterone-induced cell volume increase of endothelial cells measured by the atomic force microscope. Cell Biol Int 1997;21:759–768.
33.
Oberleithner H, Schneider SW, Albermann L, Hillebrand U, Ludwig T, Riethmuller C, Shahin V, Schafer C, Schillers H: Endothelial cell swelling by aldosterone. J Membr Biol 2003;196:163–172.
34.
Oberleithner H, Riethmuller C, Ludwig T, Hausberg M, Schillers H: Aldosterone remodels human endothelium. Acta Physiol (Oxf) 2006;187:305–312.
35.
Hillebrand U, Schillers H, Riethmuller C, Stock C, Wilhelmi M, Oberleithner H, Hausberg M: Dose-dependent endothelial cell growth and stiffening by aldosterone: endothelial protection by eplerenone. J Hypertens 2007;25:639–647.
36.
Oberleithner H, Riethmuller C, Schillers H, MacGregor GA, de Wardener HE, Hausberg M: Plasma sodium stiffens vascular endothelium and reduces nitric oxide release. Proc Natl Acad Sci USA 2007;104:16281–16286.
37.
Thum T, Schmitter K, Fleissner F, Wiebking V, Dietrich B, Widder JD, Jazbutyte V, Hahner S, Ertl G, Bauersachs J: Impairment of endothelial progenitor cell function and vascularization capacity by aldosterone in mice and humans. Eur Heart J 2011;32:1275–1286.
38.
Wu VC, Lo SC, Chen YL, Huang PH, Tsai CT, Liang CJ, Kuo CC, Kuo YS, Lee BC, Wu EL, et al: Endothelial progenitor cells in primary aldosteronism: a biomarker of severity for aldosterone vasculopathy and prognosis. J Clin Endocrinol Metab 2011;96:3175–3183.
39.
Schiffrin EL, Gutkowska J, Genest J: Effect of angiotensin II and deoxycorticosterone infusion on vascular angiotensin II receptors in rats. Am J Physiol 1984;246:H608–H614.
40.
Schiffrin EL, Franks DJ, Gutkowska J: Effect of aldosterone on vascular angiotensin II receptors in the rat. Can J Physiol Pharmacol 1985;63:1522–1527.
41.
Hatakeyama H, Miyamori I, Fujita T, Takeda Y, Takeda R, Yamamoto H: Vascular aldosterone: biosynthesis and a link to angiotensin II-induced hypertrophy of vascular smooth muscle cells. J Biol Chem 1994;269:24316–24320.
42.
Takeda Y, Miyamori I, Yoneda T, Iki K, Hatakeyama H, Blair IA, Hsieh FY, Takeda R: Production of aldosterone in isolated rat blood vessels. Hypertension 1995;25:170–173.
43.
Ahmad N, Romero DG, Gomez-Sanchez EP, Gomez-Sanchez CE: Do human vascular endothelial cells produce aldosterone? Endocrinology 2004;145:3626–3629.
44.
Fiebeler A, Nussberger J, Shagdarsuren E, Rong S, Hilfenhaus G, Al-Saadi N, Dechend R, Wellner M, Meiners S, Maser-Gluth C, et al: Aldosterone synthase inhibitor ameliorates angiotensin II-induced organ damage. Circulation 2005;111:3087–3094.
45.
Xiao F, Puddefoot JR, Barker S, Vinson GP: Mechanism for aldosterone potentiation of angiotensin II-stimulated rat arterial smooth muscle cell proliferation. Hypertension 2004;44:340–345.
46.
Min LJ, Mogi M, Li JM, Iwanami J, Iwai M, Horiuchi M: Aldosterone and angiotensin II synergistically induce mitogenic response in vascular smooth muscle cells. Circ Res 2005;97:434–442.
47.
Min LJ, Mogi M, Iwanami J, Li JM, Sakata A, Fujita T, Tsukuda K, Iwai M, Horiuchi M: Cross-talk between aldosterone and angiotensin II in vascular smooth muscle cell senescence. Cardiovasc Res 2007;76:506–516.
48.
Montezano AC, Callera GE, Yogi A, He Y, Tostes RC, He G, Schiffrin EL, Touyz RM: Aldosterone and angiotensin II synergistically stimulate migration in vascular smooth muscle cells through c-Src-regulated redox-sensitive RhoA pathways. Arterioscler Thromb Vasc Biol 2008;28:1511–1518.
49.
Batenburg WW, Jansen PM, van den Bogaerdt AJ, Danser AHJ: Angiotensin II-aldosterone interaction in human coronary microarteries involves GPR30, EGFR, and endothelial NO synthase. Cardiovasc Res 2012;94:136–143.
50.
Lemarie CA, Simeone SM, Nikonova A, Ebrahimian T, Deschenes ME, Coffman TM, Paradis P, Schiffrin EL: Aldosterone-induced activation of signaling pathways requires activity of angiotensin type 1a receptors. Circ Res 2009;105:852–859.
51.
Lariviere R, Thibault G, Schiffrin EL: Increased endothelin-1 content in blood vessels of deoxycorticosterone acetate-salt hypertensive but not in spontaneously hypertensive rats. Hypertension 1993;21:294–300.
52.
Schiffrin EL, Lariviere R, Li JS, Sventek P, Touyz RM: Deoxycorticosterone acetate plus salt induces overexpression of vascular endothelin-1 and severe vascular hypertrophy in spontaneously hypertensive rats. Hypertension 1995;25:769–773.
53.
Amiri F, Virdis A, Neves MF, Iglarz M, Seidah NG, Touyz RM, Reudelhuber TL, Schiffrin EL: Endothelium-restricted overexpression of human endothelin-1 causes vascular remodeling and endothelial dysfunction. Circulation 2004;110:2233–2240.
54.
Schiffrin EL, Sventek P, Li JS, Turgeon A, Reudelhuber T: Antihypertensive effect of an endothelin receptor antagonist in DOCA-salt spontaneously hypertensive rats. Br J Pharmacol 1995;115:1377–1381.
55.
Li JS, Turgeon A, Schiffrin EL: Effect of chronic treatment with two different ET(A) selective endothelin receptor antagonists on blood pressure and small artery structure of deoxycorticosterone acetate (DOCA)-salt hypertensive rats. Am J Hypertens 1998;11:554–562.
56.
Pu Q, Neves MF, Virdis A, Touyz RM, Schiffrin EL: Endothelin antagonism on aldosterone-induced oxidative stress and vascular remodeling. Hypertension 2003;42:49–55.
57.
Stow LR, Gumz ML, Lynch IJ, Greenlee MM, Rudin A, Cain BD, Wingo CS: Aldosterone modulates steroid receptor binding to the endothelin-1 gene (edn1). J Biol Chem 2009;284:30087–30096.
58.
Wehling M, Spes CH, Win N, Janson CP, Schmidt BM, Theisen K, Christ M: Rapid cardiovascular action of aldosterone in man. J Clin Endocrinol Metab 1998;83:3517–3522.
59.
Schmidt BM, Montealegre A, Janson CP, Martin N, Stein-Kemmesies C, Scherhag A, Feuring M, Christ M, Wehling M: Short term cardiovascular effects of aldosterone in healthy male volunteers. J Clin Endocrinol Metab 1999;84:3528–3533.
60.
Schmidt BM, Oehmer S, Delles C, Bratke R, Schneider MP, Klingbeil A, Fleischmann EH, Schmieder RE: Rapid nongenomic effects of aldosterone on human forearm vasculature. Hypertension 2003;42:156–160.
61.
Gunaruwan P, Schmitt M, Sharman J, Lee L, Struthers A, Frenneaux M: Effects of aldosterone on forearm vasculature in treated chronic heart failure. Am J Cardiol 2005;95:412–414.
62.
Schmidt BM, Sammer U, Fleischmann I, Schlaich M, Delles C, Schmieder RE: Rapid nongenomic effects of aldosterone on the renal vasculature in humans. Hypertension 2006;47:650–655.
63.
Liu SL, Schmuck S, Chorazcyzewski JZ, Gros R, Feldman RD: Aldosterone regulates vascular reactivity: short-term effects mediated by phosphatidylinositol 3-kinase-dependent nitric oxide synthase activation. Circulation 2003;108:2400–2406.
64.
Michea L, Delpiano AM, Hitschfeld C, Lobos L, Lavandero S, Marusic ET: Eplerenone blocks nongenomic effects of aldosterone on the Na+/H+ exchanger, intracellular Ca2+ levels, and vasoconstriction in mesenteric resistance vessels. Endocrinology 2005;146:973–980.
65.
Yamada M, Kushibiki M, Osanai T, Tomita H, Okumura K: Vasoconstrictor effect of aldosterone via angiotensin II type 1 (AT1) receptor: possible role of AT1 receptor dimerization. Cardiovasc Res 2008;79:169–178.
66.
Gros R, Ding Q, Sklar LA, Prossnitz EE, Arterburn JB, Chorazyczewski J, Feldman RD: GPR30 expression is required for the mineralocorticoid receptor-independent rapid vascular effects of aldosterone. Hypertension 2011;57:442–451.
67.
Christ M, Douwes K, Eisen C, Bechtner G, Theisen K, Wehling M: Rapid effects of aldosterone on sodium transport in vascular smooth muscle cells. Hypertension 1995;25:117–123.
68.
Wehling M, Neylon CB, Fullerton M, Bobik A, Funder JW: Nongenomic effects of aldosterone on intracellular Ca2+ in vascular smooth muscle cells. Circ Res 1995;76:973–979.
69.
Gros R, Ding Q, Armstrong S, O’Neil C, Pickering JG, Feldman RD: Rapid effects of aldosterone on clonal human vascular smooth muscle cells. Am J Physiol Cell Physiol 2007;292:C788–C794.
70.
Ishizawa K, Izawa Y, Ito H, Miki C, Miyata K, Fujita Y, Kanematsu Y, Tsuchiya K, Tamaki T, Nishiyama A, et al: Aldosterone stimulates vascular smooth muscle cell proliferation via big mitogen-activated protein kinase 1 activation. Hypertension 2005;46:1046–1052.
71.
Ingelsson E, Pencina MJ, Tofler GH, Benjamin EJ, Lanier KJ, Jacques PF, Fox CS, Meigs JB, Levy D, Larson MG, et al: Multimarker approach to evaluate the incidence of the metabolic syndrome and longitudinal changes in metabolic risk factors: the Framingham Offspring Study. Circulation 2007;116:984–992.
72.
Rossi GP, Belfiore A, Bernini G, Fabris B, Caridi G, Ferri C, Giacchetti G, Letizia C, Maccario M, Mannelli M, et al: Body mass index predicts plasma aldosterone concentrations in overweight-obese primary hypertensive patients. J Clin Endocrinol Metab 2008;93:2566–2571.
73.
Bentley-Lewis R, Adler GK, Perlstein T, Seely EW, Hopkins PN, Williams GH, Garg R: Body mass index predicts aldosterone production in normotensive adults on a high-salt diet. J Clin Endocrinol Metab 2007;92:4472–4475.
74.
Goodfriend TL, Kelley DE, Goodpaster BH, Winters SJ: Visceral obesity and insulin resistance are associated with plasma aldosterone levels in women. Obes Res 1999;7:355–362.
75.
Petrasek D, Jensen G, Tuck M, Stern N: In vitro effects of insulin on aldosterone production in rat zona glomerulosa cells. Life Sci 1992;50:1781–1787.
76.
Jeon JH, Kim KY, Kim JH, Baek A, Cho H, Lee YH, Kim JW, Kim D, Han SH, Lim JS, et al: A novel adipokine CTRP1 stimulates aldosterone production. FASEB J 2008;22:1502–1511.
77.
Goodfriend TL, Ball DL, Raff H, Bruder ED, Gardner HW, Spiteller G: Oxidized products of linoleic acid stimulate adrenal steroidogenesis. Endocr Res 2002;28:325–330.
78.
Ehrhart-Bornstein M, Lamounier-Zepter V, Schraven A, Langenbach J, Willenberg HS, Barthel A, Hauner H, McCann SM, Scherbaum WA, Bornstein SR: Human adipocytes secrete mineralocorticoid-releasing factors. Proc Natl Acad Sci USA 2003;100:14211–14216.
79.
Briones AM, Cat AN, Callera GE, Yogi A, Burger D, He Y, Correa JW, Gagnon AM, Gomez-Sanchez CE, Gomez-Sanchez EP, et al: Adipocytes produce aldosterone through calcineurin-dependent signaling pathways: implications in diabetes mellitus-associated obesity and vascular dysfunction. Hypertension 2012;59:1069–1078.
80.
Nguyen Dinh Cat A, Briones AM, Callera GE, Yogi A, He Y, Montezano AC, Touyz RM: Adipocyte-derived factors regulate vascular smooth muscle cells through mineralocorticoid and glucocorticoid receptors. Hypertension 2011;58:479–488.
81.
Sherajee SJ, Fujita Y, Rafiq K, Nakano D, Mori H, Masaki T, Hara T, Kohno M, Nishiyama A, Hitomi H: Aldosterone induces vascular insulin resistance by increasing insulin-like growth factor-1 receptor and hybrid receptor. Arterioscler Thromb Vasc Biol 2012;32:257–263.
82.
Hitomi H, Kiyomoto H, Nishiyama A, Hara T, Moriwaki K, Kaifu K, Ihara G, Fujita Y, Ugawa T, Kohno M: Aldosterone suppresses insulin signaling via the downregulation of insulin receptor substrate-1 in vascular smooth muscle cells. Hypertension 2007;50:750–755.
83.
Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, Bittman R, Hurley S, Kleiman J, Gatlin M: Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med 2003;348:1309–1321.
84.
Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, Palensky J, Wittes J: 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.
85.
Savoia C, Touyz RM, Amiri F, Schiffrin EL: Selective mineralocorticoid receptor blocker eplerenone reduces resistance artery stiffness in hypertensive patients. Hypertension 2008;51:432–439.
86.
Amar L, Azizi M, Menard J, Peyrard S, Watson C, Plouin PF: Aldosterone synthase inhibition with LCI699: a proof-of-concept study in patients with primary aldosteronism. Hypertension 2010;56:831–838.
87.
Calhoun DA, White WB, Krum H, Guo W, Bermann G, Trapani A, Lefkowitz MP, Menard J: Effects of a novel aldosterone synthase inhibitor for treatment of primary hypertension: results of a randomized, double-blind, placebo- and active-controlled phase 2 trial. Circulation 2011;124:1945–1955.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.