Open access

Atrial Fibrillation and the Renin-Angiotensin-Aldosterone System

Written By

Stefano Perlini, Fabio Belluzzi, Francesco Salinaro and Francesco Musca

Submitted: 15 May 2012 Published: 27 February 2013

DOI: 10.5772/53917

From the Edited Volume

Atrial Fibrillation - Mechanisms and Treatment

Edited by Tong Liu

Chapter metrics overview

2,097 Chapter Downloads

View Full Metrics

1. Introduction

Atrial fibrillation (AF) is the most common cardiac arrhythmia, affecting approximately 1% of the general population and up to 8% of subjects over the age of 80 years.[1] AF is a major contributor to cardiovascular mortality and morbidity, being associated with decreased quality of life, increased incidence of congestive heart failure,[2] embolic phenomena, including stroke,[2,3] and a 30 % higher risk of death.[3,4] AF-associated morbidity includes a four- to five-fold increased risk for stroke, [2,5] a two-fold increased risk for dementia,[6,7] and a tripling of risk for heart failure.[5] According to the Framingham Study, the percentage of strokes attributable to AF increases steeply from 1.5% at 50–59 years of age to 23.5% at 80–89 years of age, [2] and the presence of AF accounts for a 50–90% increased risk for overall mortality.[3] From the viewpoint of the AF-related socio-economic burden, it has been estimated that it is consuming between 0.9% and 2.4% of total National Health Service expenditure in the UK,[8] while in the USA, total costs are 8.6–22.6% higher for AF patients in all age- and sex- population strata.[9] Therefore significant clinical, human, social and economical benefits are therefore expected from any improvement in AF prevention and treatment.

It has to be noted that although multiple treatment options are currently available, no single modality is effective for all patients.[10] AF can occasionally affect a structurally normal heart of otherwise healthy individuals (so-called “lone AF”)[11], but most typically it occurs in subjects with previous cardiovascular damage due to hypertension, coronary artery disease and diabetes. Moreover, it can be associated with clinical conditions such as hyperthyroidism, acute infections, recent cardiothoracic or abdominal surgery, and systemic inflammatory diseases. Whatever the cause, AF is characterized by very rapid, chaotic electrical activity of the atria, resulting in accelerated and irregular ventricular activity, loss of atrial mechanical function and increased risk of atrial clot formation.

Many studies have shown that the recurrence of AF may be partially related to a phenomenon known as “atrial remodeling”, in which the electrical, mechanical, and structural properties of the atrial tissue and cardiac cells are progressively altered, creating a more favorable substrate for AF development and maintenance.[12,13] Atrial remodeling is both a cause and a consequence of the arrhythmia, and in recent years it has become more and more evident that treatment should also be based on an “upstream” therapy[14,10] aimed at modifying the arrhythmia substrate and at reducing the extent of atrial remodelling.

Advertisement

2. Atrial remodeling: electrical and structural factors

According to Coumel´s triangle of arrhythmogenesis, three cornerstones are required in the onset of clinical arrhythmia[15] – the arrhythmogenic substrate, the trigger factor and the modulation factors such as autonomic nervous system or inflammation. Once established, AF itself alters electrical and subsequently structural properties of the atrial tissue and these changes cause or “beget” further AF self-perpetuation.[12] The mechanisms responsible for the onset and persistence of the arrhythmia involve electrical as well as structural determinants, that are very complex and yet poorly understood. From the electrical standpoint, there is still debate on the three models that were proposed in 1924[16] by Garrey for describing the mechanisms of spatiotemporal organization of electrical activity in the atria during AF. According to the focal mechanism theory, AF is provoked and perhaps also driven further by the rapid firing of a single or multiple ectopic foci, whereas the single circuit re-entry theory assumes the presence of a single dominant re-entry circuit, and the multiple wavelet theory postulates the existence of multiple reentry circuits with randomly propagating wave-fronts that must find receptive tissue in order to persist.[17] It has to be recognized that all three models are non-exclusive and each may be applicable to certain subgroups of AF patients, or that they may even coexist in the same subject during different stages of AF development. Moreover, AF persistence is associated with modifications in the atrial myocyte electrical properties (the so-called electrical remodeling), that may stabilize the arrhythmia by decreasing the circuit size. The electrophysiological properties of the atrial myocardium may be further modified by changes in autonomic nervous system activity as well as by the interference of drugs and hormones, that may therefore participate in arrhythmogenesis.

Beyond these electrical determinants, AF onset and persistence may be affected by the structural factors, such as the dimensions and geometry of the atrial chambers, the atrial tissue structure and the amount and the composition of the extracellular matrix surrounding the atrial myocytes (i.e. structural remodeling). Together, these alterations create an arrhythmogenic substrate essential for the persistence of AF. Atrial structure is modified by volume and pressure overload, due to either mitral valve disease or left ventricular diastolic dysfunction in the setting of arterial hypertension, coronary artery disease or aortic valve disease. Also diabetes is associated with changes in atrial structure and function. It is not therefore surprising that all these clinical conditions are associated with an increased AF incidence and prevalence. Beyond being a possible substrate for AF onset, atrial structure is profoundly altered by the effects of rapid atrial rate. Prolonged rapid atrial pacing induces changes in atrial myocytes such as an increase in cell-size, myocyte lysis, perinuclear accumulation of glycogen, alterations in connexin expression, fragmentation of sarcoplasmic reticulum and changes in mitochondrial shape.[18] Moreover, structural remodeling is characterized by changes in extracellular matrix composition, with both diffuse interstitial and patchy fibrosis.[19] All these alterations results in electrical tissue non-homogeneity, slowed conduction and electrical uncoupling, that facilitate AF continuation. In contrast to electrical remodeling, structural changes are far less reversible and they tend to persist even after sinus rhythm restoration. Among the several mechanisms and signaling pathways involved in structural remodeling and atrial fibrosis, a key role is played by the renin-angiotensin system, and by the transforming growth-factor β1 (TGF-β1) pathway, associated with tissue inflammation[19] and reactive oxygen species production.[20,21]

Profibrotic signals act on the balance between matrix metalloproteinases (MMPs) – the main enzymes responsible for extracellular matrix degradation – and their local tissue inhibitors (TIMPs), that can be differentially altered in compensated as opposed to decompensated pressure-overload hypertrophy.[22-25] Furthermore, profibrotic signals stimulate the proliferation of fibroblasts and extracellular deposition of fibronectin, collagens I and III, proteglycans and other matrix components. In a canine model of congestive heart failure, Li et al. showed that the development of atrial fibrosis is angiotensin-II dependent,[26] via mechanisms that are partly mediated by the local production of cytokine TGF-β1.[27] In transgenic mice, overexpression of the latter cytokine has been shown to lead to selective atrial fibrosis, increased conduction heterogeneity and enhanced AF susceptibility, despite normal atrial action potential duration and normal ventricular structure and function.[28]

Advertisement

3. The renin-angiotensin-aldosterone system (RAAS) as a “novel” risk factor for AF

Among many others, two factors contribute to the search of different therapeutic approaches to AF specifically targeting substrate development and maintenance:[29] the recognition of novel risk factors for the development of this arrhythmia and the well-known limitations of the current antiarrhythmic drug therapy to maintain sinus rhythm, still having inadequate efficacy and potentially serious adverse effects.[30] In this setting, the inhibition of the renin-angiotensin-aldosterone system (RAAS) has been considered useful in both primary and secondary prevention of AF, particularly in patients presenting left ventricular hypertrophy (LVH) or heart failure. The RAAS is a major endocrine/paracrine system involved in the regulation of the cardiovascular system.[31] Its key mediator is angiotensin II, an octapeptide that is cleaved from the liver-derived 485-aminoacid precursor angiotensinogen through a process involving the enzymatic activities of renin and angiotensin converting enzyme (ACE). Two main angiotensin II receptors exist, i.e angiotensin II type 1 (AT1) and type 2 (AT2). AT1-receptor mediated pathways lead to vasoconstriction, water retention, increased renal tubular sodium reabsorption, stimulation of cell growth and connective tissue deposition, and impaired endothelial function. AT2-receptor has opposing effects, inasmuch as it mediates vasodilation, decreases renal tubular sodium reabsorption, inhibits cell growth and connective tissue deposition, and improves endothelial function. These two angiotensin receptors have different expression patterns, AT1 being constitutively expressed in a wide range of tissues of the cardiovascular, renal, endocrine, and nervous system, and AT2 expression being activated during stress conditions.[32] It is becoming increasingly evident that all these mechanisms are involved in atrial remodeling and hence in AF development and maintenance. Moreover, among the other biologically active RAAS components that are involved in these processes, angiotensin-(1-7) [Ang-(1-7)] seems to be particularly important. In an experimental canine model of chronic atrial pacing, Ang-(1-7) has been shown to reduce AF vulnerability and atrial fibrosis,[33] influencing atrial tachycardia-induced atrial ionic remodeling. [34]

Among the compounds that may interfere RAAS four classes of drugs are particularly relevant in cardiovascular therapy: angiotensin receptor blockers (ARBs), ACE inhibitors (ACEIs), aldosterone antagonists and direct renin inhibitors. ARBs directly block AT1 receptor activation, ACEIs inhibit ACE-mediated production of angiotensin II, and the recently developed direct renin inhibitor aliskiren blocks RAAS further upstream.[32,35,36] Over the last decade, these drugs have been tested in the setting of AF treatment and prevention.

Advertisement

4. The role of RAAS in the pathogenesis of AF

4.1. Atrial stretch and AF

Atrial arrhythmias frequently occur under conditions associated with atrial dilatation and increased atrial pressure, causing atrial tissue stretch and modifying atrial refractoriness, and it has been shown in several animal as well as clinical models.[37-40] These factors increase susceptibility to AF, that is associated with shortening of the atrial effective refractory period (AERP), possibly by opening of stretch-activated ion channels. In the setting of arterial hypertension and congestive heart failure (CHF), angiotensin II has been associated with increased left atrial and left ventricular end-diastolic pressure,[41] and both ACEIs and ARBs have been shown to reduce left atrial pressure.[42-45] Therefore, one potential mechanism by which ACEIs and ARBs may reduce atrial susceptibility to AF is by reducing atrial stretch. Many other mechanisms appear to be involved in the antiarrhythmic properties of RAAS inhibition, and in an animal model of ventricular tachycardia-induced CHF it has been shown that ACE inhibition is more successful than hydralazine/isosorbide mononitrate association in reducing burst pacing-induced AF promotion, despite a similar reduction in left atrial pressure.[26] As described below, angiotensin II-mediated mechanisms contribute to both structural and electrical remodeling of the atrial tissue.

4.2. The role of RAAS in structural remodeling

Atrial fibrosis causes conduction heterogeneity, hence playing a key role in the development of a vulnerable structural substrate for AF, and the proinflammatory and profibrotic effects of angiotensin II have been extensively described.[46-48] Excessive fibrillar collagen deposition, resulting from deregulated extracellular matrix metabolism, leads to atrial fibrosis, and it has been shown that angiotensin II has a direct effect in stimulating cardiac fibroblast proliferation and collagen synthesis, via AT1 receptor – mediated mechanisms involving a mitogen-activated protein kinases (MAPKs) phosphorylation pathway. [49-51] The latter cascade is inhibited by AT2 receptor activation, that has an antiproliferative effects.[52] Moreover, cardiac fibroblast function is modulated by angiotensin II through mechanisms involving TGF-1, osteopontin (OPN), and endothelin-1 (ET-1). [49,53-55] Interestingly, Nakajima and coworkers showed that selective atrial fibrosis, conduction heterogeneity, and AF propensity are enhanced in a TGFβ1 cardiac overexpression transgenic mice model,[56] as also confirmed by others.[27,28]

Beyond having both direct and indirect effects on collagen synthesis, angiotensin II interferes with collagen degradation by modulating interstitial matrix metalloproteinase (MMP) activity and tissue inhibitor of metalloproteinase (TIMP) concentrations,[52] and an atrial tissue imbalance between MMPs and TIMPs has been reported in both clinical and animal studies on AF. [52,57] Goette and coworkers showed increased atrial expression of ACE and increased activation of the angiotensin II-related intracellular signal transduction pathway in human atrial tissue derived from AF patients,[58] and atrial overexpression of angiotensin II has also been shown in a canine model of ventricular tachycardia-induced CHF[26,59] In transgenic mice experiments with cardiac-restricted ACE overexpression, Xiao et al. have demonstrated that elevated atrial tissue angiotensin II concentrations stimulates atrial fibrosis and hence an AF-promoting substrate.[60] In contrast, RAAS inhibition reduces tissue angiotensin II concentration, and attenuates atrial structural remodeling and fibrosis, thereby contrasting AF maintenance.[26,59,61-64]

4.3. The role of RAAS in electrical remodeling

Electrical remodeling has been hypothesized as a main mechanism by which, once established, “AF begets further AF” self-perpetuation.[12] In the clinical practice, this phenomenon is evident when considering that over time it becomes more and more difficult to keep in sinus rhythm a patient with AF. The concept of electrical remodeling has been originally proposed by Wijffels et al.[12] to explain the experimental observation that when AF is maintained artificially, the duration of burst pacing-induced paroxysms progressively increases until AF becomes sustained. This indicates that AF itself alters the atrial tissue electrical properties, thereby developing a functional substrate that promotes AF perpetuation and may involve alterations in ionic currents and in excitability cellular properties.[65] In their study, Wijffels et al. demonstrated that the increased propensity to AF is associated with shortening of the atrial effective refractory period (AERP) in accordance with the multiple wavelet theory,[12] a mechanism that was subsequently attributed to a reduction of action potential duration (APD) secondary to the progressive downregulation of the transient outward current (Ito) and of the L-typeCa2+ current (ICa,L).[66] As to the modulation of the ICa,L current, the role of angiotensin II is controversial, with studies reporting increase, decrease, or even no effect.[29,67] In contrast, angiotensin II has been demonstrated to downregulate Ito current,[68,67] inasmuch as AT1 receptor stimulation leads to internalization of the Kv4.3 (i.e., the pore-forming α-subunit underlying Ito), regulating its cell-surface expression.[68] As shown by Liu and coworkers, chronic Ang-(1-7) infusion prevented the decrease of Ito, ICa,L, and of Kv4.3 mRNA expression induced by chronic atrial pacing, [34] thereby contributing to reduce AF vulnerability.[33] Subsequently, Nakashima et al. showed that ACEI or ARB treatment results in complete inhibition of the shortening of AERP, that is normally induced by rapid atrial pacing.[69] A further mechanism by which the RAAS may exert a proarrhythmic effect is the modulation of gap junctions, that are low-resistance pathways for the propagation of impulses between cardiomyocytes formed by connexins (Cx).[70] Cx40 gene polymorphisms have been associated with the development of non familial AF,[71] and angiotensin II has been implicated in Cx43 downward remodeling.[72-74] Moreover, angiotensin II directly induces delayed after-depolarizations and accelerates the automatic rhythm of isolated pulmonary vein cardiomyocytes.[75] These cells are considered an important source of ectopic beats and of atrial fibrillation bursts, representing the target of AF treatment with radio-frequency ablation.[76] Therefore these experimental results demonstrate that angiotensin II may play a role in the pathophysiology of atrial fibrillation also by modulating the pulmonary vein electrical activity via an electrophysiological effect that was shown to be AT1 receptor – mediated, being inhibited by losartan, [75] and that is attenuated by heat-stress responses.[77] Recently, also the direct renin inhibitor aliskiren was shown to reduce the arrhythmogenic activity of pulmonary vein cardiomyocytes.[36] It has also been demonstrated that aldosterone promotes atrial fibrillation, causing a substrate for atrial arrhythmias characterized by atrial fibrosis, myocyte hypertrophy, and conduction disturbances,[78] and the specific antagonist spironolactone has been shown to prevent aldosterone-induced increased duration of atrial fibrillation in a rat model.[79]

4.4. RAAS gene polymorphisms and AF

The ACE DD (deletion/deletion) genotype of the ACE gene has been shown to be a predisposing factor for persistent AF,[80] and it was recently reported that the same genotype is associated with lowest rates of symptomatic response in patients with lone AF.[81] Moreover, polymorphisms of the angiotensinogen gene have also been associated with nonfamilial AF,[82] and it has been shown that significant interactions exist between angiotensinogen gene haplotypes and ACE I/D (insertion/deletion) polymorphism resulting in increased susceptibility to AF.[83,84] Also aldosterone synthase (CYP11B2) T-344C polymorphism, which is associated with increased aldosterone activity, was shown to be an independent predictor of AF in patients with HF.[85] According to Sun and coworkers, this aldosterone synthase gene polymorphism might also be associated with atrial remodelling in hypertensive patients.[86]

Advertisement

5. Atrial fibrillation and the renin-angiotensin-aldosterone system (RAAS): Clinical observations

A possible relationship between the RAAS and the risk of developing AF was brought about by several clinical data, derived from patient series in different settings, that are here summarized.

5.1. Heart failure

In heart failure, several observations indicate a possible effect of RAAS inhibition in reducing the incidence of new onset AF. In a retrospective analysis of the SOLVD trial, Vermes et al. showed that enalapril reduces the risk of AF development in patients with various degrees of heart failure.[87] Similarly, Maggioni et al. demonstrated that use of the ARB valsartan is associated with a reduction in the risk of AF in the Val-HeFT trial population.[88] Since the vast majority of these patients (92.5%) were already receiving an ACEI, a combination effect was hypothesized, and the benefit of combined treatment with both an ARB and an ACEI was also supported by the results of the CHARM trial with candesartan.[89] The latter study was composed by three component trials based on left ventricular ejection fraction (LVEF) and ACEI treatment. CHARM-Alternative trial enrolled patients with LVEF ≤40% not treated with ACEIs because of prior intolerance, CHARM-Added recruited patients with LVEF ≤40% already treated with an ACEI, and CHARM-Preserved included patients with LVEF >40%, independent of ACEI treatment. The incidence of new-onset AF was reduced in candesartan-treated patients, especially (but not exclusively) in the CHARM-Alternative trial.[89] These data indicate additional benefits in AF prevention, on the top of the already known effects of ACEI/ARB treatment in patients with heart failure.

5.2. Post-MI

After an acute myocardial infarction, treatment with the ACEI trandolapril reduced the incidence AF in patients with impaired left ventricular function, irrespective of the effects on ejection fraction per se.[90] Similar results were reported by Pizzetti et al. with lisinopril in their analysis of the GISSI-3 trial.[91]

5.3. Hypertension

The issue of the possible role of ACEI/ARB drug treatment in the primary prevention of AF in hypertensive patients derives from several conflicting observations. According to the CAPPP and the STOP-H2 trials, ACEIs were comparable to other antihypertensive regiments in preventing AF.[92,93] In contrast, a retrospective, longitudinal, cohort study by L’Allier et al. reported a benefit of ACEIs over calcium channel blockers in terms of new onset AF and AF-related hospitalizations.[94] Similar results were derived from the LIFE trial, showing that when compared with the β-blocker atenolol, patients receiving the ARB losartan had significantly lower incidence of new-onset AF and associated stroke.[95] A recent nested case-control observational study showed that compared with treatment with calcium channel blockers, long-term antihypertensive treatment with ACEIs, ARBs, or β-blockers may decrease the risk of new-onset AF.[96]

5.4. Increased cardiovascular risk

In patients with increased cardiovascular risk, the rate of new onset AF was not reduced by ramipril in a subanalysis of the HOPE clinical trial by Salehian and coworkers,[97] although in a population with a rather low incidence of AF (2.1%). Also in the ACTIVE I trial, there was no benefit of irbesartan treatment in preventing hospitalization for atrial fibrillation or atrial fibrillation recorded by 12-lead electrocardiography, nor was there a benefit in a subgroup of patients who underwent transtelephonic monitoring.[98] In contrast, according to Schmieder et al. the VALUE trial showed that valsartan-based antihypertensive treatment reduced the development of new-onset AF compared to amlodipine,[99] in subjects at higher risk of this arrhythmia due to an almost 25% prevalence of electrocardiographically-defined left ventricular hypertrophy. These conflicting data may indicate that a possible benefit of ACEI or ARB treatment can at best be observed in patients with the highest probability of increased RAAS activation.

5.5. Postoperative AF

A reduced incidence of new-onset AF was observed in patients undergoing coronary artery bypass graft surgery who were treated with ACEIs,[100] in a large multicenter prospective trial recruiting 4,657 subjects. These results were confirmed with the use of ACEIs alone or associated with candesartan,[101] whereas the reduced risk of developing postoperative AF did not reach the statistical significance in the post hoc evaluation of patients enrolled in the AFIST II and III trials.[102]

5.6. Secondary prevention after cardioversion and after catheter ablation

In the setting of secondary prevention, patients undergoing AF cardioversion represent a group in which the potential role of RAAS inhibition has been first investigated by van den Berg et al.[103], iwho studied 30 CHF patients treated with lisinopril or placebo before and after the procedure. Although the reduced incidence of recurrent AF in ACE-I treated patients did not reach the statistical significance, this study was followed by many others. Dagres et al. [104] demonstrated that treatment with the ARB irbesartan is associated with attenuated left atrial stunning after cardioversion. Subsequent studies showed that the association of an ACEI or an ARB with amiodarone prevents AF recurrences after cardioversion when compared with amiodarone alone.[105-107] Interestingly, irbesartan showed a dose-dependent preventive effect.[106] In contrast, Tveit and coworkers did not find any benefit by treating with the ARB candesartan for 3-6 weeks before and 6 months after electrical cardioversion.[108] We contributed to this debate by showing that also in the setting of lone AF,[11] long-term treatment with the ACE-I ramipril is effective in preventing relapses of AF after successful cardioversion.[109] Moreover, at the end of a 3-year follow-up, ramipril treatment also prevented left atrium enlargement,[109] which has been demonstrated to occur in the natural history of lone AF.[110]

In patients undergoing catheter ablation for drug refractory AF, ACEIs or ARBs did not show the same promising results,[111-115] raising the question whether these interventions are indeed able to revert atrial remodeling in this clinical setting.[116]

5.7. Paroxysmal AF prevention

Both ACEIs and ARBs have shown some promise in the setting of the prevention of paroxysmal AF recurrences. In two long-term clinical trials on amiodarone-treated patients, losartan or perindopril were more effective than amlodipine in the maintenance of sinus rhythm. [117,118] The same held true for telmisartan, that Fogari et al. showed as more effective than ramipril in reducing AF recurrence and severity as well as in improving P-wave dispersion, suggesting a possible specific effect of telmisartan on atrial electric remodeling.[119] In a retrospective analysis of patients with predominantly paroxysmal AF, Komatsu and coworkers showed that the enalapril added to amiodarone reduced the rate of AF recurrence and prevented the development of atrial structural remodeling.[120] In a post hoc subgroup analysis of the AFFIRM trial, Murray et al. showed that ACEIs and ARBs reduced the risk of AF recurrence in patients with a history of CHF or impaired left ventricular function. [121] The GISSI-AF trial did not show any significant effect of valsartan treatment on the rate of AF recurrences in a cohort of 1,442 patients with a history of recent AF.[122] Although it has to be noted that valsartan-treated patients had a significantly higher prevalence of coronary artery disease and peripheral artery disease, and that more than half of the patients were already taking concomitant ACEI treatment, the GISSI-AF shed some doubt on the whole issue of the preventive role of RAAS inhibition in AF prevention.[122] In the same line, the very recent ANTIPAF trial concluded that 12-month treatment with the ARB olmesartan did not reduce the number of AF episodes in patients with documented paroxysmal AF without structural heart disease.[123] Similar results were shown by the J-RHYTHM II study comparing the ARB candesartan with the calcium antagonist amlodipine in the treatment of paroxysmal AF associated with hypertension.[124] Both studies used daily transtelephonic monitoring to examine asymptomatic and symptomatic paroxysmal AF episodes. [123,124]

5.8. Emerging role for aldosterone antagonists

In the recent years, it has been suggested that upstream therapy using aldosterone antagonists, such as spironolactone or eplerenone, may reduce the deleterious effect of excessive aldosterone secretion on atrial tissue, thereby contributing to modify the risk of developing and of maintaining AF.[125] Dabrowski et al. showed that combined spironolactone plus beta-blocker treatment might be a simple and valuable option in preventing AF episodes in patients with normal left ventricular function and history of refractory paroxysmal AF.[126] In patients with AF, spironolactone treatment was associated with a reduction in the AF burden, as reflected by a combination of hospitalizations for AF and electrical cardioversion.[127] In a recent trial in patients with systolic heart failure and mild symptoms (EMPHASIS-HF), the aldosterone antagonist eplerenone reduced the incidence of new-onset AF or atrial flutter.[128]

5.9. Meta-analyses

The promise of a protective role of RAAS inhibition is largely based on the analysis of retrospective data, although on several thousands of patients. Another limitation is the fact that in most cases, the detection of AF recurrences is based on annual electrocardiograms, periodical 24-hour Holter analysis, or patient self-reported symptoms symptoms. In recent years, with the analysis of data from patients with an implanted pacemaker, it is becoming increasingly clear that continuous monitoring is much more reliable in identifying the presence of asymptomatic recurrences, with a mean sensitivity in detecting an AF episode lasting >5 minutes that was 44.4%, 50.4%, and 65.1% for 24-hour Holter, 1-week Holter, and 1-month Holter monitoring, respectively.[129] To partially overcome some of these limitations, several meta-analyses of the available trials have been conducted.[130-141] In synthesis, despite the promising preliminary experimental and clinical data, the efficacy of RAAS inhibition in the prevention of atrial fibrillation recurrences is still under debate, leading Disertori et al. in a very recent review article to the definition of “an unfulfilled hope”.[136] In meta-analysis including 92,817 randomized patients, Khatib and coworkers concluded that although RAAS inhibition appears to reduce the risk of developing new onset atrial fibrillation in different patient groups, further research with stronger quality trials is required to draw definitive conclusions.[141]

Indeed, ACE-I or ARBs cannot be considered as an alternative to the established antiarrhythmic agents and transcatheter ablation. However, since they are recommended for most concomitant cardiovascular diseases that are associated with an increased risk of AF (i.e., hypertension, heart failure, ischemic heart disease) and since there are several lines of evidence that increased angiotensin II tissue levels are involved in both structural and electrical remodeling of the atrial tissue, it appears reasonable to use these drugs. In general, no substantial difference was found in the comparison between ACE-I and ARB treatment, a finding that was confirmed also by the results of the the ONTARGET and TRANSCEND trials.[142]

5.10. Atrial remodeling as a therapeutic target: modulation of the renin-angiotensin-aldosterone system

Since angiotensin II plays a central role in the development of atrial fibrosis, inhibition of atrial angiotensin converting enzyme (ACE) and AT1 angiotensin receptors might be beneficial in AF. In experimental models, AF susceptibility and atrial fibrosis were decreased by candesartan or enalapril, but not by hydralazine or isosorbide mononitrate despite similar hemodynamic effects,[26,63] thus suggesting a key role of targeting renin-angiotensin system, rather than of improving the hemodynamics. This concept was further underscored after demonstrating a preventive role of ramipril treatment in patients with lone AF.[109] Also spironolactone was able to prevent AF episodes in patients with normal left ventricular function and a history of refractory paroxysmal AF.[126] With the notable exception of the GISSI-AF,[122] ANTIPAF,[123] and J-RHYTHM II[124] trials, the majority of the available studies showed that modulation of the renin-angiotensin-aldosterone system is able to reduce the incidence of AF, as well as its recurrence after electrical cardioversion.[134] These data are summarized in several meta-analyses,[131,132,140,143] also including the GISSI-AF data.[135] In a broader view, although ACE inhibitors and angiotensin-II receptor blockers (ARBs) are not to be considered antiarrhythmic drugs, several studies have shown that they are associated with a lower incidence of ventricular arrhythmias in patients with ischemic heart disease and left ventricular (LV) dysfunction,[90,144,145] possibly because of the adverse effects of angiotensin II on the cardiac remodeling process. Indeed, it must be recognized that in the presence of a cardiac disease causing atrial overload and/or dysfunction, the effectiveness of ACE inhibitors and/or ARBs might be attributable either to a direct antiarrhythmic effect or to an effect on atrial structure and/or function likely able to favorably modify the arrhythmic substrate, such as the increase in left atrial (LA) dimensions that is frequently observed in patients with arterial hypertension and/or LV dysfunction.

In the setting of AF, it has to be remembered that angiotensin II not only has several effects on the structure of the atrial myocardium, but also on its electrical properties, as it has been elegantly shown in isolated pulmonary vein cardiomyocytes,[75] and in instrumented animal studies.[69] Therefore, the protective effect of ACE inhibition or angiotensin II antagonists on the electrical and structural remodeling of the atria is very likely, due to a combination of their actions on atrial distension/stretch, sympathetic tone, local renin-angiotensin system, electrolyte concentrations, and cardiac loading conditions.

Advertisement

6. Conclusions

The onset of atrial fibrillation results from a complex interaction between triggers, arrhythmogenic substrate, and modulator factors. Once established, AF itself alters the electrical and structural properties of the atrial myocardium, thereby perpetuating the arrhythmia. Among many other factors, angiotensin II and aldosterone play an important role not only in determining atrial fibrosis, but also in modulating the electrical properties of the atrial myocardium. These aspects may be relevant in explaining the many clinical observations indicating the role of drugs modulating the renin-angiotensin-aldosterone system in preventing atrial fibrillation in different settings.

References

  1. 1. Fuster V, Ryden LE, Cannom DS, Crijns HJ, Curtis AB, Ellenbogen KA, Halperin JL, Le Heuzey JY, Kay GN, Lowe JE, Olsson SB, Prystowsky EN, Tamargo JL, Wann S (2006) ACC/AHA/ESC 2006 guidelines for the management of patients with atrial fibrillation-executive summary: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Revise the 2001 Guidelines for the Management of Patients with Atrial Fibrillation). Eur Heart J 27 (16):1979-2030
  2. 2. Wolf PA, Abbott RD, Kannel WB (1991) Atrial fibrillation as an independent risk factor for stroke: the Framingham Study. Stroke 22 (8):983-988
  3. 3. Benjamin EJ, Wolf PA, D'Agostino RB, Silbershatz H, Kannel WB, Levy D (1998) Impact of atrial fibrillation on the risk of death: the Framingham Heart Study. Circulation 98 (10):946-952
  4. 4. Stewart S, Hart CL, Hole DJ, McMurray JJ (2002) A population-based study of the long-term risks associated with atrial fibrillation: 20-year follow-up of the Renfrew/Paisley study. Am J Med 113 (5):359-364
  5. 5. Krahn AD, Manfreda J, Tate RB, Mathewson FA, Cuddy TE (1995) The natural history of atrial fibrillation: incidence, risk factors, and prognosis in the Manitoba Follow-Up Study. Am J Med 98 (5):476-484. doi:10.1016/S0002-9343(99)80348-9
  6. 6. Ott A, Breteler MM, de Bruyne MC, van Harskamp F, Grobbee DE, Hofman A (1997) Atrial fibrillation and dementia in a population-based study. The Rotterdam Study. Stroke 28 (2):316-321
  7. 7. Miyasaka Y, Barnes ME, Petersen RC, Cha SS, Bailey KR, Gersh BJ, Casaclang-Verzosa G, Abhayaratna WP, Seward JB, Iwasaka T, Tsang TS (2007) Risk of dementia in stroke-free patients diagnosed with atrial fibrillation: data from a community-based cohort. Eur Heart J 28 (16):1962-1967. doi:10.1093/eurheartj/ehm012
  8. 8. Wachtell K, Devereux RB, Lyle AP (2007) The effect of angiotensin receptor blockers for preventing atrial fibrillation. Curr Hypertens Rep 9 (4):278-283
  9. 9. Wolf PA, Mitchell JB, Baker CS, Kannel WB, D'Agostino RB (1998) Impact of atrial fibrillation on mortality, stroke, and medical costs. Arch Intern Med 158 (3):229-234
  10. 10. Camm AJ, Kirchhof P, Lip GY, Schotten U, Savelieva I, Ernst S, Van Gelder IC, Al-Attar N, Hindricks G, Prendergast B, Heidbuchel H, Alfieri O, Angelini A, Atar D, Colonna P, De Caterina R, De Sutter J, Goette A, Gorenek B, Heldal M, Hohloser SH, Kolh P, Le Heuzey JY, Ponikowski P, Rutten FH (2010) Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur Heart J 31 (19):2369-2429. doi:10.1093/eurheartj/ehq278
  11. 11. Evans W, Swann P (1954) Lone auricular fibrillation. Br Heart J 16 (2):189-194
  12. 12. Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA (1995) Atrial fibrillation begets atrial fibrillation. A study in awake chronically instrumented goats. Circulation 92 (7):1954-1968
  13. 13. Allessie M, Ausma J, Schotten U (2002) Electrical, contractile and structural remodeling during atrial fibrillation. Cardiovasc Res 54 (2):230-246
  14. 14. Smit MD, Van Gelder IC (2009) Upstream therapy of atrial fibrillation. Expert Rev Cardiovasc Ther 7 (7):763-778
  15. 15. Farre J, Wellens HJ (2004) Philippe Coumel: a founding father of modern arrhythmology. Europace 6 (5):464-465
  16. 16. Garrey WE (1924) Auricular Fibrillation. Physiological Reviews 4 (2):215-250
  17. 17. Moe GK, Rheinboldt WC, Abildskov JA (1964) A Computer Model of Atrial Fibrillation. Am Heart J 67:200-220
  18. 18. Ausma J, Wijffels M, Thone F, Wouters L, Allessie M, Borgers M (1997) Structural changes of atrial myocardium due to sustained atrial fibrillation in the goat. Circulation 96 (9):3157-3163
  19. 19. Frustaci A, Chimenti C, Bellocci F, Morgante E, Russo MA, Maseri A (1997) Histological substrate of atrial biopsies in patients with lone atrial fibrillation. Circulation 96 (4):1180-1184
  20. 20. Bruins P, te Velthuis H, Yazdanbakhsh AP, Jansen PG, van Hardevelt FW, de Beaumont EM, Wildevuur CR, Eijsman L, Trouwborst A, Hack CE (1997) Activation of the complement system during and after cardiopulmonary bypass surgery: postsurgery activation involves C-reactive protein and is associated with postoperative arrhythmia. Circulation 96 (10):3542-3548
  21. 21. Kim YH, Lim DS, Lee JH, Shim WJ, Ro YM, Park GH, Becker KG, Cho-Chung YS, Kim MK (2003) Gene expression profiling of oxidative stress on atrial fibrillation in humans. Exp Mol Med 35 (5):336-349
  22. 22. Tozzi R, Palladini G, Fallarini S, Nano R, Gatti C, Presotto C, Schiavone A, Micheletti R, Ferrari P, Fogari R, Perlini S (2007) Matrix metalloprotease activity is enhanced in the compensated but not in the decompensated phase of pressure overload hypertrophy. Am J Hypertens 20 (6):663-669. doi:10.1016/j.amjhyper.2007.01.016
  23. 23. Falcao-Pires I, Palladini G, Goncalves N, van der Velden J, Moreira-Goncalves D, Miranda-Silva D, Salinaro F, Paulus WJ, Niessen HW, Perlini S, Leite-Moreira AF (2011) Distinct mechanisms for diastolic dysfunction in diabetes mellitus and chronic pressure-overload. Basic Res Cardiol 106 (5):801-814. doi:10.1007/s00395-011-0184-x
  24. 24. Castoldi G, di Gioia CR, Bombardi C, Perego C, Perego L, Mancini M, Leopizzi M, Corradi B, Perlini S, Zerbini G, Stella A (2010) Prevention of myocardial fibrosis by N-acetyl-seryl-aspartyl-lysyl-proline in diabetic rats. Clin Sci (Lond) 118 (3):211-220
  25. 25. Perlini S, Palladini G, Ferrero I, Tozzi R, Fallarini S, Facoetti A, Nano R, Clari F, Busca G, Fogari R, Ferrari AU (2005) Sympathectomy or doxazosin, but not propranolol, blunt myocardial interstitial fibrosis in pressure-overload hypertrophy. Hypertension 46 (5):1213-1218
  26. 26. Li D, Shinagawa K, Pang L, Leung TK, Cardin S, Wang Z, Nattel S (2001) Effects of angiotensin-converting enzyme inhibition on the development of the atrial fibrillation substrate in dogs with ventricular tachypacing-induced congestive heart failure. Circulation 104 (21):2608-2614
  27. 27. Kupfahl C, Pink D, Friedrich K, Zurbrugg HR, Neuss M, Warnecke C, Fielitz J, Graf K, Fleck E, Regitz-Zagrosek V (2000) Angiotensin II directly increases transforming growth factor beta1 and osteopontin and indirectly affects collagen mRNA expression in the human heart. Cardiovasc Res 46 (3):463-475
  28. 28. Verheule S, Sato T, Everett Tt, Engle SK, Otten D, Rubart-von der Lohe M, Nakajima HO, Nakajima H, Field LJ, Olgin JE (2004) Increased vulnerability to atrial fibrillation in transgenic mice with selective atrial fibrosis caused by overexpression of TGF-beta1. Circ Res 94 (11):1458-1465
  29. 29. Ehrlich JR, Hohnloser SH, Nattel S (2006) Role of angiotensin system and effects of its inhibition in atrial fibrillation: clinical and experimental evidence. Eur Heart J 27 (5):512-518
  30. 30. Lafuente-Lafuente C, Mouly S, Longas-Tejero MA, Mahe I, Bergmann JF (2006) Antiarrhythmic drugs for maintaining sinus rhythm after cardioversion of atrial fibrillation: a systematic review of randomized controlled trials. Arch Intern Med 166 (7):719-728. doi:10.1001/archinte.166.7.719
  31. 31. Iravanian S, Dudley SC, Jr. (2008) The renin-angiotensin-aldosterone system (RAAS) and cardiac arrhythmias. Heart Rhythm 5 (6 Suppl):S12-17
  32. 32. Ram CV (2008) Angiotensin receptor blockers: current status and future prospects. Am J Med 121 (8):656-663
  33. 33. Liu E, Yang S, Xu Z, Li J, Yang W, Li G (2010) Angiotensin-(1-7) prevents atrial fibrosis and atrial fibrillation in long-term atrial tachycardia dogs. Regul Pept 162 (1-3):73-78. doi:10.1016/j.regpep.2009.12.020
  34. 34. Liu E, Xu Z, Li J, Yang S, Yang W, Li G (2011) Enalapril, irbesartan, and angiotensin-(1-7) prevent atrial tachycardia-induced ionic remodeling. Int J Cardiol 146 (3):364-370. doi:10.1016/j.ijcard.2009.07.015
  35. 35. Perlini S, Salinaro F, Fonte ML (2008) Direct renin inhibition: another weapon to modulate the renin-angiotensin system in postinfarction remodeling? Hypertension 52 (6):1019-1021. doi:10.1161/HYPERTENSIONAHA.108.121590
  36. 36. Tsai CF, Chen YC, Lin YK, Chen SA, Chen YJ (2011) Electromechanical effects of the direct renin inhibitor (aliskiren) on the pulmonary vein and atrium. Basic Res Cardiol 106 (6):979-993. doi:10.1007/s00395-011-0206-8
  37. 37. Calkins H, el-Atassi R, Kalbfleisch S, Langberg J, Morady F (1992) Effects of an acute increase in atrial pressure on atrial refractoriness in humans. Pacing Clin Electrophysiol 15 (11 Pt 1):1674-1680
  38. 38. Solti F, Vecsey T, Kekesi V, Juhasz-Nagy A (1989) The effect of atrial dilatation on the genesis of atrial arrhythmias. Cardiovasc Res 23 (10):882-886
  39. 39. Murgatroyd FD, Camm AJ (1993) Atrial arrhythmias. Lancet 341 (8856):1317-1322
  40. 40. Ravelli F, Allessie M (1997) Effects of atrial dilatation on refractory period and vulnerability to atrial fibrillation in the isolated Langendorff-perfused rabbit heart. Circulation 96 (5):1686-1695
  41. 41. Matsuda Y, Toma Y, Matsuzaki M, Moritani K, Satoh A, Shiomi K, Ohtani N, Kohno M, Fujii T, Katayama K, et al. (1990) Change of left atrial systolic pressure waveform in relation to left ventricular end-diastolic pressure. Circulation 82 (5):1659-1667
  42. 42. Chatterjee K, Parmley WW, Cohn JN, Levine TB, Awan NA, Mason DT, Faxon DP, Creager M, Gavras HP, Fouad FM, et al. (1985) A cooperative multicenter study of captopril in congestive heart failure: hemodynamic effects and long-term response. Am Heart J 110 (2):439-447
  43. 43. Fitzpatrick MA, Rademaker MT, Charles CJ, Yandle TG, Espiner EA, Ikram H (1992) Angiotensin II receptor antagonism in ovine heart failure: acute hemodynamic, hormonal, and renal effects. Am J Physiol 263 (1 Pt 2):H250-256
  44. 44. Rademaker MT, Charles CJ, Espiner EA, Frampton CM, Nicholls MG, Richards AM (2004) Combined inhibition of angiotensin II and endothelin suppresses the brain natriuretic peptide response to developing heart failure. Clin Sci (Lond) 106 (6):569-576. doi:10.1042/CS20030366
  45. 45. de Graeff PA, Kingma JH, Dunselman PH, Wesseling H, Lie KI (1987) Acute hemodynamic and hormonal effects of ramipril in chronic congestive heart failure and comparison with captopril. Am J Cardiol 59 (10):164D-170D
  46. 46. Burstein B, Nattel S (2008) Atrial structural remodeling as an antiarrhythmic target. J Cardiovasc Pharmacol 52 (1):4-10
  47. 47. Burstein B, Nattel S (2008) Atrial fibrosis: mechanisms and clinical relevance in atrial fibrillation. J Am Coll Cardiol 51 (8):802-809
  48. 48. Corradi D, Callegari S, Maestri R, Benussi S, Alfieri O (2008) Structural remodeling in atrial fibrillation. Nat Clin Pract Cardiovasc Med 5 (12):782-796. doi:10.1038/ncpcardio1370
  49. 49. Sadoshima J, Izumo S (1993) Molecular characterization of angiotensin II--induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts. Critical role of the AT1 receptor subtype. Circ Res 73 (3):413-423
  50. 50. Crabos M, Roth M, Hahn AW, Erne P (1994) Characterization of angiotensin II receptors in cultured adult rat cardiac fibroblasts. Coupling to signaling systems and gene expression. J Clin Invest 93 (6):2372-2378. doi:10.1172/JCI117243
  51. 51. Zhou G, Kandala JC, Tyagi SC, Katwa LC, Weber KT (1996) Effects of angiotensin II and aldosterone on collagen gene expression and protein turnover in cardiac fibroblasts. Mol Cell Biochem 154 (2):171-178
  52. 52. Lin CS, Pan CH (2008) Regulatory mechanisms of atrial fibrotic remodeling in atrial fibrillation. Cell Mol Life Sci 65 (10):1489-1508
  53. 53. Dostal DE (2001) Regulation of cardiac collagen: angiotensin and cross-talk with local growth factors. Hypertension 37 (3):841-844
  54. 54. Lee AA, Dillmann WH, McCulloch AD, Villarreal FJ (1995) Angiotensin II stimulates the autocrine production of transforming growth factor-beta 1 in adult rat cardiac fibroblasts. J Mol Cell Cardiol 27 (10):2347-2357
  55. 55. Lijnen PJ, Petrov VV, Fagard RH (2000) Induction of cardiac fibrosis by transforming growth factor-beta(1). Mol Genet Metab 71 (1-2):418-435. doi:10.1006/mgme.2000.3032
  56. 56. Nakajima H, Nakajima HO, Salcher O, Dittie AS, Dembowsky K, Jing S, Field LJ (2000) Atrial but not ventricular fibrosis in mice expressing a mutant transforming growth factor-beta(1) transgene in the heart. Circ Res 86 (5):571-579
  57. 57. Kallergis EM, Manios EG, Kanoupakis EM, Mavrakis HE, Arfanakis DA, Maliaraki NE, Lathourakis CE, Chlouverakis GI, Vardas PE (2008) Extracellular matrix alterations in patients with paroxysmal and persistent atrial fibrillation: biochemical assessment of collagen type-I turnover. J Am Coll Cardiol 52 (3):211-215. doi:10.1016/j.jacc.2008.03.045
  58. 58. Goette A, Staack T, Rocken C, Arndt M, Geller JC, Huth C, Ansorge S, Klein HU, Lendeckel U (2000) Increased expression of extracellular signal-regulated kinase and angiotensin-converting enzyme in human atria during atrial fibrillation. J Am Coll Cardiol 35 (6):1669-1677
  59. 59. Cardin S, Li D, Thorin-Trescases N, Leung TK, Thorin E, Nattel S (2003) Evolution of the atrial fibrillation substrate in experimental congestive heart failure: angiotensin-dependent and -independent pathways. Cardiovasc Res 60 (2):315-325
  60. 60. Xiao HD, Fuchs S, Campbell DJ, Lewis W, Dudley SC, Jr., Kasi VS, Hoit BD, Keshelava G, Zhao H, Capecchi MR, Bernstein KE (2004) Mice with cardiac-restricted angiotensin-converting enzyme (ACE) have atrial enlargement, cardiac arrhythmia, and sudden death. Am J Pathol 165 (3):1019-1032. doi:10.1016/S0002-9440(10)63363-9
  61. 61. Sakabe M, Fujiki A, Nishida K, Sugao M, Nagasawa H, Tsuneda T, Mizumaki K, Inoue H (2004) Enalapril prevents perpetuation of atrial fibrillation by suppressing atrial fibrosis and over-expression of connexin43 in a canine model of atrial pacing-induced left ventricular dysfunction. J Cardiovasc Pharmacol 43 (6):851-859
  62. 62. Li Y, Li W, Yang B, Han W, Dong D, Xue J, Li B, Yang S, Sheng L (2007) Effects of Cilazapril on atrial electrical, structural and functional remodeling in atrial fibrillation dogs. J Electrocardiol 40 (1):100 e101-106. doi:10.1016/j.jelectrocard.2006.04.001
  63. 63. Okazaki H, Minamino T, Tsukamoto O, Kim J, Okada K, Myoishi M, Wakeno M, Takashima S, Mochizuki N, Kitakaze M (2006) Angiotensin II type 1 receptor blocker prevents atrial structural remodeling in rats with hypertension induced by chronic nitric oxide inhibition. Hypertens Res 29 (4):277-284
  64. 64. Kumagai K, Nakashima H, Urata H, Gondo N, Arakawa K, Saku K (2003) Effects of angiotensin II type 1 receptor antagonist on electrical and structural remodeling in atrial fibrillation. J Am Coll Cardiol 41 (12):2197-2204
  65. 65. Nattel S, Maguy A, Le Bouter S, Yeh YH (2007) Arrhythmogenic ion-channel remodeling in the heart: heart failure, myocardial infarction, and atrial fibrillation. Physiol Rev 87 (2):425-456. doi:10.1152/physrev.00014.2006
  66. 66. Yue L, Feng J, Gaspo R, Li GR, Wang Z, Nattel S (1997) Ionic remodeling underlying action potential changes in a canine model of atrial fibrillation. Circ Res 81 (4):512-525
  67. 67. Laszlo R, Eick C, Rueb N, Weretka S, Weig HJ, Schreieck J, Bosch RF (2008) Inhibition of the renin-angiotensin system: effects on tachycardia-induced early electrical remodelling in rabbit atrium. J Renin Angiotensin Aldosterone System 9 (3):125-132. doi:10.1177/1470320308095262
  68. 68. Doronin SV, Potapova IA, Lu Z, Cohen IS (2004) Angiotensin receptor type 1 forms a complex with the transient outward potassium channel Kv4.3 and regulates its gating properties and intracellular localization. J Biol Chem 279 (46):48231-48237. doi:10.1074/jbc.M405789200
  69. 69. Nakashima H, Kumagai K, Urata H, Gondo N, Ideishi M, Arakawa K (2000) Angiotensin II antagonist prevents electrical remodeling in atrial fibrillation. Circulation 101 (22):2612-2617
  70. 70. Valderrabano M (2007) Influence of anisotropic conduction properties in the propagation of the cardiac action potential. Prog Biophys Mol Biol 94 (1-2):144-168. doi:10.1016/j.pbiomolbio.2007.03.014
  71. 71. Tsai CT, Lai LP, Hwang JJ, Lin JL, Chiang FT (2008) Molecular genetics of atrial fibrillation. J Am Coll Cardiol 52 (4):241-250
  72. 72. Emdad L, Uzzaman M, Takagishi Y, Honjo H, Uchida T, Severs NJ, Kodama I, Murata Y (2001) Gap junction remodeling in hypertrophied left ventricles of aortic-banded rats: prevention by angiotensin II type 1 receptor blockade. J Mol Cell Cardiol 33 (2):219-231. doi:10.1006/jmcc.2000.1293
  73. 73. Fischer R, Dechend R, Gapelyuk A, Shagdarsuren E, Gruner K, Gruner A, Gratze P, Qadri F, Wellner M, Fiebeler A, Dietz R, Luft FC, Muller DN, Schirdewan A (2007) Angiotensin II-induced sudden arrhythmic death and electrical remodeling. American J Physiol Heart Circ Physiol 293 (2):H1242-1253. doi:10.1152/ajpheart.01400.2006
  74. 74. Mayama T, Matsumura K, Lin H, Ogawa K, Imanaga I (2007) Remodelling of cardiac gap junction connexin 43 and arrhythmogenesis. Exp Clin Cardiol 12 (2):67-76
  75. 75. Chen YJ, Chen YC, Tai CT, Yeh HI, Lin CI, Chen SA (2006) Angiotensin II and angiotensin II receptor blocker modulate the arrhythmogenic activity of pulmonary veins. Br J Pharmacol 147 (1):12-22. doi:10.1038/sj.bjp.0706445
  76. 76. Haissaguerre M, Jais P, Shah DC, Takahashi A, Hocini M, Quiniou G, Garrigue S, Le Mouroux A, Le Metayer P, Clementy J (1998) Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 339 (10):659-666
  77. 77. Cheng CC, Huang CF, Chen YC, Lin YK, Kao YH, Chen YJ, Chen SA (2011) Heat-stress responses modulate beta-adrenergic agonist and angiotensin II effects on the arrhythmogenesis of pulmonary vein cardiomyocytes. J Cardiovasc Electrophysiol 22 (2):183-190. doi:10.1111/j.1540-8167.2010.01849.x
  78. 78. Reil JC, Hohl M, Selejan S, Lipp P, Drautz F, Kazakow A, Munz BM, Muller P, Steendijk P, Reil GH, Allessie MA, Bohm M, Neuberger HR (2012) Aldosterone promotes atrial fibrillation. Eur Heart J 33 (16):2098-2108. doi:10.1093/eurheartj/ehr266
  79. 79. Lammers C, Dartsch T, Brandt MC, Rottlander D, Halbach M, Peinkofer G, Ockenpoehler S, Weiergraeber M, Schneider T, Reuter H, Muller-Ehmsen J, Hescheler J, Hoppe UC, Zobel C (2012) Spironolactone prevents aldosterone induced increased duration of atrial fibrillation in rat. Cell Physiol Biochem 29 (5-6):833-840. doi:10.1159/000178483
  80. 80. Gensini F, Padeletti L, Fatini C, Sticchi E, Gensini GF, Michelucci A (2003) Angiotensin-converting enzyme and endothelial nitric oxide synthase polymorphisms in patients with atrial fibrillation. Pacing Clin Electrophysiol 26 (1 Pt 2):295-298
  81. 81. Darbar D, Motsinger AA, Ritchie MD, Gainer JV, Roden DM (2007) Polymorphism modulates symptomatic response to antiarrhythmic drug therapy in patients with lone atrial fibrillation. Heart Rhythm 4 (6):743-749
  82. 82. Tsai CT, Lai LP, Lin JL, Chiang FT, Hwang JJ, Ritchie MD, Moore JH, Hsu KL, Tseng CD, Liau CS, Tseng YZ (2004) Renin-angiotensin system gene polymorphisms and atrial fibrillation. Circulation 109 (13):1640-1646
  83. 83. Ravn LS, Benn M, Nordestgaard BG, Sethi AA, Agerholm-Larsen B, Jensen GB, Tybjaerg-Hansen A (2008) Angiotensinogen and ACE gene polymorphisms and risk of atrial fibrillation in the general population. Pharmacogenet Genomics 18 (6):525-533. doi:10.1097/FPC.0b013e3282fce3bd
  84. 84. Tsai CT, Hwang JJ, Chiang FT, Wang YC, Tseng CD, Tseng YZ, Lin JL (2008) Renin-angiotensin system gene polymorphisms and atrial fibrillation: a regression approach for the detection of gene-gene interactions in a large hospitalized population. Cardiology 111 (1):1-7. doi:10.1159/000113419
  85. 85. Amir O, Amir RE, Paz H, Mor R, Sagiv M, Lewis BS (2008) Aldosterone synthase gene polymorphism as a determinant of atrial fibrillation in patients with heart failure. Am J Cardiol 102 (3):326-329. doi:10.1016/j.amjcard.2008.03.063
  86. 86. Sun X, Yang J, Hou X, Li J, Shi Y, Jing Y (2011) Relationship between -344T/C polymorphism in the aldosterone synthase gene and atrial fibrillation in patients with essential hypertension. J Renin Angiotensin Aldosterone System 12 (4):557-563. doi:10.1177/1470320311417654
  87. 87. Vermes E, Tardif JC, Bourassa MG, Racine N, Levesque S, White M, Guerra PG, Ducharme A (2003) Enalapril decreases the incidence of atrial fibrillation in patients with left ventricular dysfunction: insight from the Studies Of Left Ventricular Dysfunction (SOLVD) trials. Circulation 107 (23):2926-2931
  88. 88. Maggioni AP, Latini R, Carson PE, Singh SN, Barlera S, Glazer R, Masson S, Cere E, Tognoni G, Cohn JN (2005) Valsartan reduces the incidence of atrial fibrillation in patients with heart failure: results from the Valsartan Heart Failure Trial (Val-HeFT). Am Heart J 149 (3):548-557. doi:10.1016/j.ahj.2004.09.033
  89. 89. Ducharme A, Swedberg K, Pfeffer MA, Cohen-Solal A, Granger CB, Maggioni AP, Michelson EL, McMurray JJ, Olsson L, Rouleau JL, Young JB, Olofsson B, Puu M, Yusuf S (2006) Prevention of atrial fibrillation in patients with symptomatic chronic heart failure by candesartan in the Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity (CHARM) program. Am Heart J 152 (1):86-92
  90. 90. Pedersen OD, Bagger H, Kober L, Torp-Pedersen C (1999) Trandolapril reduces the incidence of atrial fibrillation after acute myocardial infarction in patients with left ventricular dysfunction. Circulation 100 (4):376-380
  91. 91. Pizzetti F, Turazza FM, Franzosi MG, Barlera S, Ledda A, Maggioni AP, Santoro L, Tognoni G (2001) Incidence and prognostic significance of atrial fibrillation in acute myocardial infarction: the GISSI-3 data. Heart 86 (5):527-532
  92. 92. Hansson L, Lindholm LH, Ekbom T, Dahlof B, Lanke J, Schersten B, Wester PO, Hedner T, de Faire U (1999) Randomised trial of old and new antihypertensive drugs in elderly patients: cardiovascular mortality and morbidity the Swedish Trial in Old Patients with Hypertension-2 study. Lancet 354 (9192):1751-1756
  93. 93. Hansson L, Lindholm LH, Niskanen L, Lanke J, Hedner T, Niklason A, Luomanmaki K, Dahlof B, de Faire U, Morlin C, Karlberg BE, Wester PO, Bjorck JE (1999) Effect of angiotensin-converting-enzyme inhibition compared with conventional therapy on cardiovascular morbidity and mortality in hypertension: the Captopril Prevention Project (CAPPP) randomised trial. Lancet 353 (9153):611-616
  94. 94. L'Allier PL, Ducharme A, Keller PF, Yu H, Guertin MC, Tardif JC (2004) Angiotensin-converting enzyme inhibition in hypertensive patients is associated with a reduction in the occurrence of atrial fibrillation. J Am Coll Cardiol 44 (1):159-164
  95. 95. Wachtell K, Hornestam B, Lehto M, Slotwiner DJ, Gerdts E, Olsen MH, Aurup P, Dahlof B, Ibsen H, Julius S, Kjeldsen SE, Lindholm LH, Nieminen MS, Rokkedal J, Devereux RB (2005) Cardiovascular morbidity and mortality in hypertensive patients with a history of atrial fibrillation: The Losartan Intervention For End Point Reduction in Hypertension (LIFE) study. J Am Coll Cardiol 45 (5):705-711
  96. 96. Schaer BA, Schneider C, Jick SS, Conen D, Osswald S, Meier CR (2010) Risk for incident atrial fibrillation in patients who receive antihypertensive drugs: a nested case-control study. Ann Intern Med 152 (2):78-84. doi:10.1059/0003-4819-152-2-201001190-00005
  97. 97. Salehian O, Healey J, Stambler B, Alnemer K, Almerri K, Grover J, Bata I, Mann J, Matthew J, Pogue J, Yusuf S, Dagenais G, Lonn E (2007) Impact of ramipril on the incidence of atrial fibrillation: results of the Heart Outcomes Prevention Evaluation study. Am Heart J 154 (3):448-453. doi:10.1016/j.ahj.2007.04.062
  98. 98. Investigators AI, Yusuf S, Healey JS, Pogue J, Chrolavicius S, Flather M, Hart RG, Hohnloser SH, Joyner CD, Pfeffer MA, Connolly SJ (2011) Irbesartan in patients with atrial fibrillation. N Engl J Med 364 (10):928-938. doi:10.1056/NEJMoa1008816
  99. 99. Schmieder RE, Kjeldsen SE, Julius S, McInnes GT, Zanchetti A, Hua TA (2008) Reduced incidence of new-onset atrial fibrillation with angiotensin II receptor blockade: the VALUE trial. J Hypertens 26 (3):403-411
  100. 100. Mathew JP, Fontes ML, Tudor IC, Ramsay J, Duke P, Mazer CD, Barash PG, Hsu PH, Mangano DT (2004) A multicenter risk index for atrial fibrillation after cardiac surgery. Jama 291 (14):1720-1729
  101. 101. Ozaydin M, Dede O, Varol E, Kapan S, Turker Y, Peker O, Duver H, Ibrisim E (2008) Effect of renin-angiotensin aldosteron system blockers on postoperative atrial fibrillation. Int J Cardiol 127 (3):362-367
  102. 102. White CM, Kluger J, Lertsburapa K, Faheem O, Coleman CI (2007) Effect of preoperative angiotensin converting enzyme inhibitor or angiotensin receptor blocker use on the frequency of atrial fibrillation after cardiac surgery: a cohort study from the atrial fibrillation suppression trials II and III. Eur J Cardiothorac Surg 31 (5):817-820. doi:10.1016/j.ejcts.2007.02.010
  103. 103. Van Den Berg MP, Crijns HJ, Van Veldhuisen DJ, Griep N, De Kam PJ, Lie KI (1995) Effects of lisinopril in patients with heart failure and chronic atrial fibrillation. J Card Fail 1 (5):355-363
  104. 104. Dagres N, Karatasakis G, Panou F, Athanassopoulos G, Maounis T, Tsougos E, Kourea K, Malakos I, Kremastinos DT, Cokkinos DV (2006) Pre-treatment with Irbesartan attenuates left atrial stunning after electrical cardioversion of atrial fibrillation. Eur Heart J 27 (17):2062-2068
  105. 105. Madrid AH, Bueno MG, Rebollo JM, Marin I, Pena G, Bernal E, Rodriguez A, Cano L, Cano JM, Cabeza P, Moro C (2002) Use of irbesartan to maintain sinus rhythm in patients with long-lasting persistent atrial fibrillation: a prospective and randomized study. Circulation 106 (3):331-336
  106. 106. Madrid AH, Marin IM, Cervantes CE, Morell EB, Estevez JE, Moreno G, Parajon JR, Peng J, Limon L, Nannini S, Moro C (2004) Prevention of recurrences in patients with lone atrial fibrillation. The dose-dependent effect of angiotensin II receptor blockers. J Renin Angiotensin Aldosterone System 5 (3):114-120
  107. 107. Ueng KC, Tsai TP, Yu WC, Tsai CF, Lin MC, Chan KC, Chen CY, Wu DJ, Lin CS, Chen SA (2003) Use of enalapril to facilitate sinus rhythm maintenance after external cardioversion of long-standing persistent atrial fibrillation. Results of a prospective and controlled study. Eur Heart J 24 (23):2090-2098
  108. 108. Tveit A, Grundvold I, Olufsen M, Seljeflot I, Abdelnoor M, Arnesen H, Smith P (2007) Candesartan in the prevention of relapsing atrial fibrillation. Int J Cardiol 120 (1):85-91. doi:10.1016/j.ijcard.2006.08.086
  109. 109. Belluzzi F, Sernesi L, Preti P, Salinaro F, Fonte ML, Perlini S (2009) Prevention of recurrent lone atrial fibrillation by the angiotensin-II converting enzyme inhibitor ramipril in normotensive patients. J Am Coll Cardiol 53 (1):24-29
  110. 110. Katritsis DG, Toumpoulis IK, Giazitzoglou E, Korovesis S, Karabinos I, Paxinos G, Zambartas C, Anagnostopoulos CE (2005) Latent arterial hypertension in apparently lone atrial fibrillation. J Interv Card Electrophysiol 13 (3):203-207
  111. 111. Richter B, Derntl M, Marx M, Lercher P, Gossinger HD (2007) Therapy with angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, and statins: no effect on ablation outcome after ablation of atrial fibrillation. Am Heart J 153 (1):113-119. doi:10.1016/j.ahj.2006.09.006
  112. 112. Al Chekakie MO, Akar JG, Wang F, Al Muradi H, Wu J, Santucci P, Varma N, Wilber DJ (2007) The effects of statins and renin-angiotensin system blockers on atrial fibrillation recurrence following antral pulmonary vein isolation. J Cardiovasc Electrophysiol 18 (9):942-946. doi:10.1111/j.1540-8167.2007.00887.x
  113. 113. Park JH, Oh YS, Kim JH, Chung WB, Oh SS, Lee DH, Choi YS, Shin WS, Park CS, Youn HJ, Chung WS, Lee MY, Seung KB, Rho TH, Hong SJ (2009) Effect of Angiotensin converting enzyme inhibitors and Angiotensin receptor blockers on patients following ablation of atrial fibrillation. Korean Circ J 39 (5):185-189. doi:10.4070/kcj.2009.39.5.185
  114. 114. Zheng B, Kang J, Tian Y, Tang R, Long D, Yu R, He H, Zhang M, Shi L, Tao H, Liu X, Dong J, Ma C (2009) Angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers have no beneficial effect on ablation outcome in chronic persistent atrial fibrillation. Acta Cardiol 64 (3):335-340
  115. 115. Patel D, Mohanty P, Di Biase L, Wang Y, Shaheen MH, Sanchez JE, Horton RP, Gallinghouse GJ, Zagrodzky JD, Bailey SM, Burkhardt JD, Lewis WR, Diaz A, Beheiry S, Hongo R, Al-Ahmad A, Wang P, Schweikert R, Natale A (2010) The impact of statins and renin-angiotensin-aldosterone system blockers on pulmonary vein antrum isolation outcomes in post-menopausal females. Europace 12 (3):322-330. doi:10.1093/europace/eup387
  116. 116. Folkeringa RJ, Crijns HJ (2010) Do non-antiarrhythmic drugs have enough pleiotropic power to reduce atrial fibrillation? Europace 12 (3):299-300. doi:10.1093/europace/euq009
  117. 117. Yin Y, Dalal D, Liu Z, Wu J, Liu D, Lan X, Dai Y, Su L, Ling Z, She Q, Luo K, Woo K, Dong J (2006) Prospective randomized study comparing amiodarone vs. amiodarone plus losartan vs. amiodarone plus perindopril for the prevention of atrial fibrillation recurrence in patients with lone paroxysmal atrial fibrillation. Eur Heart J 27 (15):1841-1846. doi:10.1093/eurheartj/ehl135
  118. 118. Fogari R, Mugellini A, Destro M, Corradi L, Zoppi A, Fogari E, Rinaldi A (2006) Losartan and prevention of atrial fibrillation recurrence in hypertensive patients. J Cardiovasc Pharmacol 47 (1):46-50
  119. 119. Fogari R, Mugellini A, Zoppi A, Preti P, Destro M, Lazzari P, Derosa G (2012) Effect of telmisartan and ramipril on atrial fibrillation recurrence and severity in hypertensive patients with metabolic syndrome and recurrent symptomatic paroxysmal and persistent atrial fibrillation. J Cardiovasc Pharmacol Ther 17 (1):34-43. doi:10.1177/1074248410395018
  120. 120. Komatsu T, Ozawa M, Tachibana H, Sato Y, Orii M, Kunugida F, Nakamura M (2008) Combination therapy with amiodarone and enalapril in patients with paroxysmal atrial fibrillation prevents the development of structural atrial remodeling. Int Heart J 49 (4):435-447
  121. 121. Murray KT, Rottman JN, Arbogast PG, Shemanski L, Primm RK, Campbell WB, Solomon AJ, Olgin JE, Wilson MJ, Dimarco JP, Beckman KJ, Dennish G, Naccarelli GV, Ray WA (2004) Inhibition of angiotensin II signaling and recurrence of atrial fibrillation in AFFIRM. Heart Rhythm 1 (6):669-675. doi:10.1016/j.hrthm.2004.08.008
  122. 122. Disertori M, Latini R, Barlera S, Franzosi MG, Staszewsky L, Maggioni AP, Lucci D, Di Pasquale G, Tognoni G (2009) Valsartan for prevention of recurrent atrial fibrillation. N Engl J Med 360 (16):1606-1617
  123. 123. Goette A, Schon N, Kirchhof P, Breithardt G, Fetsch T, Hausler KG, Klein HU, Steinbeck G, Wegscheider K, Meinertz T (2012) Angiotensin II-antagonist in paroxysmal atrial fibrillation (ANTIPAF) trial. Circ Arrhythm Electrophysiol 5 (1):43-51. doi:10.1161/CIRCEP.111.965178
  124. 124. Yamashita T, Inoue H, Okumura K, Kodama I, Aizawa Y, Atarashi H, Ohe T, Ohtsu H, Kato T, Kamakura S, Kumagai K, Kurachi Y, Koretsune Y, Saikawa T, Sakurai M, Sato T, Sugi K, Nakaya H, Hirai M, Hirayama A, Fukatani M, Mitamura H, Yamazaki T, Watanabe E, Ogawa S, Investigators JRI (2011) Randomized trial of angiotensin II-receptor blocker vs. dihydropiridine calcium channel blocker in the treatment of paroxysmal atrial fibrillation with hypertension (J-RHYTHM II study). Europace 13 (4):473-479. doi:10.1093/europace/euq439
  125. 125. Dabrowski R, Szwed H (2012) Antiarrhythmic potential of aldosterone antagonists in atrial fibrillation. Cardiol J 19 (3):223-229
  126. 126. Dabrowski R, Borowiec A, Smolis-Bak E, Kowalik I, Sosnowski C, Kraska A, Kazimierska B, Wozniak J, Zareba W, Szwed H (2010) Effect of combined spironolactone-beta-blocker +/- enalapril treatment on occurrence of symptomatic atrial fibrillation episodes in patients with a history of paroxysmal atrial fibrillation (SPIR-AF study). Am J Cardiol 106 (11):1609-1614
  127. 127. Williams RS, deLemos JA, Dimas V, Reisch J, Hill JA, Naseem RH (2011) Effect of spironolactone on patients with atrial fibrillation and structural heart disease. Clin Cardiol 34 (7):415-419. doi:10.1002/clc.20914
  128. 128. Swedberg K, Zannad F, McMurray JJ, Krum H, van Veldhuisen DJ, Shi H, Vincent J, Pitt B, Investigators E-HS (2012) Eplerenone and atrial fibrillation in mild systolic heart failure: results from the EMPHASIS-HF (Eplerenone in Mild Patients Hospitalization And SurvIval Study in Heart Failure) study. J Am Coll Cardiol 59 (18):1598-1603. doi:10.1016/j.jacc.2011.11.063
  129. 129. Botto GL, Padeletti L, Santini M, Capucci A, Gulizia M, Zolezzi F, Favale S, Molon G, Ricci R, Biffi M, Russo G, Vimercati M, Corbucci G, Boriani G (2009) Presence and duration of atrial fibrillation detected by continuous monitoring: crucial implications for the risk of thromboembolic events. J Cardiovasc Electrophysiol 20 (3):241-248
  130. 130. Madrid AH, Peng J, Zamora J, Marin I, Bernal E, Escobar C, Munos-Tinoco C, Rebollo JM, Moro C (2004) The role of angiotensin receptor blockers and/or angiotensin converting enzyme inhibitors in the prevention of atrial fibrillation in patients with cardiovascular diseases: meta-analysis of randomized controlled clinical trials. Pacing Clin Electrophysiol 27 (10):1405-1410. doi:10.1111/j.1540-8159.2004.00645.x
  131. 131. Healey JS, Baranchuk A, Crystal E, Morillo CA, Garfinkle M, Yusuf S, Connolly SJ (2005) Prevention of atrial fibrillation with angiotensin-converting enzyme inhibitors and angiotensin receptor blockers: a meta-analysis. J Am Coll Cardiol 45 (11):1832-1839
  132. 132. Anand K, Mooss AN, Hee TT, Mohiuddin SM (2006) Meta-analysis: inhibition of renin-angiotensin system prevents new-onset atrial fibrillation. Am Heart J 152 (2):217-222
  133. 133. Jibrini MB, Molnar J, Arora RR (2008) Prevention of atrial fibrillation by way of abrogation of the renin-angiotensin system: a systematic review and meta-analysis. Am J Ther 15 (1):36-43. doi:10.1097/MJT.0b013e31804beb59
  134. 134. Kalus JS, Coleman CI, White CM (2006) The impact of suppressing the renin-angiotensin system on atrial fibrillation. J Clin Pharmacol 46 (1):21-28
  135. 135. Schneider MP, Hua TA, Bohm M, Wachtell K, Kjeldsen SE, Schmieder RE (2010) Prevention of atrial fibrillation by Renin-Angiotensin system inhibition a meta-analysis. J Am Coll Cardiol 55 (21):2299-2307
  136. 136. Disertori M, Barlera S, Staszewsky L, Latini R, Quintarelli S, Franzosi MG (2012) Systematic review and meta-analysis: renin-Angiotensin system inhibitors in the prevention of atrial fibrillation recurrences: an unfulfilled hope. Cardiovasc Drugs Ther 26 (1):47-54. doi:10.1007/s10557-011-6346-0
  137. 137. Liu T, Korantzopoulos P, Xu G, Shehata M, Li D, Wang X, Li G (2011) Association between angiotensin-converting enzyme insertion/deletion gene polymorphism and atrial fibrillation: a meta-analysis. Europace 13 (3):346-354. doi:10.1093/europace/euq407
  138. 138. Huang G, Xu JB, Liu JX, He Y, Nie XL, Li Q, Hu YM, Zhao SQ, Wang M, Zhang WY, Liu XR, Wu T, Arkin A, Zhang TJ (2011) Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers decrease the incidence of atrial fibrillation: a meta-analysis. Eur J Clin Invest 41 (7):719-733. doi:10.1111/j.1365-2362.2010.02460.x
  139. 139. Bhuriya R, Singh M, Sethi A, Molnar J, Bahekar A, Singh PP, Khosla S, Arora R (2011) Prevention of recurrent atrial fibrillation with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers: a systematic review and meta-analysis of randomized trials. J Cardiovasc Pharmacol Ther 16 (2):178-184. doi:10.1177/1074248410389045
  140. 140. Zhang Y, Zhang P, Mu Y, Gao M, Wang JR, Wang Y, Su LQ, Hou YL (2010) The role of renin-angiotensin system blockade therapy in the prevention of atrial fibrillation: a meta-analysis of randomized controlled trials. Clin Pharmacol Ther 88 (4):521-531
  141. 141. Khatib R, Joseph P, Briel M, Yusuf S, Healey J (2012) Blockade of the renin-angiotensin-aldosterone system (RAAS) for primary prevention of non-valvular atrial fibrillation: A systematic review and meta analysis of randomized controlled trials. Int J Cardiol. doi:10.1016/j.ijcard.2012.02.009
  142. 142. Kintscher U (2009) ONTARGET, TRANSCEND, and PRoFESS: new-onset diabetes, atrial fibrillation, and left ventricular hypertrophy. J Hypertens Suppl 27 (2):S36-39. doi:10.1097/01.hjh.0000354519.67451.96
  143. 143. Coleman CI, Makanji S, Kluger J, White CM (2007) Effect of angiotensin-converting enzyme inhibitors or angiotensin receptor blockers on the frequency of post-cardiothoracic surgery atrial fibrillation. Ann Pharmacother 41 (3):433-437
  144. 144. Campbell RW (1996) ACE inhibitors and arrhythmias. Heart 76 (3 Suppl 3):79-82
  145. 145. Fletcher RD, Cintron GB, Johnson G, Orndorff J, Carson P, Cohn JN (1993) Enalapril decreases prevalence of ventricular tachycardia in patients with chronic congestive heart failure. The V-HeFT II VA Cooperative Studies Group. Circulation 87 (6 Suppl):VI49-55

Written By

Stefano Perlini, Fabio Belluzzi, Francesco Salinaro and Francesco Musca

Submitted: 15 May 2012 Published: 27 February 2013