university of groningen towards personalized

31
University of Groningen Towards personalized cardiovascular risk management in renal transplant recipients de Vries, Laura Victorine IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2018 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): de Vries, L. V. (2018). Towards personalized cardiovascular risk management in renal transplant recipients. Rijksuniversiteit Groningen. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license. More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne- amendment. Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 04-10-2021

Upload: others

Post on 04-Oct-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: University of Groningen Towards personalized

University of Groningen

Towards personalized cardiovascular risk management in renal transplant recipientsde Vries, Laura Victorine

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):de Vries, L. V. (2018). Towards personalized cardiovascular risk management in renal transplant recipients.Rijksuniversiteit Groningen.

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license.More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne-amendment.

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 04-10-2021

Page 2: University of Groningen Towards personalized

Chapter 3ALDOSTERONE, FROM (PATHO)PHYSIOLOGY TO TREATMENT IN CARDIOVASCULAR AND

RENAL DAMAGE

Femke Waanders Laura V. de Vries Harry van Goor

Jan-Luuk HillebrandsGozewijn LavermanStephan J. L. Bakker

Gerjan Navis

Curr Vasc Pharmacol. 2011; 9: 594-605.

Page 3: University of Groningen Towards personalized

Chapter 3

54

ABSTRACT

Aldosterone, a steroid hormone with mineralocorticoid activity, is far more than merely a salt-and-water hormone. Aldosterone has a number of non-classical, mineralocor-ticoid receptor (MR)-mediated actions, including tissue remodeling, modulation of vascular tone, and stimulating inflammation and fibrosis, which may fuel progression of end organ damage. Aldosterone breakthrough during blockade of the renin-angio-tensin aldosterone system (RAAS) may explain why this treatment regimen only confers partial cardiovascular and renal protection. Of major interest, aldosterone is delete-rious only if inappropriately high for its sodium status i.e. high aldosterone and high sodium. The mechanism of sodium dependence of aldosterone-induced renal and car-diovascular damage continues to fascinate. Aldosterone excess increases sodium and fluid retention and consequently increases blood pressure, which, in turn, mediates target organ damage. Moreover, blood pressure independent effects play a role with dissociation of low circulating and high tissue aldosterone levels during high sodium intake and possibly enhanced MR signaling. MR blockade is a valuable strategy, which has potency to halt the progressive end organ damage as observed during current treatments. In heart failure, MR blockade on top of RAAS blockade reduces hard clini-cal endpoints. Despite encouraging results on the intermediate endpoint proteinuria, long-term data on the efficacy and safety of MR blockade in preventing dialysis and/or cardiovascular endpoints in chronic kidney disease are still lacking. It is obligatory that future clinical studies on the effects of MR blockade on end-organ damage take into account the sodium status.

Page 4: University of Groningen Towards personalized

Aldosterone and Renal Damage

55

3

INTRODUCTION

Aldosterone is classically known as a main regulator of sodium and potassium homeo-stasis. As one of the effector hormones of the renin-angiotensin-aldosterone system (RAAS), it plays a main role in the homeostatic response to volume depletion, leading to increased renal tubular sodium reabsorption in order to restore volume status. It also induces potassium loss, during conditions of high plasma potassium and sodium depletion. In classical experiments, continuous infusion of aldosterone induced sodium retention with a positive volume balance and hence hypertension, followed by the so-called aldosterone-escape, with re-establishment of steady state despite ongoing aldosterone infusion.1

In more recent years, the label “aldosterone-escape” is used to indicate a different phenomenon, namely the restoration of aldosterone towards baseline values after an initial decrease during blockade of the RAAS by angiotensin converting enzyme (ACE) inhibition or angiotensin II receptor blockade (ARB).2 This phenomenon is also known as aldosterone breakthrough. The transient response of circulating aldoste-rone levels to ACE inhibition is illustrated in Figure 1, showing the day-to-day values of aldosterone in patients with essential hypertension, instituted on the ACE-inhibitor enalapril on low (50 mmol/d) and high (200 mmol/d) sodium intake.3 It shows that the decrease in aldosterone is rapid, occurs on both sodium intakes, and is followed by breakthrough towards baseline values already during the first week of treatment.3 This may well reflect a homeostatic response to the concurrent negative sodium balance, and/or “escape” due to the reactive rise in renin that occurs. Interestingly, the values during high sodium parallel those during low sodium, at an appropriately suppressed level, demonstrating that the relationship between sodium intake and circulating aldosterone remains intact during this pharmacological blockade of the RAAS by ACE inhibition.

ACE inhibitors and ARBs have long been the dominant strategies for RAAS blockade in hypertension, heart failure and renal disease. Aldosterone blockade had a relatively minor role, mainly consisting of the use of spironolactone as a potassium sparing diuretic for conditions characterized by edema and secondary hyperaldosteronism, like heart failure and cirrhosis, conditions in which normal aldosterone escape is impaired.1 However, a shift of paradigm occurred when Pitt et al. reported specific cardioprotective effects in heart failure that could not be attributed to diuretic effects, but rather pointed towards a specific anti-fibrotic action. This observation fueled a new wave of interest in the pathophysiological effects of aldosterone, and its phar-macological blockade as a tool for intervention in cardiovascular and renal disease.4,5

Page 5: University of Groningen Towards personalized

Chapter 3

56

Interestingly, some of the novel, non-classical effects of aldosterone also appear to have a link with sodium status, with more prominent deleterious effects of aldosterone during conditions of sodium excess (Figure 2).

Here, we will review the recent insights of the role of aldosterone in renal and vascular damage and its interaction with sodium status. Furthermore, we will discuss the effi-cacy of mineralocorticoid receptor (MR) blockade in the treatment of patients with a high cardiovascular risk profile as well as in patients with chronic kidney disease (CKD).

Aldosterone, From (Patho)Physiology to Treatment Current Vascular Pharmacology, 2011, Vol. 9, No. 5 595

Fig. (1). Transient response of circulating aldosterone levels to

ACE-inhibition. Day-to-day values plasma renin activity (PRA) and

plasma aldosterone concentration (PAC) in patients with essential

hypertension treated with ACE inhibition, enalapril 10 mg. The

continuous lines represent low sodium intake (50 mmol/d) and the

broken lines a liberal sodium diet (200 mmol/d). The decrease in

aldosterone is rapid, occurs on both sodium intakes, and is followed

by breakthrough towards baseline values already during the first

week of treatment. Reprinted from reference [3].

Here, we will review the recent insights of the role of aldosterone in renal and vascular damage and its interaction with sodium status. Furthermore, we will discuss the efficacy of mineralocorticoid receptor (MR) blockade in the treatment of patients with a high cardiovascular risk profile as well as in patients with chronic kidney disease (CKD).

CLASSICAL EFFECTS OF ALDOSTERONE ON

SODIUM,POTASSIUM AND WATER HOMEOSTASIS

Aldosterone is a steroid hormone with mineralocorticoid activity. Not only angiotensin II, but also potassium and adrenocorticotropic hormone (ACTH) stimulate the adrenal zona glomerulosa, the outer layer of the adrenal cortex, to synthesize aldosterone. Aldosterone can also be synthesized locally in tissues such as blood vessels, heart and kidneys [6, 7]. Synthesis at extra-adrenal sites is regulated by the RAAS and the amount produced is <1% compared with the amount produced by the adrenal glands [6, 7]. Consequently, none of these extra-adrenal sources significantly contribute to plasma levels; circulating aldosterone levels fall essentially to zero with adrenalectomy. Circulating aldosterone binds to the inactive cytosolic MR on target cells with epithelial sodium channels. Main target cells in the kidney are located on the principal cells of the cortical collecting duct in the distal nephron. However, the classical effects of aldosterone (i.e. promotion of sodium retention and potassium loss) are not confined to the kidney. Aldosterone exerts similar, but lesser effects on epithelial cells in the colon, sweat, and salivary

Fig. (2). Proposed interaction between the classical pathway of

aldosterone release, sodium status, and metabolic status in cardio-

vascular and renal disease. The left box shows classical associations

between aldosterone and sodium intake. These mainly involve in-

creased circulating aldosterone during sodium depletion and sup-

pression of circulating aldosterone during high sodium intake. The

right upper and right lower boxes show recent insights on interac-

tion of sodium and metabolic status with this classical pathway. The

box shows dissociation of adequately suppressed circulating aldos-

terone levels with high aldosterone levels in target tissues during

high sodium intake [7, 59]. The right lower box shows impaired

negative feed-back by high sodium intake on circulating aldoster-

one in obesity, which is likely to be caused by secretion of aldoster-

one-releasing factors by adipocytes [44, 64]. Enhanced MR activa-

tion by increased levels of both circulating and tissue aldosterone

directly contributes to progressive cardiovascular and renal damage

through combined effects of blood pressure with fibrosis and in-

flammation [20-23]. Abbreviations: BMI: body mass index; RAAS:

renin-angiotensin aldosterone system; MR: mineralocorticoid re-

ceptor.

glands. Binding of aldosterone to the MR on target cells results in translocation of the ligand-activated MR into the nucleus, where it exerts its transcriptional effects. It binds to hormone-response elements in the regulatory region of target gene promoters (the MR is a nuclear hormone receptor), re-sulting in protein synthesis. In the distal nephron of the kid-ney, MR-induced serum- and glucocorticoid-inducible kinase-1 (sgk-1) gene expression triggers a cascade of mo-lecular events that leads to three important changes. First, it

�������������������

��

����

����

�� �

�� �

��������

��������

��������

�������

����

������

�������

����

����������������������������� ���������

��������������������������������

����������������

���������������

�����������

�����������

������������

����������������

�������������������������

�������

������

�����������

���������������������������

���

� ���������

����� �������������������

Figure 1. Transient response of circulating aldosterone levels to ACE-inhibition. Day-to-day values plasma renin activity (PRA) and plasma aldosterone concentration (PAC) in patients with essential hypertension treated with ACE inhibition, enalapril 10 mg. The continuous lines represent low sodium intake (50 mmol/d) and the broken lines a liberal sodium diet (200 mmol/d). The decrease in aldosterone is rapid, occurs on both sodium intakes, and is followed by breakthrough towards baseline values already during the first week of treatment. Reprinted from reference.3

Page 6: University of Groningen Towards personalized

Aldosterone and Renal Damage

57

3

Classical effects of aldosterone on sodium, potassium, and water homeostasis Aldosterone is a steroid hormone with mineralocorticoid activity. Not only angiotensin II, but also potassium and adrenocorticotropic hormone (ACTH) stimulate the adrenal zona glomerulosa, the outer layer of the adrenal cortex, to synthesize aldosterone. Aldosterone can also be synthesized locally in tissues such as blood vessels, heart and kidneys.6,7 Synthesis at extra-adrenal sites is regulated by the RAAS and the amount produced is <1% compared with the amount produced by the adrenal glands.6,7 Conse-quently, none of these extra-adrenal sources significantly contribute to plasma levels; circulating aldosterone levels fall essentially to zero with adrenalectomy. Circulating aldosterone binds to the inactive cytosolic MR on target cells with epithelial sodium channels. Main target cells in the kidney are located on the principal cells of the cortical collecting duct in the distal nephron. However, the classical effects of aldosterone (i.e. promotion of sodium retention and potassium loss) are not confined to the kidney. Aldosterone exerts similar, but lesser effects on epithelial cells in the colon, sweat and salivary glands. Binding of aldosterone to the MR on target cells results in translocation of the ligand-activated MR into the nucleus, where it exerts its transcriptional effects. It binds to hormone-response elements in the regulatory region of target gene pro-moters (the MR is a nuclear hormone receptor), resulting in protein synthesis. In the distal nephron of the kidney, MR-induced serum- and glucocorticoid-inducible kinase-1 (sgk-1) gene expression triggers a cascade of molecular events that leads to three important changes. First, it increases the expression of apical epithelial sodium chan-nels, resulting in reabsorption of sodium and water. Second, it increases the expression of basolateral sodium/potassium ATPase for cellular sodium extrusion and potassium entry and third, it increases the expression of apical renal outer medullary potassium channels which are involved in passive excretion of potassium.8 Collectively, these processes lead to sodium and water reabsorption with subsequent volume expansion and an increase in blood pressure and potassium excretion.

Pre-receptor metabolismMineralocorticoid selectivity is achieved through co-expression of the MR and the enzyme 11beta-hydroxysteroid dehydrogenase type 2 (11β-HSD2). 11β-HSD2 metab-olizes glucocorticoid hormones into receptor-inactive 11-keto cogeners, preventing occupancy of MR by circulating cortisol (a glucocorticoid hormone). This is also known as ‘pre-receptor metabolism’. The affinity of the MR for aldosterone and glucocorti-coids is similar,9 however cortisol is 100- to 1000-fold more abundantly present in plasma than aldosterone. Clinical importance of this pre-receptor metabolism by 11β-HSD2 is highlighted by inactivating mutations found in the syndrome of apparent mineralocorticoid excess. This is a rare genetic defect characterized by early onset of severe low-renin hypokalemic hypertension.10 Moreover, consumption of liquorice also

Page 7: University of Groningen Towards personalized

Chapter 3

58

induces hypokalemic hypertension. Its active metabolite, glycyrrhetinic acid, inhibits 11β-HSD2 activity, allowing cortisol to escape pre-receptor metabolism and occupy and activate MR. Reduced activity of 11β-HSD2 can also contribute to the pathogenesis of human essential hypertension. Almost 40% reduction in ex vivo 11β-HSD2 catalytic activity, measured directly on sweat gland cells, was found in patients with essential hypertension as compared to normotensive subjects.11 Data from a very recent experi-mental study in rats revealed that 11β-HSD2 expression (and presumably also activity) was >50% reduced in the distal nephron of offspring of protein-restricted mothers.12 This suggests that the observed elevated blood pressure in the offspring may be the result of insufficient protection of the MR from inappropriate binding by glucocorti-coids, as only halve the level of glucocorticoids are required to inappropriately activate the MR in the distal nephron.

Primary hyperaldosteronismPrimary hyperaldosteronism is a syndrome, in which secretion of aldosterone is rela-tively autonomous of its normal stimuli, due to hyperplasia, adenoma, or carcinoma of the adrenal cortex. Patients with primary hyperaldosteronism, or presumed idiopathic hyperaldosteronism have an increased risk of stroke, myocardial infarction, and atrial fibrillation and have higher urinary albumin excretion rates than patients with pri-mary hypertension with similar blood pressure levels.13,14 In these patients aldosterone excess is associated with increased cardiac mass, fibrosis and cardiovascular tissue remodeling, independent of blood pressure.15 These data confirm the clinical relevance of blood pressure independent, direct adverse effects of aldosterone excess.16 Treat-ment of aldosterone excess by laparoscopic adrenalectomy (for patients with unilateral primary hyperaldosteronism) or by MR blockade reverses the excess in cardiovascular morbidity and improves the metabolic complications related to hyperaldosteronism in these patients.13,17-19

Beyond classical actions: profibrotic and proliferative effects of aldosteroneOver the last decade it has become increasingly clear that aldosterone can directly contribute to processes of tissue remodeling and progressive cardiovascular and renal damage through its pro-fibrotic actions (Figure 2).20-23 This appears to affect many cell types, including vascular cells, and different cell types in the heart and kidneys, medi-ating target organ damage.24-26 The cell-signaling pathways of the aldosterone-induced pro-fibrotic effects are not yet fully elucidated, but reactive oxygen species (ROS), NFκB, PAI-1, TGF-β, and increased collagen gene expression and synthesis are likely to be involved and can be abolished by MR blockade.23,27-30 Recent findings showed that the G allele of the common functional genetic polymorphism c.- 2G>C in the MR gene is associated with decreased MR protein levels compared with the C allele.31 Paradoxi-

Page 8: University of Groningen Towards personalized

Aldosterone and Renal Damage

59

3

cally, the G allele of this polymorphism is associated with increased RAAS activation and increased blood pressure.31 Mechanisms of aldosterone induced injury, downstream pathways of MR activation and cross-talk between aldosterone and angiotensin II sig-naling were reviewed recently elsewhere.32

Figure 2. Proposed interaction between the classical pathway of aldosterone release, sodium status, and metabolic status in cardiovascular and renal disease. The left box shows classical associations between aldosterone and sodium intake. These mainly involve increased circulating aldosterone during sodium depletion and suppression of circulating aldosterone during high sodium intake. The right upper and right lower boxes show recent insights on interaction of sodium and metabolic status with this classical pathway. The box shows dissociation of adequately suppressed circulating aldosterone levels with high aldosterone levels in target tissues during high sodium intake.7,58 The right lower box shows impaired negative feed-back by high sodium intake on circulating aldosterone in obesity, which is likely to be caused by secretion of aldosterone-releasing factors by adipocytes.43,63 Enhanced MR activation by increased levels of both circulating and tissue aldosterone directly contributes to progressive cardiovascular and renal damage through combined effects of blood pressure with fibrosis and inflammation.20-23 Abbreviations: BMI, body mass index; RAAS, renin-angiotensin-aldosterone system; MR, mineralocorticoid receptor.

Page 9: University of Groningen Towards personalized

Chapter 3

60

Beyond classical effects: interaction with metabolic statusClassically, glucocorticoids but not mineralocorticoids were thought to exert their effects on metabolic status. However, it has become increasingly clear that cross-talk between both types of steroids and their receptors exists, probably due to structural similar-ities of the MR and glucocorticoid receptor.9 Mounting evidence is pointing towards mutual interaction of aldosterone and MR activation with metabolic status (Figure 2). The impact of aldosterone on metabolic status, was first described in patients with primary hyperaldosteronism who also had impaired glucose tolerance.33 More recent studies performed in obese individuals and patients with primary hyperaldosteronism demonstrate that increased plasma aldosterone levels are independently associated with the metabolic syndrome, but also with its separate metabolic components i.e. impaired glucose metabolism and insulin resistance, high waist circumference and low HDL-cholesterol levels.17,34,35 Reduction of plasma aldosterone levels with either adrenalectomy or MR blockade with spironolactone normalizes plasma glucose and insulin levels.17,36

Several mechanisms have been proposed for the interaction of aldosterone with met-abolic status, of which the most important ones are the interaction of aldosterone with adipocytes and its effect on insulin signaling. Chronic exposure to aldosterone promotes adipogenesis in an MR-dependent manner in vitro.37,38 Adipocytes are able to secrete cytokines which can induce insulin resistance, such as interleukin-6, TNF-α and MCP-1. MR blockade reduced expression of these pro-inflammatory cytokines in obese diabetic mice.39 In a model of RAAS activation and insulin resistance in rats, MR blockade improved systemic insulin sensitivity and skeletal muscle glucose uptake, which was accompanied by a decrease in oxidative stress.40 Aldosterone also exerts inhibitory effects on insulin release by pancreatic β-cells and stimulates hepatic gluco-neogenesis.41,42 The other way round, adipocytes have been demonstrated to secrete as- yet-unidentified aldosterone-releasing factors which are able to induce synthe-sis and secretion of aldosterone in adrenal cells.43,44 This suggests that interaction of aldosterone with adipose tissue probably is bidirectional (Figure 2). Candidates for adipocyte-derived stimulation of aldosterone synthesis include TNF-α, C1q/TNF-α-re-lated protein, leptin, linoleic acid oxidative products, and interleukin-6.45-47 Moreover, free fatty acids can stimulate the adrenal production of aldosterone, reflecting another effect of metabolic status on aldosterone.48

Interaction between profibrotic effects of aldosterone and sodium statusInterestingly, aldosterone-induced target organ damage depends on sodium and volume status, even independent of blood pressure (Figure 2). Aldosterone-induced cardiac hypertrophy or cardiac fibrosis is completely prevented by a low sodium diet

Page 10: University of Groningen Towards personalized

Aldosterone and Renal Damage

61

3

in uninephrectomized rats.15,49 Effects of aldosterone on the severity of hypertension, endothelial dysfunction, and cardiac and vascular remodeling are sodium-dependent in stroke-prone spontaneously hypertensive rats.50 Moreover, aldosterone-induced renal inflammatory cell infiltration and vasculopathy only occurs during high sodium intake.51,52 In human, various conditions of hyperaldosteronism secondary to volume depletion, such as habitual low sodium intake in Yanomami Indians or Gitelman or Bartter syndrome with renal sodium loss, have no hypertension or vascular target organ damage.53,54 The other way round, in patients with resistant hypertension the del-eterious effects of high sodium intake on proteinuria are most pronounced in patients with the highest aldosterone levels.55 Taken together, these data suggest that aldo-sterone exerts adverse effects only when its serum concentration is inappropriate for sodium status (i.e. high aldosterone, high sodium). Sodium status might thus have an important role in sensitizing target organs to the damaging effects of aldosterone. The mechanisms of this sensitization are of great interest, in particular because the habitual sodium intake in most populations is high, both in general population and in patient cohorts with renal or cardiac disease.56,57

The classical association between aldosterone and sodium intake involves suppres-sion of aldosterone during high sodium intake (Figure 2), which facilitates renal excretion of the excess sodium. However, recent evidence showed, first, dissocia-tion of adequately suppressed circulating aldosterone levels with increased levels in target tissues in several conditions (Figure 2).7,58 In stroke-prone spontaneously hypertensive rats high sodium intake increased vascular aldosterone in association with vascular damage.58 In healthy rats high sodium intake increases cardiac aldo-sterone synthase activity and hence cardiac aldosterone, associated with cardiac hypertrophy and perivascular and interstitial fibrosis, independent of blood pressure, along with a reduction in circulating aldosterone levels.59 Interestingly, aging might represent another major condition with dissociation of circulating and tissue aldo-sterone effects. Aging is associated with lower circulating aldosterone levels,60 but higher vascular MR expression. Moreover, sensitivity for aldosterone-induced ERK 1/2 activation is enhanced in aged vascular smooth muscle cells,61 and MR blockade prevents age-associated upregulation of pro-inflammatory gene expression, support-ing relevance of MR and aldosterone signaling in the development of inflammation that is associated with arterial aging.61 Second, suppression of circulating aldosterone levels by high sodium intake is insufficient in several conditions, and associated with end organ damage. For instance, insufficient sodium-induced suppression of aldo-sterone is associated with left ventricular structural changes and cardiac dysfunction in hypertensive patients.62

Page 11: University of Groningen Towards personalized

Chapter 3

62

Adipocytes are likely to be involved in the impaired negative feed-back regulation by sodium loading on aldosterone, by release of aldosterone-producing factors. In obese spontaneously hypertensive rats (SHR), for instance, high sodium less effectively sup-presses aldosterone than in lean littermates.63 Renal target organ damage in obese SHRs, apparent from proteinuria, podocyte injury and advanced renal lesions, was dramatically ameliorated by MR blockade using eplerenone.63 Since nuclear MR con-tent and sgk-1 expression in the kidney were also increased in this study, MR signaling might underlie the sodium-induced renal damage in these obese SHRs.63

Genetic factors can also affect aldosterone regulation and its response to sodium. The ACE insertion/deletion (I/D) polymorphism affects not only ACE activity levels, but also levels of tissue aldosterone, with higher levels in DD genotype. In healthy DD homozygotes angiotensin I infusion (as a marker of increased tissue ACE activity and angiotensin II generation) led to increased responses of blood pressure during high sodium intake only,64 which is in line with other studies showing that high sodium conditions induce tissue ACE activity, and possibly aldosterone. This might have clini-cal relevance, as high sodium intake is associated with resistance to ACE inhibition in renal patients and healthy subjects with the DD genotype.65,66 Furthermore, patients with heart failure who have high circulating aldosterone levels despite treatment with ACE inhibition are more likely to have the DD genotype compared to patients lacking this aldosterone breakthrough.67

The mechanism of sodium dependence of aldosterone-induced renal and cardiovascu-lar disease continues to fascinate scientists and clinicians. The pathological processes by which aldosterone and high sodium intake promote target organ damage are undoubtedly multifactorial (Figure 2). Aldosterone excess increases sodium and fluid retention through its classical effects and increases blood pressure, which, in turn, mediates target organ damage. Moreover, blood pressure independent effects play a role with dissociation of low circulating and high tissue aldosterone levels during high sodium intake and possibly enhanced MR signaling. It is currently unknown why aldosterone-MR-induced downstream pathways leading to inflammatory cell activa-tion and pro-inflammatory gene upregulation are not activated in the absence of high sodium.51, 52

Aldosterone and effects of MR blockade on hypertension and the vasculatureSpironolactone and eplerenone are MR antagonists, which are currently available for the treatment of hypertension. The effects of MR blockade on blood pressure are clearly related to blocking of the genomic effects of aldosterone on renal sodium handling, with consequent a slow-onset natriuretic effect associated with potassium

Page 12: University of Groningen Towards personalized

Aldosterone and Renal Damage

63

3

retention. Interestingly, the effects of mineralocorticoid antagonism on blood pressure are not solely dependent on its diuretic effect. MR blockade during one year reduces stiffness of resistance arteries and decreases the collagen/elastin ratio as compared to treatment with the beta-blocker atenolol in hypertensive patients with similarly well controlled blood pressures.68 Moreover, pre-dialysis systolic blood pressure is decreased by spironolactone in anuric hemodialysis patients, without differences in serum potassium levels.69 This strongly suggests direct effects on vascular resistance.69 The vasculo-protective potential of MR blockade is supported by experimental studies showing reduced neointima formation after coronary angioplasty and coronary stent-ing in pigs.70,71 Furthermore, overexpression of aldosterone synthase in the heart is associated with coronary vascular dysfunction without altering cardiac structure and function in transgenic mice, suggesting direct vascular effects of aldosterone.72 Direct effects of aldosterone on the vasculature are further supported by experimental data in rats on a high sodium diet infused with angiotensin II and co-treated with L-NAME to inhibit nitric oxide synthesis.73 Adrenalectomy or MR blockade prevented the asso-ciated myocardial necrosis, proteinuria, and vascular lesions without altering systolic blood pressure. This protective effect was lost by aldosterone infusion, suggesting that the noxious effects of NO-depletion plus angiotensin II infusion on a high sodium diet are mediated, at least in part, by aldosterone independent of its classic effects.73

We recently demonstrated that MR blockade by spironolactone significantly amelio-rates transplant vasculopathy (TV) in renal chronic transplant dysfunction (CTD) in rats.74 TV is characterized by severe neo-intima formation, which leads to impaired perfusion and subsequent allograft dysfunction with reduced survival.75 Spironolac-tone ameliorated the development of TV by reducing the number of affected arteries (Figure 3).74 This suggests that MR activation plays a role in the initiation rather than the progression of neointima formation, possibly by protecting against endothelial activation. The latter plays an important role in the initiation of TV, and MR blockade has been shown to ameliorate aldosterone-associated endothelial activation and dys-function.76-80

Aldosterone and effects of MR blockade on cardiac fibrosis and heart failureDirect effects of aldosterone on the heart were suggested already more than a decade ago by the strong correlation of aldosterone levels with left ventricular mass in patients with hypertension, independent of blood pressure.81 Decreased compliance of the aorta was shown to inversely correlate with aldosterone levels in patients with heart failure.82 Interestingly, a recent randomized controlled study showed left ventricu-lar mass reduction and improved arterial stiffness by MR blockade in non-diabetic patients with stage 2 and 3 CKD.83 These effects occurred along with a reduction in systolic blood pressure (placebo vs. spironolactone: 124 vs. 119 mmHg respectively).

Page 13: University of Groningen Towards personalized

Chapter 3

64

Figure 3. Spironolactone ameliorates transplant vasculopathy (TV) by reducing the number of affected arteries in experimental chronic transplant dysfunction (CTD). Effects of MR blockade by spironolactone on TV in the Dark Agouti-to-Wistar Furth renal allograft trans-plant model of CTD. Treatment with spironolactone or vehicle daily by oral gavage was given from 2 days prior to transplantation (donors and recipients) throughout the exper-iment of (12 weeks, recipients). Dark Agouti-to-Dark Agouti isografts served as negative controls. [A, B] Photomicrograph of an artery from a vehicle-treated allograft without TV (A) and with TV (B). NI, neointima; arrow head indicates elastin. [C] Percentage of affected arteries present within small (25-49 µm), medium 25 (50-99 µm) and large (≥100 µm) diameter arteries. [D] Total TV index, expressed as the mean occlusion in all elastin-posi-tive arteries with a diameter ≥100 µm. [E] Percent occlusion in TV affected arteries with a with a diameter ≥100 µm. Data are presented as mean ± SE. *P < 0.05 vs. vehicle-treated allografts. Reprinted from reference.74

Page 14: University of Groningen Towards personalized

Aldosterone and Renal Damage

65

3

However, these effects on blood pressure were much weaker than expected based on the observed effects on arterial stiffness. So, it is plausible that blockade of the cardiac and vascular MR reduced aldosterone-induced inflammation, fibrosis and hypertrophy in a blood pressure-independent manner.84

Increased aldosterone levels are associated with the development of cardiac fibrosis.85 Atrial fibrosis causes local conduction disturbances, which may lead to the develop-ment of arrhythmia. Atrial fibrillation is the most frequently observed arrhythmia, which accounts for substantial morbidity and mortality.86 Aldosterone levels are increased in patients with persistent atrial fibrillation.87 In an experimental study, MR blockade by eplerenone not only prevented development of de novo cardiac fibrosis and inflammation, but also reduced established mineralocorticoid-induced cardiac fibrosis and inflammation.88 It would be of great interest to investigate whether MR blockade can reduce conduction abnormalities by reducing atrial fibrosis. This would offer new approaches for anti-arrhythmic pharmacotherapy in atrial fibrillation.

The MR is present in the heart and aldosterone is produced locally in the heart at low levels.6 However, after myocardial infarction local aldosterone production increases, contributing to cardiac fibrosis,85 which may lead to heart failure. Aldosterone synthase mRNA expression is upregulated in ventricles of patients with heart failure89 and/or hypertrophic cardiomyopathy.90 These observations suggest that cardiac aldosterone may indeed have harmful effects in the myocardium. A recent study highlights the importance of macrophages in cardiac inflammation and fibrosis and assigns a key role to the MR in macrophages.91 Using macrophage-specific MR knockout mice the authors demonstrated that macrophages infiltrating the heart need to express the MR in order to exert their detrimental effects on blood pressure and cardiac fibrosis.91

In line with the cardiovascular effects of aldosterone-MR mentioned above, two large randomized controlled clinical trials (the RALES [randomized aldactone evaluation study] and EPHESUS [eplerenone post-acute myocardial infarction heart failure efficacy and survival study]) have demonstrated the efficacy of MR blockade on top of stan-dard medical treatment in patients with heart failure.4,5 Treatment with spironolactone or eplerenone improved patient survival and reduced cardiovascular events inde-pendently of blood pressure.4,5 Results from a recent post-hoc biomarker study from EPHESUS suggest that these beneficial effects of MR blockade may be due to reduced cardiac extracellular matrix remodeling.92 As compared to placebo-treatment, patients treated with eplerenone had lower levels of propeptide of type I procollagen and the amino-terminal propeptide of type III procollagen, which are biomarkers of type I and III collagen synthesis. In contrast, there was no difference in levels of biomarkers for

Page 15: University of Groningen Towards personalized

Chapter 3

66

collagen degradation. Thus MR blockade may prevent cardiac matrix remodeling rather than facilitating degradation of existing deposited collagen. In addition, effects on baro-reflex sensitivity and the autonomic nervous system may be involved in the improved survival in patients with heart failure during MR blockade. Aldosterone is a risk factor for decreased heart rate variability and prolonged QT interval, factors that predict sudden cardiac death.93-95 MR blockade not only reduces myocardial collagen turnover but also improves time-domain heart rate variability in patients with heart failure.94

Direct renal effects of aldosteroneThere is increasing evidence that aldosterone can be directly involved in the develop-ment and progression of renal disease via non-epithelial MR-mediated effects.20,26,28,96 In vitro studies showed that aldosterone increases the production of TGF-β, ROS, PAI-1 and collagen which can be abolished by MR blockade. Studies in different rat models support a role for non-hemodynamic effects of aldosterone and the MR in renal damage. Uninephrectomized rats develop progressive proteinuria when they are continuously infused with aldosterone and fed a high-salt diet. These effects could be prevented by MR blockade or tempol, a ROS scavenger.29 In the same study it was shown that addition of aldosterone to cultured podocytes induced sgk-1 via the MR, which could be prevented by the addition of tempol,29 suggesting that aldosterone-in-duced podocyte injury is in part caused by an increase in oxidative stress.

MR blockade slows the progression of glomerulosclerosis, and can sometimes induce regression of existing glomerulosclerosis in rats with 5/6 nephrectomy.20 Despite these glomeruloprotective effects, proteinuria continued to increase in this study, demon-strating that the effects of MR blockade on proteinuria and structural damage can be discordant.20 It may also be that podocyte injury persists even after regression of glomerulosclerosis. Podocytes have only limited regenerative capacity and may have less ability to heal and/or require longer time for regression of injury than remodeling of the extracellular matrix of sclerotic lesions. We showed that aldosterone induces heparanase expression in podocytes, which could be blocked by spironolactone.97 Heparanase is an endo-β(1-4)-D-glucuronidase that cleaves heparan sulphate side chains, which is involved in tissue remodeling. Spironolactone reduced glomerular heparanase expression and partially restored heparan sulphate expression in the glo-merular basement membrane of rats with adriamycin nephropathy.97,98 Despite these beneficial effects on structural damage, MR blockade did not affect proteinuria,97,98 again underlining that effects of MR blockade on proteinuria and tissue remodeling can be discordant. MR antagonists also markedly ameliorated tubulo-interstitial injury and/or glomerular damage in several models of nephropathy including spontaneously hypertensive stroke-prone rats without effects on systemic blood pressure or volume

Page 16: University of Groningen Towards personalized

Aldosterone and Renal Damage

67

3

status.26,99 Moreover, exogenous aldosterone infusion completely reversed the reno-protective effects of ACE inhibition in this model.26 Furthermore, in the remnant kidney model in which rats develop hyperaldosteronism, the renoprotective effect of dual RAAS blockade by the combination of ACE inhibition and ARB was reversed by exoge-nous aldosterone infusion.96

Interestingly, in adriamycin-induced proteinuria mono-therapy MR blockade was inef-fective, but in combination with ACE inhibition it effectively reduced blood pressure, proteinuria, interstitial and glomerular damage.98 It is unclear whether the added effect of MR blockade and ACE inhibition is due to the diuretic effect of MR blockade or by a direct anti-fibrotic effect. In contrast, we found progressive interstitial fibrosis during ACE inhibition in a separate study where dietary sodium restriction was used to induce the volume depletion required to maximize the effects of ACE inhibition, despite similarly effective reduction of blood pressure, proteinuria and glomerulosclerosis.100 Although a head-to-head comparison of these two different studies is not warranted, the difference in effect on interstitial fibrosis, all other components of the therapy response being equal, is remarkable (Figure 4). Similarly low systolic blood pressure can thus be tolerated without inducing hypoxia-driven interstitial fibrosis, depending on the treatment regimen by which it is achieved.

The mechanisms underlying the adverse effects of aldosterone on the kidney are com-plex and multifactorial. First, aldosterone increases sodium and fluid retention and accordingly increases blood pressure, which, in turn, mediates target organ damage. Volume overload, moreover, blunts the efficacy of RAAS blockade.57,101,102 Aldosterone also increases intraglomerular pressure due to direct effects on the renal microcircu-lation. Aldosterone exerts a vasoconstrictor effect on the efferent arteriole and inhibits vasoconstriction in perfused rabbit afferent arterioles, which is abolished by spirono-lactone.103-105 This is consistent with the assumption of aldosterone-induced increased intraglomerular pressure. NO modulates the actions of aldosterone, as endothelial denudation and pharmacological blockade of eNOS increased the sensitivity of the afferent arterioles to aldosterone.105

MR blockade on top of treatment with ACE inhibition or ARB induces a larger decrease in urinary albumin excretion in diabetic patients.106,107 However, the antiproteinuric effects of MR blockade can be dissociated from circulating aldosterone levels. The syndrome of hyporenin-hypoaldosteronism in diabetic patients with autonomic dys-function is associated with cardiovascular damage. In these patients with long-standing diabetes the conversion of prorenin to renin is impaired by the diabetic kidney, result-ing in lower circulating aldosterone levels.108 These results suggest that MR blockade

Page 17: University of Groningen Towards personalized

Chapter 3

68

may be effective in preventing the progression of cardiovascular and renal compli-cations in patients with diabetic nephropathy who are known to have low circulating aldosterone levels. This may also suggest the importance of a tissue renin-angiotensin aldosterone system, with high tissue aldosterone levels. Not only diabetes, but also aging is associated with lower circulating aldosterone levels.60 However, there is no evidence that MR blockade would be less effective in the elderly.

Aldosterone breakthrough is associated with a poor response to anti-proteinuric treatment and an enhanced decline of renal function in patients with (diabetic) nephropathy.109,110 It is yet unknown whether this worse clinical prognosis of patients with aldosterone breakthrough is due to the noxious effects of aldosterone, induc-ing inflammation and fibrosis. In line with this hypothesis, studies in patients with chronic renal failure and early diabetic nephropathy show that MR blockade on top of ACE inhibition and/or ARB exerts added renoprotective effects.21,22,109-111 The reduc-tion of proteinuria by the addition of spironolactone to ACE inhibitors, ARBs or their combination, is related to aldosterone levels in non-diabetic proteinuric patients with CKD.21,109,111 This strongly suggests that aldosterone is a component of the renal damage that is associated with CKD and that its inhibition by ACE inhibitors, ARBs or their combination can be incomplete.21 Several small human interventional studies showed greater reductions of proteinuria and blood pressure when MR blockade was added to ACE inhibitors and/or ARBs.21,112,113 In spite of these encouraging results on proteinuria, long-term data on the efficacy of MR blockade on hard end points, for example the development of ESRD or patient survival, are still lacking in CKD.

Aldosterone breakthrough during RAAS blockade and intervention in sodium status Sodium restriction is known to reduce long-term cardiovascular risk,114 and is usually required to obtain an effective response to RAAS blockade.101,102 However, sodium restriction and diuretic treatment increase aldosterone levels not only in the untreated condition, but also during RAAS blockade by ARB (Figure 5, left panel, proteinuric patients)102 or by ACE inhibition (Figure 5, right panel, data from healthy volunteers).65 Short-term treatment with ACE inhibition reduces aldosterone during high and low sodium intake, due to decreased angiotensin II levels.65 However, this suppression cannot be sustained in 10% to 50% of patients treated with ACE inhibitors and/or ARBs.115 This aldosterone breakthrough particularly occurs during long-term treatment with ACE inhibition116 or during sodium restriction,117 which potentiates the adrenal response to angiotensin II.118 ARB treatment, on the other hand, did not affect aldo-sterone levels during either diet in proteinuric patients.102 Thus, during RAAS blockade aldosterone still responds to changes in volume status. Formerly, this preservation of the homeostatic response of the RAAS was considered neutral from the point of

Page 18: University of Groningen Towards personalized

Aldosterone and Renal Damage

69

3

Figure 4. Combined treatment with ACE inhibition and MR blockade on a normal sodium diet does not induce interstitial fibrosis despite similarly low blood pressure. Two differ-ent studies with proteinuria induced renal damage, in which treatment was started six weeks after adriamycin injection, are shown. ACE inhibition with lisinopril combined with MR blockade with spironolactone on a normal sodium diet, given for 12 weeks,98 similarly reduced blood pressure [A] and proteinuria [B] compared to ACE inhibition with lisinopril on a low sodium diet, given for 3 weeks.100 The difference in effect on interstitial fibrosis is remarkable [C]. Progressive interstitial fibrosis during ACE inhibition combined with a low sodium diet was seen, while only mild interstitial abnormalities were observed after ACE inhibition combined with MR blockade. Similarly low systolic blood pressure can thus be tolerated without inducing hypoxia-driven interstitial fibrosis, depending on the treat-ment regimen by which it is achieved. Abbreviations: BL, baseline; NS, normal sodium; ACEi, angiotensin converting enzyme inhibition; SPIR, spironolactone; LS, low sodium; SMA, smooth muscle actin. Data are presented as mean ± SD.

Page 19: University of Groningen Towards personalized

Chapter 3

70

view of therapeutic efficacy. Given the well-established pro-fibrotic effects of aldoste-rone23,27-30 and the association of aldosterone breakthrough with worse outcome,109,110 this point of view should be reconsidered. During RAAS blockade, co-treatment with sodium restriction and/or diuretic is usually required to obtain an effective therapy response of blood pressure and proteinuria.101,102 As shown here, this occurs at the expense of elevated circulating aldosterone. The clinical consequences of the rise in aldosterone induced by volume depletion during RAAS blockade are still unclear, but it might be reassuring that this is not a high sodium-high aldosterone condition. The eventual effects on long term outcome might be dependent on the concurrent effect on tissue aldosterone. In theory, overcoming aldosterone breakthrough induced by sodium restriction and/or diuretic treatment might contribute to long-term protection of heart, vasculature and kidneys. It would thus be of interest to investigate whether MR blockade is specifically beneficial during sodium restriction, or conversely, during conditions of volume excess. Moreover, it would be of interest to study whether the benefits of MR blockade derive from its diuretic effects or in its ability to modulate non-volume-mediated effects of aldosterone and whether its renoprotective effects on top of ACE inhibition outweigh the effects of conventional diuretics. Therefore, studies are warranted to investigate extracellular fluid volume in a cross-over design with and without MR blockade during a high and a low sodium diet in proteinuric patients on conventional RAAS blockade and compare these effects to hydrochlorothiazide. This is topic of a current study (ESCAPE study, Dutch Trial Register, NTR2133).

Pharmacology of MR blocking agents and effects on potassium homeostasisTwo different MR blocking agents exist, spironolactone and eplerenone. Both are steroid analogues with structural similarity to aldosterone, thereby functioning as com-petitive antagonists. Spironolactone is absorbed for 80-90% and is rapidly metabolized in the liver into a number of metabolites, including canrenone, 7α-methylspironolac-tone and 6β- hydroxy-7α-methylspironolactone, having mean half-lives of 1.4, 16.5, 13.8, and 15 hours respectively. The onset of action for spironolactone is slow, with a peak response on natriuresis occurring 48 hours after the first dose. In eplerenone the 17-α-thioacylgroup of spironolactone is replaced with a carbomethoxy group.119 This confers selectivity of eplerenone to the MR, translating into less affinity of epler-enone for androgen and progestogenic receptors compared to spironolactone. This selectivity results in less endocrine side effects including gynecomastia, menstrual irregularities, loss of libido and impotence. No active metabolites have been identified for eplerenone,119 resulting in a shorter effective half-life and therefore quicker time to peak response than spironolactone. The maximum concentration of eplerenone is reached in 1.3 hours.119 The primary route of elimination is through CYP4503A4-medi-ated metabolism, its elimination half-life being 4-6 hours.119

Page 20: University of Groningen Towards personalized

Aldosterone and Renal Damage

71

3

Figure 5. Circulating aldosterone levels are increased by intervention in sodium status. The left panel shows circulating aldosterone levels in proteinuric patients treated during 6 weeks with placebo, ARB (losartan 100 mg/d) and ARB plus diuretic (losartan/hydrochlo-rothiazide 100/25 mg/d) on a high (200 mmol/d) and a low sodium diet (50 mmol/d) in a randomized double-blind cross-over trial.102 ARB treatment did not affect aldosterone levels during either diet. Sodium restriction and diuretic treatment both increased aldo-sterone levels not only in the untreated condition, but also during RAAS blockade by ARB. The right panel shows circulating aldosterone levels in healthy male volunteers treated during one week with ACE inhibition (enalapril 20 mg/d) on a high (200 mmol/d) and a low sodium diet (50 mmol/d) in a randomized double-blind cross-over trial.65 Short-term treatment with ACE inhibition reduced circulating aldosterone levels during both sodium intakes. Sodium restriction increased aldosterone levels during placebo and during RAAS blockade by ACE inhibition. Abbreviations: HCT, hydrochlorothiazide; ACEi, angiotensin converting enzyme inhibition; LS, low sodium diet. #p<0.05 vs. same treatment on high sodium (effect of LS), †p<0.05 vs. losartan treatment on same diet (effect of HCT), ‡p<0.05 vs. placebo on same diet, *p<0.05 vs. losartan+HCT high sodium (comparison between addition of low sodium and HCT to losartan). Data are presented as mean ± SE.

An important risk of MR blockade is hyperkalemia, which can induce life threatening cardiac arrhythmias. Impaired renal function is the most important risk factor for hyperkalemia. Many patients with hypertension, heart failure or kidney disease are already treated with ACE inhibitors or ARBs, which further increases the risk for MR

Page 21: University of Groningen Towards personalized

Chapter 3

72

blockade-induced hyperkalemia. Both spironolactone and eplerenone are associated with dose-related increases in serum potassium values. Spironolactone dosed 25, 50, and 75 mg/d induced hyperkalemia (serum potassium > 5.5 mmol/L) in 13, 20 and 24% of the participants of a dose-finding study for RALES.120 Experiences with the RALES and EPHESUS trials on hyperkalemia are somewhat reassuring. Spironolactone dosed 25 mg on top of ACE inhibition increased serum potassium levels with 0.3 mmol/L in the RALES study.4 There were only 10/841 and 14/822 cases of serious hyperkalemia in the placebo- and spironolactone-treated group, respectively.4 Importantly, this study excluded patients with renal function impairment (serum creatinine > 221 µmol/L) or a baseline serum potassium level of > 5.0 mmol/L. In the EPHESUS study potassium levels increased significantly more during eplerenone 50 mg (0.3 mmol/L) than during placebo (0.2 mmol/L) and serious hyperkalemia (serum potassium > 6.0 mmol/L) was more prevalent during eplerenone as well (126/3301, 3.9% vs. 180/3307, 5.5%). The incidence of hyperkalemia was higher in patients with renal function impairment (cre-atinine clearance < 50 mL/min); eplerenone 10.1% vs. placebo 5.9%. A recent post-hoc analysis of the EPHESUS study demonstrates that eplerenone still exerts beneficial effects on mortality regardless of baseline risk factors for development of hyperka-lemia (serum potassium > 6.0 mmol/L).121 The four independent baseline predictors of hyperkalemia were baseline potassium greater than the median of 4.3 mmol/L, impaired renal function (eGFR < 60 mL/min/1.73m2), history of diabetes mellitus, and prior use of antiarrhythmic agents.121

To obtain beneficial effects of MR blockade, it is necessary to ensure its safe use. According to current guidelines,122,123 this involves measuring both serum potassium and estimating renal function either by eGFR or creatinine clearance before initiating treatment with MR blockade. MR blocking agents should not be prescribed to patients with serum potassium greater than 5.0 mmol/L and/or impaired renal function as defined by serum creatinine > 221 µmol/L and/or creatinine clearance < 30 mL/min. Periodical monitoring of serum potassium and renal function is recommended with dose adjustment of the MR blocking agent as necessary.

Page 22: University of Groningen Towards personalized

Aldosterone and Renal Damage

73

3

CONCLUSIONS

Prevailing evidence suggests that aldosterone is far more than merely a salt-and-wa-ter hormone. In addition to its mineralocorticoid function, aldosterone has a number of non-classical, MR mediated actions, including tissue remodeling, modulation of vascular tone, and stimulating inflammation and fibrosis. All these actions may fuel progression of cardiovascular and renal end organ damage. Aldosterone break-through during RAAS blockade with ACE inhibition and/or ARB may explain why this treatment regimen only confers partial cardiovascular and renal protection. We have delineated that tissue aldosterone rather than its circulating level is relevant in this respect and that the former and latter do not necessarily run in parallel. Of major inter-est, sodium status appears to be an important determinant of the end-organ effects of aldosterone. Aldosterone is deleterious only if inappropriately high for its sodium status i.e. high aldosterone and high sodium.

MR blockade is a valuable strategy, which has potency to halt the progressive end organ damage as observed during current treatments. In heart failure, MR blockade on top of RAAS blockade reduces hard clinical endpoints. Despite encouraging results on the intermediate endpoint proteinuria, long-term data on the efficacy and safety of MR blockade in preventing dialysis and/or cardiovascular endpoints in CKD are still lacking. It is obligatory that future clinical studies on the effects of MR blockade on end-organ damage take into account the sodium status.

Page 23: University of Groningen Towards personalized

Chapter 3

74

REFERENCES

1. Schrier RW: Aldosterone ‘escape’ vs ‘breakthrough’. Nat Rev Nephrol 6(2):61, 2010

2. Athyros VG, Mikhailidis DP, Kakafika AI, Tziomalos K, Karagiannis A: Angiotensin II reactivation and

aldosterone escape phenomena in renin-angiotensin-aldosterone system blockade: Is oral renin

inhibition the solution? Expert Opin Pharmacother 8(5):529-535, 2007

3. Navis G, de Jong PE, Donker AJ, van der Hem GK, de Zeeuw D: Moderate sodium restriction in

hypertensive subjects: Renal effects of ACE-inhibition. Kidney Int 31(3):815-819, 1987

4. 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 341(10):709-717, 1999

5. Pitt B, Remme W, Zannad F, et al: Eplerenone, a selective aldosterone blocker, in patients with left

ventricular dysfunction after myocardial infarction. N Engl J Med 348(14):1309-1321, 2003

6. Silvestre JS, Robert V, Heymes C, et al: Myocardial production of aldosterone and corticosterone in

the rat. physiological regulation. J Biol Chem 273(9):4883-4891, 1998

7. Takeda Y: Role of cardiovascular aldosterone in hypertension. Curr Med Chem Cardiovasc Hematol

Agents 3(3):261-266, 2005

8. Marney AM, Brown NJ: Aldosterone and end-organ damage. Clin Sci (Lond) 113(6):267-278, 2007

9. Arriza JL, Weinberger C, Cerelli G, et al: Cloning of human mineralocorticoid receptor complementary

DNA: Structural and functional kinship with the glucocorticoid receptor. Science 237(4812):268-275,

1987

10. Anagnostis P, Karagiannis A, Tziomalos K, Athyros VG, Kita M, Mikhailidis DP: Endocrine hyperten-

sion: Diagnosis and management of a complex clinical entity. Curr Vasc Pharmacol 8(5):646-660,

2010

11. Bocchi B, Kenouch S, Lamarre-Cliche M, et al: Impaired 11-beta hydroxysteroid dehydrogenase

type 2 activity in sweat gland ducts in human essential hypertension. Hypertension 43(4):803-808,

2004

12. Ostreicher I, Almeida JR, Campean V, et al: Changes in 11beta-hydroxysteroid dehydrogenase type

2 expression in a low-protein rat model of intrauterine growth restriction. Nephrol Dial Transplant

25(10):3195-3203, 2010

13. Stowasser M: Update in primary aldosteronism. J Clin Endocrinol Metab 94(10):3623-3630, 2009

14. Rossi GP, Bernini G, Desideri G, et al: Renal damage in primary aldosteronism: Results of the PAPY

study. Hypertension 48(2):232-238, 2006

15. Brilla CG, Weber KT: Mineralocorticoid excess, dietary sodium, and myocardial fibrosis. J Lab Clin

Med 120(6):893-901, 1992

16. Funder JW: Minireview: Aldosterone and mineralocorticoid receptors: Past, present, and future.

Endocrinology 151(11):5098-5102, 2010

17. Giacchetti G, Ronconi V, Turchi F, et al: Aldosterone as a key mediator of the cardiometabolic syn-

drome in primary aldosteronism: An observational study. J Hypertens 25(1):177-186, 2007

18. Young WF: Primary aldosteronism: Renaissance of a syndrome. Clin Endocrinol (Oxf) 66(5):607-618,

2007

19. Karagiannis A, Tziomalos K, Kakafika AI, Athyros VG, Harsoulis F, Mikhailidis DP: Medical treatment

as an alternative to adrenalectomy in patients with aldosterone-producing adenomas. Endocr Relat

Cancer 15(3):693-700, 2008

20. Aldigier JC, Kanjanbuch T, Ma LJ, Brown NJ, Fogo AB: Regression of existing glomerulosclerosis by

Page 24: University of Groningen Towards personalized

Aldosterone and Renal Damage

75

3

inhibition of aldosterone. J Am Soc Nephrol 16(11):3306-3314, 2005

21. Bianchi S, Bigazzi R, Campese VM: Long-term effects of spironolactone on proteinuria and kidney

function in patients with chronic kidney disease. Kidney Int 70(12):2116-2123, 2006

22. Chrysostomou A, Becker G: Spironolactone in addition to ACE inhibition to reduce proteinuria in

patients with chronic renal disease. N Engl J Med 345(12):925-926, 2001

23. Miyata K, Rahman M, Shokoji T, et al: Aldosterone stimulates reactive oxygen species production

through activation of NADPH oxidase in rat mesangial cells. J Am Soc Nephrol 16(10):2906-2912,

2005

24. Fiebeler A, Muller DN, Shagdarsuren E, Luft FC: Aldosterone, mineralocorticoid receptors, and

vascular inflammation. Curr Opin Nephrol Hypertens 16(2):134-142, 2007

25. Funder JW: Aldosterone, salt and cardiac fibrosis. Clin Exp Hypertens 19(5-6):885-899, 1997

26. Rocha R, Chander PN, Zuckerman A, Stier CT,Jr: Role of aldosterone in renal vascular injury in

stroke-prone hypertensive rats. Hypertension 33(1 Pt 2):232-237, 1999

27. Oberleithner H, Riethmuller C, Ludwig T, Hausberg M, Schillers H: Aldosterone remodels human

endothelium. Acta Physiol (Oxf) 187(1-2):305-312, 2006

28. Del Vecchio L, Procaccio M, Vigano S, Cusi D: Mechanisms of disease: The role of aldosterone in

kidney damage and clinical benefits of its blockade. Nat Clin Pract Nephrol 3(1):42-49, 2007

29. Shibata S, Nagase M, Yoshida S, Kawachi H, Fujita T: Podocyte as the target for aldosterone: Roles

of oxidative stress and Sgk1. Hypertension 49(2):355-364, 2007

30. Nagai Y, Miyata K, Sun GP, et al: Aldosterone stimulates collagen gene expression and synthesis

via activation of ERK1/2 in rat renal fibroblasts. Hypertension 46(4):1039-1045, 2005

31. van Leeuwen N, Caprio M, Blaya C, et al: The functional c.-2G>C variant of the mineralocorticoid

receptor modulates blood pressure, renin, and aldosterone levels. Hypertension 56(5):995-1002,

2010

32. Briet M, Schiffrin EL: Aldosterone: Effects on the kidney and cardiovascular system. Nat Rev Nephrol

6(5):261-273, 2010

33. Conn JW: Hypertension, the potassium ion and impaired carbohydrate tolerance. N Engl J Med

273(21):1135-1143, 1965

34. Bochud M, Nussberger J, Bovet P, et al: Plasma aldosterone is independently associated with the

metabolic syndrome. Hypertension 48(2):239-245, 2006

35. Fallo F, Della Mea P, Sonino N, et al: Adiponectin and insulin sensitivity in primary aldosteronism.

Am J Hypertens 20(8):855-861, 2007

36. Catena C, Lapenna R, Baroselli S, et al: Insulin sensitivity in patients with primary aldosteronism: A

follow-up study. J Clin Endocrinol Metab 91(9):3457-3463, 2006

37. Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC: Pivotal role of the mineralo-

corticoid receptor in corticosteroid-induced adipogenesis. FASEB J 21(9):2185-2194, 2007

38. Rondinone CM, Rodbard D, Baker ME: Aldosterone stimulated differentiation of mouse 3T3-L1 cells

into adipocytes. Endocrinology 132(6):2421-2426, 1993

39. Guo C, Ricchiuti V, Lian BQ, et al: Mineralocorticoid receptor blockade reverses obesity-related

changes in expression of adiponectin, peroxisome proliferator-activated receptor-gamma, and

proinflammatory adipokines. Circulation 117(17):2253-2261, 2008

40. Lastra G, Whaley-Connell A, Manrique C, et al: Low-dose spironolactone reduces reactive oxygen

species generation and improves insulin-stimulated glucose transport in skeletal muscle in the

TG(mRen2)27 rat. Am J Physiol Endocrinol Metab 295(1):E110-6, 2008

41. Mosso LM, Carvajal CA, Maiz A, et al: A possible association between primary aldosteronism and

a lower beta-cell function. J Hypertens 25(10):2125-2130, 2007

Page 25: University of Groningen Towards personalized

Chapter 3

76

42. Yamashita R, Kikuchi T, Mori Y, et al: Aldosterone stimulates gene expression of hepatic gluconeo-

genic enzymes through the glucocorticoid receptor in a manner independent of the protein kinase

B cascade. Endocr J 51(2):243-251, 2004

43. Ehrhart-Bornstein M, Lamounier-Zepter V, Schraven A, et al: Human adipocytes secrete mineralo-

corticoid-releasing factors. Proc Natl Acad Sci U S A 100(24):14211-14216, 2003

44. Ehrhart-Bornstein M, Arakelyan K, Krug AW, Scherbaum WA, Bornstein SR: Fat cells may be the

obesity-hypertension link: Human adipogenic factors stimulate aldosterone secretion from adre-

nocortical cells. Endocr Res 30(4):865-870, 2004

45. Fujita T: Mineralocorticoid receptors, salt-sensitive hypertension, and metabolic syndrome. Hyper-

tension 55(4):813-818, 2010

46. Goodfriend TL, Ball DL, Egan BM, Campbell WB, Nithipatikom K: Epoxy-keto derivative of linoleic

acid stimulates aldosterone secretion. Hypertension 43(2):358-363, 2004

47. Jeon JH, Kim KY, Kim JH, et al: A novel adipokine CTRP1 stimulates aldosterone production. FASEB

J 22(5):1502-1511, 2008

48. Sowers JR, Whaley-Connell A, Epstein M: Narrative review: The emerging clinical implications of

the role of aldosterone in the metabolic syndrome and resistant hypertension. Ann Intern Med

150(11):776-783, 2009

49. Wang Q, Clement S, Gabbiani G, et al: Chronic hyperaldosteronism in a transgenic mouse model

fails to induce cardiac remodeling and fibrosis under a normal-salt diet. Am J Physiol Renal Physiol

286(6):F1178-84, 2004

50. Endemann DH, Touyz RM, Iglarz M, Savoia C, Schiffrin EL: Eplerenone prevents salt-induced vascular

remodeling and cardiac fibrosis in stroke-prone spontaneously hypertensive rats. Hypertension

43(6):1252-1257, 2004

51. Blasi ER, Rocha R, Rudolph AE, Blomme EA, Polly ML, McMahon EG: Aldosterone/salt induces renal

inflammation and fibrosis in hypertensive rats. Kidney Int 63(5):1791-1800, 2003

52. Hartner A, Porst M, Gauer S, Prols F, Veelken R, Hilgers KF: Glomerular osteopontin expression and

macrophage infiltration in glomerulosclerosis of DOCA-salt rats. Am J Kidney Dis 38(1):153-164,

2001

53. Nowaczynski W, Oliver WJ, Neel JV: Serum aldosterone and protein-binding variables in yanomama

indians: A no-salt culture as compared to partially acculturated guaymi indians. Clin Physiol Bio-

chem 3(6):289-306, 1985

54. Calo LA, Puato M, Schiavo S, et al: 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 23(9):2804-2809, 2008

55. Pimenta E, Gaddam KK, Pratt-Ubunama MN, et al: Relation of dietary salt and aldosterone to urinary

protein excretion in subjects with resistant hypertension. Hypertension 51(2):339-344, 2008

56. Bibbins-Domingo K, Chertow GM, Coxson PG, et al: Projected effect of dietary salt reductions on

future cardiovascular disease. N Engl J Med 362(7):590-599, 2010

57. Krikken JA, Laverman GD, Navis G: Benefits of dietary sodium restriction in the management of

chronic kidney disease. Curr Opin Nephrol Hypertens 18(6):531-538, 2009

58. Takeda Y, Yoneda T, Demura M, Furukawa K, Miyamori I, Mabuchi H: Effects of high sodium intake

on cardiovascular aldosterone synthesis in stroke-prone spontaneously hypertensive rats. J Hyper-

tens 19(3 Pt 2):635-639, 2001

59. Takeda Y, Yoneda T, Demura M, Miyamori I, Mabuchi H: Sodium-induced cardiac aldosterone

synthesis causes cardiac hypertrophy. Endocrinology 141(5):1901-1904, 2000

60. Epstein M: Aging and the kidney. J Am Soc Nephrol 7(8):1106-1122, 1996

Page 26: University of Groningen Towards personalized

Aldosterone and Renal Damage

77

3

61. Krug AW, Allenhofer L, Monticone R, et al: Elevated mineralocorticoid receptor activity in aged rat

vascular smooth muscle cells promotes a proinflammatory phenotype via extracellular signal-regu-

lated kinase 1/2 mitogen-activated protein kinase and epidermal growth factor receptor-dependent

pathways. Hypertension 55(6):1476-1483, 2010

62. Schlaich MP, Schobel HP, Hilgers K, Schmieder RE: Impact of aldosterone on left ventricular

structure and function in young normotensive and mildly hypertensive subjects. Am J Cardiol

85(10):1199-1206, 2000

63. Nagase M, Matsui H, Shibata S, Gotoda T, Fujita T: Salt-induced nephropathy in obese sponta-

neously hypertensive rats via paradoxical activation of the mineralocorticoid receptor: Role of

oxidative stress. Hypertension 50(5):877-883, 2007

64. van der Kleij FG, de Jong PE, Henning RH, de Zeeuw D, Navis G: Enhanced responses of blood

pressure, renal function, and aldosterone to angiotensin I in the DD genotype are blunted by low

sodium intake. J Am Soc Nephrol 13(4):1025-1033, 2002

65. Lely AT, Heerspink HJ, Zuurman M, et al: Response to angiotensin-converting enzyme inhibition is

selectively blunted by high sodium in angiotensin-converting enzyme DD genotype: Evidence for

gene-environment interaction in healthy volunteers. J Hypertens 28(12):2414-2421, 2010

66. van der Kleij FG, Schmidt A, Navis GJ, et al: Angiotensin converting enzyme insertion/deletion poly-

morphism and short-term renal response to ACE inhibition: Role of sodium status. Kidney Int Suppl

63:S23-6, 1997

67. Cicoira M, Zanolla L, Rossi A, et al: Failure of aldosterone suppression despite angiotensin-con-

verting enzyme (ACE) inhibitor administration in chronic heart failure is associated with ACE DD

genotype. J Am Coll Cardiol 37(7):1808-1812, 2001

68. Savoia C, Touyz RM, Amiri F, Schiffrin EL: Selective mineralocorticoid receptor blocker eplerenone

reduces resistance artery stiffness in hypertensive patients. Hypertension 51(2):432-439, 2008

69. Gross E, Rothstein M, Dombek S, Juknis HI: Effect of spironolactone on blood pressure and the

renin-angiotensin-aldosterone system in oligo-anuric hemodialysis patients. Am J Kidney Dis

46(1):94-101, 2005

70. Wakabayashi K, Suzuki H, Sato T, Iso Y, Katagiri T, Takeyama Y: Eplerenone suppresses neointimal

formation after coronary stent implantation in swine. Int J Cardiol 107(2):260-266, 2006

71. Ward MR, Kanellakis P, Ramsey D, Funder J, Bobik A: Eplerenone suppresses constrictive remodeling

and collagen accumulation after angioplasty in porcine coronary arteries. Circulation 104(4):467-

472, 2001

72. Garnier A, Bendall JK, Fuchs S, et al: Cardiac specific increase in aldosterone production induces

coronary dysfunction in aldosterone synthase-transgenic mice. Circulation 110(13):1819-1825, 2004

73. Rocha R, Stier CT,Jr, Kifor I, et al: Aldosterone: A mediator of myocardial necrosis and renal arteri-

opathy. Endocrinology 141(10):3871-3878, 2000

74. Waanders F, Rienstra H, Boer MW, et al: Spironolactone ameliorates transplant vasculopathy in

renal chronic transplant dysfunction in rats. Am J Physiol Renal Physiol 296(5):F1072-9, 2009

75. Rienstra H, Boersema M, Onuta G, et al: Donor and recipient origin of mesenchymal and endothelial

cells in chronic renal allograft remodeling. Am J Transplant 9(3):463-472, 2009

76. Ross R, Glomset J, Harker L: Response to injury and atherogenesis. Am J Pathol 86(3):675-684, 1977

77. Farquharson CA, Struthers AD: Aldosterone induces acute endothelial dysfunction in vivo in

humans: Evidence for an aldosterone-induced vasculopathy. Clin Sci (Lond) 103(4):425-431, 2002

78. Oberleithner H: Aldosterone makes human endothelium stiff and vulnerable. Kidney Int 67(5):1680-

1682, 2005

79. Farquharson CA, Struthers AD: Spironolactone increases nitric oxide bioactivity, improves endo-

Page 27: University of Groningen Towards personalized

Chapter 3

78

thelial vasodilator dysfunction, and suppresses vascular angiotensin I/angiotensin II conversion in

patients with chronic heart failure. Circulation 101(6):594-597, 2000

80. Macdonald JE, Kennedy N, Struthers AD: Effects of spironolactone on endothelial function, vascular

angiotensin converting enzyme activity, and other prognostic markers in patients with mild heart

failure already taking optimal treatment. Heart 90(7):765-770, 2004

81. Duprez DA, Bauwens FR, De Buyzere ML, et al: Influence of arterial blood pressure and aldosterone

on left ventricular hypertrophy in moderate essential hypertension. Am J Cardiol 71(3):17A-20A,

1993

82. Duprez DA, De Buyzere ML, Rietzschel ER, et al: Inverse relationship between aldosterone and large

artery compliance in chronically treated heart failure patients. Eur Heart J 19(9):1371-1376, 1998

83. Edwards NC, Steeds RP, Stewart PM, Ferro CJ, Townend JN: Effect of spironolactone on left ventric-

ular mass and aortic stiffness in early-stage chronic kidney disease: A randomized controlled trial.

J Am Coll Cardiol 54(6):505-512, 2009

84. Rocha R, Rudolph AE, Frierdich GE, et al: Aldosterone induces a vascular inflammatory phenotype

in the rat heart. Am J Physiol Heart Circ Physiol 283(5):H1802-10, 2002

85. Silvestre JS, Heymes C, Oubenaissa A, et al: Activation of cardiac aldosterone production in rat myo-

cardial infarction: Effect of angiotensin II receptor blockade and role in cardiac fibrosis. Circulation

99(20):2694-2701, 1999

86. Cameron A, Schwartz MJ, Kronmal RA, Kosinski AS: Prevalence and significance of atrial fibrillation

in coronary artery disease (CASS registry). Am J Cardiol 61(10):714-717, 1988

87. Vasan RS, Evans JC, Larson MG, et al: Serum aldosterone and the incidence of hypertension in

nonhypertensive persons. N Engl J Med 351(1):33-41, 2004

88. Young M, Funder JW: Eplerenone, but not steroid withdrawal, reverses cardiac fibrosis in deoxy-

corticosterone/salt-treated rats. Endocrinology 145(7):3153-3157, 2004

89. Yoshimura M, Nakamura S, Ito T, et al: Expression of aldosterone synthase gene in failing human

heart: Quantitative analysis using modified real-time polymerase chain reaction. J Clin Endocrinol

Metab 87(8):3936-3940, 2002

90. Tsybouleva N, Zhang L, Chen S, et al: Aldosterone, through novel signaling proteins, is a funda-

mental molecular bridge between the genetic defect and the cardiac phenotype of hypertrophic

cardiomyopathy. Circulation 109(10):1284-1291, 2004

91. Rickard AJ, Morgan J, Tesch G, Funder JW, Fuller PJ, Young MJ: Deletion of mineralocorticoid recep-

tors from macrophages protects against deoxycorticosterone/salt-induced cardiac fibrosis and

increased blood pressure. Hypertension 54(3):537-543, 2009

92. Iraqi W, Rossignol P, Angioi M, et al: Extracellular cardiac matrix biomarkers in patients with acute

myocardial infarction complicated by left ventricular dysfunction and heart failure: Insights from

the eplerenone post-acute myocardial infarction heart failure efficacy and survival study (EPHESUS)

study. Circulation 119(18):2471-2479, 2009

93. Gerritsen J, Dekker JM, TenVoorde BJ, et al: Impaired autonomic function is associated with increased

mortality, especially in subjects with diabetes, hypertension, or a history of cardiovascular disease:

The hoorn study. Diabetes Care 24(10):1793-1798, 2001

94. MacFadyen RJ, Barr CS, Struthers AD: Aldosterone blockade reduces vascular collagen turnover,

improves heart rate variability and reduces early morning rise in heart rate in heart failure patients.

Cardiovasc Res 35(1):30-34, 1997

95. Struthers AD: Two new culprits in cardiovascular disease: QT dispersion and aldosterone. J Hum

Hypertens 9(8):659-661, 1995

96. Greene EL, Kren S, Hostetter TH: Role of aldosterone in the remnant kidney model in the rat. J Clin

Page 28: University of Groningen Towards personalized

Aldosterone and Renal Damage

79

3

Invest 98(4):1063-1068, 1996

97. van den Hoven MJ, Waanders F, Rops AL, et al: Regulation of glomerular heparanase expression by

aldosterone, angiotensin II and reactive oxygen species. Nephrol Dial Transplant 24(9):2637-2645,

2009

98. Kramer AB, van der Meulen EF, Hamming I, van Goor H, Navis G: Effect of combining ACE inhibition

with aldosterone blockade on proteinuria and renal damage in experimental nephrosis. Kidney Int

71(5):417-424, 2007

99. Rocha R, Chander PN, Khanna K, Zuckerman A, Stier CT,Jr: Mineralocorticoid blockade reduces

vascular injury in stroke-prone hypertensive rats. Hypertension 31(1 Pt 2):451-458, 1998

100. Hamming I, Navis G, Kocks MJ, van Goor H: ACE inhibition has adverse renal effects during dietary

sodium restriction in proteinuric and healthy rats. J Pathol 209(1):129-139, 2006

101. Heeg JE, de Jong PE, van der Hem GK, de Zeeuw D: Efficacy and variability of the antiproteinuric

effect of ACE inhibition by lisinopril. Kidney Int 36(2):272-279, 1989

102. Vogt L, Waanders F, Boomsma F, de Zeeuw D, Navis G: Effects of dietary sodium and hydrochloro-

thiazide on the antiproteinuric efficacy of losartan. J Am Soc Nephrol 19(5):999-1007, 2008

103. Arima S, Kohagura K, Xu HL, et al: Nongenomic vascular action of aldosterone in the glomerular

microcirculation. J Am Soc Nephrol 14(9):2255-2263, 2003

104. Uhrenholt TR, Schjerning J, Hansen PB, et al: Rapid inhibition of vasoconstriction in renal afferent

arterioles by aldosterone. Circ Res 93(12):1258-1266, 2003

105. Arima S, Kohagura K, Xu HL, et al: Endothelium-derived nitric oxide modulates vascular action of

aldosterone in renal arteriole. Hypertension 43(2):352-357, 2004

106. Epstein M, Williams GH, Weinberger M, et al: Selective aldosterone blockade with eplerenone

reduces albuminuria in patients with type 2 diabetes. Clin J Am Soc Nephrol 1(5):940-951, 2006

107. Schjoedt KJ, Jacobsen P, Rossing K, Boomsma F, Parving HH: Dual blockade of the renin-angioten-

sin-aldosterone system in diabetic nephropathy: The role of aldosterone. Horm Metab Res 37 Suppl

1:4-8, 2005

108. McFarlane SI, Sowers JR: Cardiovascular endocrinology 1: Aldosterone function in diabetes mellitus:

Effects on cardiovascular and renal disease. J Clin Endocrinol Metab 88(2):516-523, 2003

109. Sato A, Hayashi K, Naruse M, Saruta T: Effectiveness of aldosterone blockade in patients with

diabetic nephropathy. Hypertension 41(1):64-68, 2003

110. Sato A, Saruta T: Aldosterone breakthrough during angiotensin-converting enzyme inhibitor ther-

apy. Am J Hypertens 16(9 Pt 1):781-788, 2003

111. Bianchi S, Bigazzi R, Campese VM: Antagonists of aldosterone and proteinuria in patients with CKD:

An uncontrolled pilot study. Am J Kidney Dis 46(1):45-51, 2005

112. Chrysostomou A, Pedagogos E, MacGregor L, Becker GJ: Double-blind, placebo-controlled study on

the effect of the aldosterone receptor antagonist spironolactone in patients who have persistent

proteinuria and are on long-term angiotensin-converting enzyme inhibitor therapy, with or without

an angiotensin II receptor blocker. Clin J Am Soc Nephrol 1(2):256-262, 2006

113. van den Meiracker AH, Baggen RG, Pauli S, et al: Spironolactone in type 2 diabetic nephropathy:

Effects on proteinuria, blood pressure and renal function. J Hypertens 24(11):2285-2292, 2006

114. Cook NR, Cutler JA, Obarzanek E, et al: Long term effects of dietary sodium reduction on cardiovas-

cular disease outcomes: Observational follow-up of the trials of hypertension prevention (TOHP).

BMJ 334(7599):885-888, 2007

115. Bomback AS, Kshirsagar AV, Amamoo MA, Klemmer PJ: Change in proteinuria after adding aldo-

sterone blockers to ACE inhibitors or angiotensin receptor blockers in CKD: A systematic review.

Am J Kidney Dis 51(2):199-211, 2008

Page 29: University of Groningen Towards personalized

Chapter 3

80

116. Lakkis J, Lu WX, Weir MR: RAAS escape: A real clinical entity that may be important in the progression

of cardiovascular and renal disease. Curr Hypertens Rep 5(5):408-417, 2003

117. Navis G, de Jong P, Donker AJ, van der Hem GK, de Zeeuw D: Diuretic effects of angiotensin-con-

verting enzyme inhibition: Comparison of low and liberal sodium diet in hypertensive patients. J

Cardiovasc Pharmacol 9(6):743-748, 1987

118. Hollenberg NK, Chenitz WR, Adams DF, Williams GH: Reciprocal influence of salt intake on adrenal

glomerulosa and renal vascular responses to angiotensin II in normal man. J Clin Invest 54(1):34-42,

1974

119. Sica DA: Pharmacokinetics and pharmacodynamics of mineralocorticoid blocking agents and their

effects on potassium homeostasis. Heart Fail Rev 10(1):23-29, 2005

120. [Anonymous]. Effectiveness of spironolactone added to an angiotensin-converting enzyme inhibitor

and a loop diuretic for severe chronic congestive heart failure (the randomized aldactone evalua-

tion study [RALES]). Am J Cardiol 78(8):902-907, 1996

121. Pitt B, Bakris G, Ruilope LM, DiCarlo L, Mukherjee R, EPHESUS Investigators: Serum potassium and

clinical outcomes in the eplerenone post-acute myocardial infarction heart failure efficacy and

survival study (EPHESUS). Circulation 118(16):1643-1650, 2008

122. Antman EM, Hand M, Armstrong PW, et al: 2007 focused update of the ACC/AHA 2004 guidelines

for the management of patients with ST-elevation myocardial infarction: A report of the american

college of Cardiology/American heart association task force on practice guidelines: Developed

in collaboration with the canadian cardiovascular society endorsed by the american academy of

family physicians: 2007 writing group to review new evidence and update the ACC/AHA 2004 guide-

lines for the management of patients with ST-elevation myocardial infarction, writing on behalf of

the 2004 writing committee. Circulation 117(2):296-329, 2008

123. Swedberg K, Cleland J, Dargie H, et al: Guidelines for the diagnosis and treatment of chronic heart

failure: Executive summary (update 2005): The task force for the diagnosis and treatment of chronic

heart failure of the european society of cardiology. Eur Heart J 26(11):1115-1140, 2005

Page 30: University of Groningen Towards personalized
Page 31: University of Groningen Towards personalized