Journal Information
Vol. 35. Issue 1.
Pages 41-57 (January 2016)
Visits
16968
Vol. 35. Issue 1.
Pages 41-57 (January 2016)
Review article
Open Access
Statins and oxidative stress in chronic heart failure
Estatinas e stresse oxidativo na insuficiência cardíaca crónica
Visits
16968
Sónia Costaa,1, Marta Reina-Coutoa,b,c,1, António Albino-Teixeiraa,b,
Corresponding author
albinote@med.up.pt

Corresponding authors.
, Teresa Sousaa,b,
Corresponding author
tsousa@med.up.pt

Corresponding authors.
a Departamento de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
b MedInUP – Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Porto, Portugal
c Departamento de Medicina Intensiva, Centro Hospitalar São João, Porto, Portugal
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (7)
Show moreShow less
Tables (3)
Table 1. Pleiotropic effects of statins and associated molecular pathways.
Table 2. Effects of statins in heart failure: non-randomized observational studies (adapted from6–8,84,90).
Table 3. Effects of statins in heart failure with reduced ejection fraction: non-randomized studies (adapted from6–8,69,85,87).
Show moreShow less
Abstract

Statins are the most commonly prescribed drugs for the treatment of dyslipidemia. They are also recommended in primary and secondary prevention of cardiovascular disease. In addition to decreasing cholesterol synthesis, statins interfere with the synthesis of isoprenoid intermediates, which may explain many of their pleiotropic properties, including their antioxidant effects.

Oxidative stress is defined as an imbalance between the synthesis of reactive oxygen species and their elimination by antioxidant defense systems, with a prevailing pro-oxidant status that results in macromolecular damage and disruption of cellular redox signaling. Reactive oxygen species interfere with various processes that affect cardiac structure and function, contributing to the contractile dysfunction, myocardial hypertrophy and fibrosis observed in the pathophysiology of heart failure. By regulating several molecular pathways that control nicotinamide adenine dinucleotide phosphate oxidase and endothelial nitric oxide synthase activity, statins help restore redox homeostasis. These drugs also contribute to the control of inflammation and appear to have a protective role in various diseases. The results of observational studies and clinical trials with statins in heart failure have not been consensual.

This review aims to analyze the role of oxidative stress in heart failure and the molecular mechanisms underlying statins’ antioxidant properties. It also examines current scientific evidence on the use of these drugs as a specific treatment for heart failure.

Keywords:
Statins
Heart failure
Oxidative stress
Endothelial nitric oxide synthase
Nicotinamide adenine dinucleotide phosphate oxidases
Antioxidant/anti-inflammatory effects
Clinical trials
Resumo

As estatinas são os fármacos mais prescritos no tratamento da dislipidemia, estando recomendadas na prevenção primária e secundária das doenças cardiovasculares. Para além de diminuírem a síntese de colesterol, interferem com a síntese de intermediários isoprenoides, o que poderá explicar muitos dos seus efeitos pleiotrópicos, incluindo a ação antioxidante.

O stresse oxidativo é definido como um desequilíbrio entre a formação de espécies reativas de oxigénio e a sua eliminação por sistemas de defesa antioxidantes, com prevalência de um estado pró-oxidante com efeitos deletérios nas macromoléculas orgânicas e na sinalização redox celular. As espécies reativas de oxigénio podem interferir com vários processos que afetam a estrutura e função cardíacas, contribuindo para a disfunção contráctil, fibrose e hipertrofia do miocárdio observadas na fisiopatologia da insuficiência cardíaca. As estatinas promovem a restauração do equilíbrio redox pela regulação de várias vias moleculares responsáveis pelo controlo da atividade de enzimas como a NADPH oxídase e a sintetase endotelial de monóxido de azoto. Estes fármacos contribuem também para o controlo de processos inflamatórios e parecem desempenhar um papel protetor em várias patologias. Os resultados de estudos observacionais e ensaios clínicos, realizados com o objetivo de esclarecer o efeito das estatinas na insuficiência cardíaca, não têm sido consensuais. Esta revisão tem como principal objetivo analisar o papel do stresse oxidativo na insuficiência cardíaca e os mecanismos moleculares inerentes às propriedades antioxidantes das estatinas. Pretende ainda reunir a evidência científica atual relativa à utilização destes fármacos como terapêutica específica da insuficiência cardíaca.

Palavras-chave:
Estatinas
Insuficiência cardíaca
Stresse oxidativo
Sintetase endotelial de monóxido de azoto
Nicotinamida adenina dinucleótido fosfato oxidase
Efeitos antioxidantes/anti-inflamatórios
Ensaios clínicos
List of abbreviations
8-OHdG

8-hydroxy-2′-deoxyguanosine

15-epi-LXA4

15-epi-lipoxin A4

5-LOX

5-lipoxygenase

ACC

American College of Cardiology

AF

atrial fibrillation

Akt

protein kinase B

AMPK

adenosine monophosphate-activated protein kinase

AP-1

activator protein 1

ASK1

apoptosis signal-regulating kinase 1

BH4

tetrahydrobiopterin

BNP

brain-type natriuretic peptide

CAD

coronary artery disease

CORONA

Controlled Rosuvastatin Multinational Trial in Heart Failure

COX-2

cyclooxygenase 2

CVD

cardiovascular disease

Cu,Zn-SOD

Copper, zinc superoxide dismutase (cytoplasmic isoform; SOD-1)

DDB2

DNA damage-binding protein 2

EC-SOD

extracellular superoxide dismutase (SOD-3)

eNOS

endothelial nitric oxide synthase

ERK1/2

extracellular signal-regulated kinases 1 and 2

ESC

European Society of Cardiology

FPP

farnesyl pyrophosphate

GGPP

geranylgeranyl pyrophosphate

GISSI-HF

Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto miocardico-Heart Failure

GPx

glutathione peroxidase

GSH

glutathione

GTP

guanosine triphosphate

GTPCH

guanosine triphosphate cyclohydrolase

H2O2

hydrogen peroxide

HClO

hypochlorous acid

HDL

high-density lipoprotein

HF

heart failure

HMG-CoA

hydroxymethylglutaryl-coenzyme A

HO-1

heme oxygenase 1

LDL

low-density lipoprotein

LVEF

left ventricular ejection fraction

MAO

monoamine oxidase

MAPK

mitogen-activated protein kinase

MDA

malondialdehyde

MI

myocardial infarction

MMPs

matrix metalloproteinases

MnSOD

manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)

MPO

myeloperoxidase

mRNA

messenger ribonucleic acid

NADPH

nicotinamide adenine dinucleotide phosphate

NF-κB

nuclear factor kappa B

NO

nitric oxide

Nox

nicotinamide adenine dinucleotide phosphate oxidases

NT-proBNP

N-terminal brain-type natriuretic peptide

NYHA

New York Heart Association

O2

superoxide

oxLDL

oxidized LDL

PI3K

phosphoinositide 3-kinase

PKA

protein kinase A

ROS

reactive oxygen species

Ser1177

serine 1177

Ser473

serine 473

Ser633

serine 633

SERCA2

sarcoplasmic Ca2+-ATPase

SOD

superoxide dismutase

XO

xanthine oxidase

Full Text
Introduction

The prevalence of heart failure (HF) in the USA is predicted to increase by 46% between 2012 and 2030.1 In Portugal, mortality rates remain at approximately 50% within five years of diagnosis.2 It is therefore essential to explore the main pathophysiological pathways of HF and to review current therapies.

HF is a complex clinical syndrome that can be defined as an abnormality of cardiac structure or function affecting ventricular filling and ejection,3 in which there is evidence of imbalances in redox status and nitric oxide (NO) production that can be redressed by appropriate therapy.4

In Portugal, hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors or statins (Figure 1) account for 90% of lipid-lowering drugs prescribed,5 but it is not known what proportion are prescribed in HF. Although various observational studies suggest that statins may reduce the morbidity and mortality associated with HF, due in part to their antioxidant properties,6–8 and that coronary artery disease (CAD), in which the value of statin therapy is well established, is responsible for around two-thirds of cases of systolic HF,3 the benefit of statins specifically for HF has been called into question by the results of randomized clinical trials. Their use is also limited by cardiac cachexia and by the paradoxical association of low serum cholesterol with worse HF prognosis.9

HMG-CoA reductase inhibitors (statins) (adapted from94). Lovastatin, simvastatin and pravastatin are of fungal origin, while more recent statins are completely synthetic. Statins of fungal origin are structurally related and have a hydronaphthalene ring in common. Lovastatin and simvastatin are administered as inactive prodrugs while pravastatin is given in the active form. Totally synthetic statins have different structures that may account for their solubility differences in water.'> Chemical structure of <span class=HMG-CoA reductase inhibitors (statins) (adapted from94). Lovastatin, simvastatin and pravastatin are of fungal origin, while more recent statins are completely synthetic. Statins of fungal origin are structurally related and have a hydronaphthalene ring in common. Lovastatin and simvastatin are administered as inactive prodrugs while pravastatin is given in the active form. Totally synthetic statins have different structures that may account for their solubility differences in water.' title='Chemical structure of HMG-CoA reductase inhibitors (statins) (adapted from94). Lovastatin, simvastatin and pravastatin are of fungal origin, while more recent statins are completely synthetic. Statins of fungal origin are structurally related and have a hydronaphthalene ring in common. Lovastatin and simvastatin are administered as inactive prodrugs while pravastatin is given in the active form. Totally synthetic statins have different structures that may account for their solubility differences in water.'/>
Figure 1.

Chemical structure of HMG-CoA reductase inhibitors (statins) (adapted from94).

Lovastatin, simvastatin and pravastatin are of fungal origin, while more recent statins are completely synthetic. Statins of fungal origin are structurally related and have a hydronaphthalene ring in common. Lovastatin and simvastatin are administered as inactive prodrugs while pravastatin is given in the active form. Totally synthetic statins have different structures that may account for their solubility differences in water.

(0.3MB).

Statins’ main mechanism of action is inhibition of HMG-CoA reductase, the rate-limiting enzyme of cholesterol synthesis. Inhibition of this enzyme interferes with the synthesis of mevalonate and isoprenoid intermediates, particularly farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP), which are responsible for the isoprenylation of a wide range of proteins including small proteins associated with guanosine triphosphate (GTP) such as Ras, Rho and Rac (Figure 2). Isoprenylation of these molecules is essential for the covalent attachment, subcellular localization, and intracellular trafficking of membrane-associated proteins.10 Inhibition of the synthesis of isoprenoid intermediates may underlie many of statins’ pleiotropic effects (Table 1),11 including their antioxidant and anti-inflammatory action.

Table 1.

Pleiotropic effects of statins and associated molecular pathways.

Properties of statins  Mechanisms of action 
Anti-inflammatory  MPO (a pro-oxidant enzyme secreted by neutrophils and monocytes under inflammatory conditions)69
↑ 15-epi-lipoxin A4 (an anti-inflammatory and pro-resolving eicosanoid)63
↑ vasodilatory and anti-inflammatory prostacyclins by activating the PLA2-COX pathway97
↑ KLF2 in endothelial cells97
↓ number of inflammatory cells in atherosclerotic plaques by reducing cell adhesion molecules such as ICAM-110 
Antioxidant  Inhibition of platelet Nox275
↓ circulating concentration of the catalytic subunit of Nox247
↑ adiponectin (inhibits Nox activation)47
↑ catalase and ↑ SOD via phosphorylation of Akt41
↑ transcription, protein expression and activity of SOD by repressing DNA of DDB2 (an epigenetic negative regulator of SOD transcription)48
↑ activity of catalase and ↑ glutathione48
↑ activity of the antioxidant enzyme thioredoxin-1 via S-nitrosylation27
mRNA and expression of HO-150
↑ activity of PON1 (responsible for the antioxidant properties of HDL cholesterol)51 
Improvement of endothelial function  mRNA of eNOS39,40
BH4 (cofactor of eNOS)39,40
GTPCH (the rate-limiting enzyme in BH4 synthesis)39,40
↑ activity of eNOS after phosphorylation mediated by PI3K/Akt and PKA39
↑ stability of eNOS mRNA due to polyadenylation42
↑ activity of eNOS after phosphorylation of AMPK44
↓ caveolin-1 (a caveolar protein (lipid domain) that negatively impacts eNOS activity through interaction with the enzyme)45,46
Inhibition of expression of caveolar endothelin-1 (a potent vasoconstrictor and mitogen)10 
Antiarrhythmic  Inhibition of Nox79 
Angiogenic  ↑ proliferation, migration and survival of endothelial progenitor cells10
↑ mobilization of endothelial progenitor cells from the bone marrow and acceleration of vascular structure formation via activation of PI3K/Akt and eNOS10 
Antithrombotic and antiplatelet  Inhibition of platelet Nox275
↑ KLF297
eNOS, ↓ TXA2 and modifications in the cholesterol content of platelet membranes10
↑ proliferation, migration and survival of endothelial progenitor cells10
↑ mobilization of endothelial progenitor cells from the bone marrow and acceleration of vascular structure formation via activation of PI3K/Akt and eNOS10 
Stabilization of atherosclerotic plaques  ↓ plaque size or modification of the physical and chemical properties of the lipid core10
↓ macrophage accumulation and ↓ production of MMPs (proteolytic enzymes)10 
Prevention of smooth muscle cell proliferation  Arresting of the cell cycle between the G1/S phase transition10
Inhibition of the small GTPase RhoA that mediates PDGF-induced SMC proliferation10 
Prevention of cardiac hypertrophy and remodeling  ↓ phosphorylation of Akt and ERK1/2 and of the transcription factor GATA498
↑ expression of mRNA of PPARα, a ligand-activated transcription factor belonging to the nuclear receptor superfamily98 
Immunomodulation  ↓ maturation of antigen-presenting cells via ↓ class II histocompatibility complex molecules99
Inhibition of antigen uptake by antigen-presenting cells99
Inhibition of cytokine secretion via suppression of the transcription factor NF-κB99 
Modulation of ANS  ↓ noradrenaline and ↓ renal sympathetic acvitity100
mRNA and protein expression of angiotensin receptor and Nox subunits100
↓ superoxide in the rostral ventrolateral medulla100 

↑: increased; ↓: decreased; Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; ANS: autonomic nervous system; BH4: tetrahydrobiopterin; COX: cyclooxygenase; DDB2: damage-binding protein 2; eNOS: endothelial nitric oxide synthase; ERK1/2: extracellular signal-regulated kinases 1 and 2; GATA4: transcription factor GATA-4; GTPCH: guanosine triphosphate cyclohydrolase; HDL: high-density lipoprotein; HO-1: heme oxygenase 1; ICAM-1: intercellular adhesion molecule 1; KLF2: Kruppel-like factor 2; MMPs: matrix metalloproteases; MPO: myeloperoxidase; mRNA: messenger RNA; NF-κB: nuclear factor kappa B; NO: nitric oxide; Nox: nicotinamide adenine dinucleotide phosphate oxidases; PDGF: platelet-derived growth factor; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; PLA2: phospholipase A2; PON1: paraoxonase 1; PPARα: peroxisome proliferator-activated receptor alpha; RhoA: Ras homolog gene family, member A; SMC: smooth muscle cells; SOD: superoxide dismutase; TXA2: thromboxane A2.

The main aim of this review is to analyze the role of oxidative stress in HF and the molecular mechanisms underlying statins’ antioxidant properties. It also examines current scientific evidence on the use of these drugs as a specific treatment for HF.

Heart failure and oxidative stress

Several experimental and clinical studies in recent years have demonstrated the importance of oxidative stress in the pathogenesis of HF. Oxidative stress is defined as an imbalance between the synthesis of reactive oxygen species (ROS) and their elimination by antioxidant defense systems, with a prevailing pro-oxidant status that results in macromolecular damage and disrupts cellular redox signaling.12 The ROS family includes free radicals such as superoxide (O2) and the hydroxyl radical (HO) as well as non-radical oxidants such as hydrogen peroxide (H2O2) and hypochlorous acid (HClO).13

Cardiomyocytes, endothelial cells and neutrophils all produce ROS in the heart.14 The principal enzymes involved in this process are mitochondrial oxidases, nicotinamide adenine dinucleotide phosphate oxidases (Nox), xanthine oxidase (XO), endothelial nitric oxide synthase (eNOS), monoamine oxidase (MAO) and myeloperoxidase (MPO).14–16

Mitochondria are the main source of ROS. More than 90% of the total oxygen (O2) consumed by aerobic organisms is utilized by mitochondrial oxidases to produce adenosine triphosphate in a process coupled to the reduction of cellular O2 to water. About 1–4% of the O2 used in these reactions is converted to O2 and H2O2, which can be detrimental to mitochondrial function if not adequately detoxified.13 Cardiomyocytes contain the greatest density of mitochondria in the body, due to the energy requirements of the heart. Cardiac mitochondria in animal models of HF produce more O2 than normal mitochondria.14

Nox are enzyme complexes that catalyze O2 reduction using reduced nicotinamide adenine dinucleotide phosphate (NADPH) as the electron donor. This process produces ROS such as O2 and H2O2.17 These enzymes were originally identified in neutrophils. The structure of Nox in these cells includes a membrane complex made up of the catalytic subunit, gp91phox and the subunit p22phox, the cytoplasmic proteins p47phox, p67phox and p40phox, and Rac, a low molecular weight G protein.17,18 Several homologs of the gp91phox subunit, now known as Nox2, have been identified, and there are seven Nox isoforms, Nox1–5 and the dual oxidases Duox1 and Duox2, which differ in expression, molecular composition, subcellular location and tissue distribution.7,19 Nox1–3 require cytosolic subunits to be translocated to form a membrane complex in order to produce ROS,17 while Nox4 is constitutively active and Nox5 remains inactive until stimulated by calcium.11,17 The most important Nox in CVD are Nox1, Nox2, Nox4 and Nox5.20 The activity of Nox1 and Nox2 can be increased by angiotensin II, growth hormones and cytokines.11,17 In terms of distribution, endothelial cells express Nox2, Nox4 and Nox5, vascular smooth muscle cells express Nox1, Nox4 and Nox5, phagocytes mainly express Nox2,11 and cardiomyocytes mainly produce Nox2 and Nox4.19 The Nox family has been implicated in the pathogenesis of HF due to pressure overload, doxorubicin-induced HF and ischemic and diabetic cardiomyopathy.19

eNOS synthesizes NO and citrulline from O2 and l-arginine. However, in conditions of oxidative stress or reduced availability of the substrate (l-arginine) or the cofactor tetrahydrobiopterin (BH4), eNOS can become uncoupled and produce O2 instead of NO (Figure 3).21 In the heart, eNOS is expressed in endothelial cells and cardiomyocytes. Uncoupling of eNOS appears to contribute to endothelial dysfunction in hypertensive diastolic HF and to ventricular hypertrophy due to pressure overload.14,22

eNOS requires the presence of a cofactor, BH4. When BH4 binds to eNOS, the enzyme is coupled. Degradation of BH4 by ROS, particularly ONOO− leads to the uncoupling of the enzyme and hence the production of O2− instead of NO. In turn, O2− reacts with NO, producing ONOO− and oxidizing BH4, leading to a vicious cycle of eNOS uncoupling. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; Poly(A): polyadenylate; Cav-1: caveolin-1; eNOS: endothelial nitric oxide synthase; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; O2−: superoxide; ONOO−: peroxynitrite; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.'> Effects of oxidative stress on endothelial nitric oxide synthase. The synthesis of NO by <span class=eNOS requires the presence of a cofactor, BH4. When BH4 binds to eNOS, the enzyme is coupled. Degradation of BH4 by ROS, particularly ONOO− leads to the uncoupling of the enzyme and hence the production of O2− instead of NO. In turn, O2− reacts with NO, producing ONOO− and oxidizing BH4, leading to a vicious cycle of eNOS uncoupling. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; Poly(A): polyadenylate; Cav-1: caveolin-1; eNOS: endothelial nitric oxide synthase; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; O2−: superoxide; ONOO−: peroxynitrite; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.' title='Effects of oxidative stress on endothelial nitric oxide synthase. The synthesis of NO by eNOS requires the presence of a cofactor, BH4. When BH4 binds to eNOS, the enzyme is coupled. Degradation of BH4 by ROS, particularly ONOO− leads to the uncoupling of the enzyme and hence the production of O2− instead of NO. In turn, O2− reacts with NO, producing ONOO− and oxidizing BH4, leading to a vicious cycle of eNOS uncoupling. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; Poly(A): polyadenylate; Cav-1: caveolin-1; eNOS: endothelial nitric oxide synthase; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; O2−: superoxide; ONOO−: peroxynitrite; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.'/>
Figure 3.

Effects of oxidative stress on endothelial nitric oxide synthase.

The synthesis of NO by eNOS requires the presence of a cofactor, BH4. When BH4 binds to eNOS, the enzyme is coupled. Degradation of BH4 by ROS, particularly ONOO leads to the uncoupling of the enzyme and hence the production of O2 instead of NO. In turn, O2 reacts with NO, producing ONOO and oxidizing BH4, leading to a vicious cycle of eNOS uncoupling. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; Poly(A): polyadenylate; Cav-1: caveolin-1; eNOS: endothelial nitric oxide synthase; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; O2: superoxide; ONOO: peroxynitrite; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.

(0.34MB).

XO is involved in purine metabolism, catalyzing the conversion of hypoxanthine to xanthine and thence to uric acid. O2 is used as an electron donor in these reactions, producing O2 and H2O2.13 Increased expression and activity of XO has been described in HF, while administration of allopurinol, an XO inhibitor, increased cardiac contractility in an animal model of HF and reduced adverse cardiac remodeling in an experimental model of myocardial infarction (MI).14

The MAO family of enzymes catalyzes the oxidative degradation of neutrotransmitters such as noradrenaline, adrenaline and dopamine, and in the process generates H2O2. When the heart is subjected to neurohormonal and/or chronic hemodynamic stress, the abundance of circulating/tissue monoamines can increase MAO-derived H2O2 production,15 with significant contributions from both isoforms. MAO-A may establish a link with the renin-angiotensin system in the pathophysiology of diabetic cardiomyopathy,23 and there is evidence of cardiomyocyte necrosis when it is over-expressed,24 while MAO-B predominates in chronic hemodynamic overload.25

MPO is an enzyme secreted by activated neutrophils and monocytes in inflammatory conditions and uses H2O2 to produce several oxidizing molecules, including HClO, chloramines and nitrogen dioxides, that cause tissue damage in the heart and blood vessels and contribute to endothelial dysfunction.13 Plasma MPO is elevated in HF patients compared to controls, and its activity is increased in severe chronic HF compared to mild to moderate HF.16,26

Oxidative stress can result not only from increased ROS production but also from dysfunctional antioxidant defenses.21 The most important antioxidant enzymes are superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx).13 Conversion of O2 to H2O2 is catalyzed by SOD isoforms found in the cytoplasm and organelles (Cu,Zn-SOD and SOD-1), mitochondria (SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD and SOD-2) or extracellularly (extracellular superoxide dismutase [EC-SOD] or SOD-3). H2O2 can then be converted to H2O and O2 by catalase in peroxisomes or by GPx in the cytoplasm and mitochondria.13 Other antioxidant enzymes include glutathione reductase, glutathione-S-transferase, the peroxiredoxins and the thioredoxins. The latter regulate the thiol-disulfide status of proteins, affecting their structure and function, and thioredoxin 1 binds directly to and metabolize ROS.27 Heme oxygenase 1 (HO-1) also has antioxidant properties; as ROS formation can be catalyzed by an excess of heme, degradation of this compound by HO-1 reduces oxidative stress.28 The human body also has non-enzyme antioxidant defenses, including glutathione (GSH), vitamins C and E, carotenoids, uric acid, bilirubin and albumin.13 High-density lipoprotein (HDL) is another antioxidant molecule; functional HDL has high levels of antioxidant and anti-inflammatory proteins and enzymes, notably paraoxonase-1, which appears to be involved in inhibition of low-density lipoprotein (LDL) oxidation.29

Although reduced antioxidant enzyme activity has been reported in animal models of HF,30 there are conflicting results in HF patients, with increases, decreases, and no change in antioxidant enzyme activity.31–33 Reductions in vitamin C and carotenoid levels have also been seen in patients with chronic HF.34

ROS are involved in several processes that affect cardiac structure and function, contributing to the genesis and progression of HF.

Reactive oxygen species and cardiac contractility

In the heart, ROS can target the function of various ion channels (L-type calcium, sodium and potassium channels) and depress the activity of the sarcoplasmic reticulum Ca2+ ATPase SERCA2.35 They may also modify proteins that are important in cardiac contractility; for example, the phosphorylation of troponin T by ROS-activated kinases may contribute to reduced cardiac contractility.35

Reactive oxygen species and cardiac hypertrophy and fibrosis

ROS stimulate various enzymes of the mitogen-activated protein kinase (MAPK) family, including the extracellular signal-regulated kinases (ERK 1/2) and apoptosis signal-regulating kinase 1 (ASK1), contributing to the development of cardiac hypertrophy.35 Activation of transcription factors such as nuclear factor kappa B and activator protein 1 (AP-1) is also involved in ROS-induced cardiac hypertrophy.35 ROS can also stimulate proliferation of cardiac fibroblasts and activate extracellular matrix metalloproteinases (MMPs), leading to remodeling of the extracellular matrix. These alterations are important factors in adverse myocardial remodeling.14

Reactive oxygen species and cardiomyocyte apoptosis

ROS can mediate cardiomyocyte apoptosis by a variety of mechanisms, including direct genotoxicity, activation of ASK-1 in response to tumor necrosis factor alpha, and stimulation of kinases that induce mitochondrial death in response to activation of beta-adrenergic receptors.35 Interestingly, induction of cardiomyocyte hypertrophy or apoptosis appears to depend on the level of ROS produced: relatively low levels of H2O2 stimulate protein synthesis, while higher levels induce cardiomyocyte apoptosis.35

Reactive oxygen species and mitochondrial dysfunction

Mitochondria are important sources of ROS in the heart, but they can also be affected by their toxic effects. ROS can damage mitochondrial DNA, reducing transcription and protein synthesis, leading to mitochondrial dysfunction, disruption of cardiac energy metabolism and cell death.14

Reactive oxygen species and ischemic cardiomyopathy

ROS can contribute to the genesis and progression of CAD. ROS produced in the vessel wall are involved in the formation of oxidized LDL (oxLDL), which is central to the pathogenesis of atherosclerosis.35 ROS-associated activation of MMPs may also play an important role in instability and rupture of atheromatous plaque in the coronary arteries, leading to thrombosis.35 ROS are also involved in reperfusion injury and tissue necrosis caused by MI.35

Interaction between reactive oxygen species and reactive nitrogen species

The interaction between ROS and NO can affect cardiac function. NO mediates protein S-nitrosylation at specific cysteine residues, which influences calcium flux and excitation-contraction coupling. High levels of O2 can inhibit protein S-nitrosylation, while the interaction of O2 with NO, as well as contributing to endothelial dysfunction by reducing NO availability, also produces peroxynitrite, a potent oxidant that induces apoptosis or cell necrosis.35,36

Statins and oxidative stressEffects of statins on endothelial nitric oxide synthase

eNOS is the main enzymatic source of NO in blood vessels. NO plays a part in vascular homeostasis, by inhibiting platelet activation and aggregation, vascular smooth muscle cell proliferation, expression of cell adhesion molecules and production of extracellular matrix.37 NO also helps regulate cardiac contractility and heart rate, limits cardiac remodeling after MI, and contributes to the protective effect of ischemic pre- and postconditioning.38

Binding to BH4 is essential for eNOS to synthesize NO. Degradation of this cofactor by ROS leads to uncoupling of the enzyme and production of O2 instead of NO, as mentioned previously11 (Figure 3). De novo synthesis of BH4 requires the enzyme guanosine triphosphate cyclohydrolase (GTPCH), a limiting factor for its formation. In human endothelial cells, fluvastatin and cerivastatin significantly increased the expression of messenger ribonucleic acid (mRNA) of GTPCH and intracellular BH4 concentrations.39,40 These statins also increased eNOS transcription.39,40 In another study, atorvastatin, pravastatin or pitavastatin also increased eNOS expression through stimulation of protein kinase B (Akt) at Ser473, inhibiting endothelial senescence induced by oxidative stress.41

Increased eNOS expression can also result from enhanced mRNA stability due to polyadenylation. Simvastatin and rosuvastatin significantly increased polyadenylation and hence the stability of eNOS mRNA in bovine aortic endothelial cells42 (Figure 4).

eNOS and by preventing eNOS uncoupling, limit the formation of ROS. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; Poly(A): polyadenylate; Cav-1: caveolin-1; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; Poly(A): polyadenylate; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.'> Action of statins on endothelial nitric oxide synthase. Statins stimulate the synthesis of NO by <span class=eNOS and by preventing eNOS uncoupling, limit the formation of ROS. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; Poly(A): polyadenylate; Cav-1: caveolin-1; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; Poly(A): polyadenylate; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.' title='Action of statins on endothelial nitric oxide synthase. Statins stimulate the synthesis of NO by eNOS and by preventing eNOS uncoupling, limit the formation of ROS. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; Poly(A): polyadenylate; Cav-1: caveolin-1; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; Poly(A): polyadenylate; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.'/>
Figure 4.

Action of statins on endothelial nitric oxide synthase.

Statins stimulate the synthesis of NO by eNOS and by preventing eNOS uncoupling, limit the formation of ROS. Akt: protein kinase B; AMPK: adenosine monophosphate-activated protein kinase; mRNA: messenger ribonucleic acid; BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; Poly(A): polyadenylate; Cav-1: caveolin-1; GTPCH-1: guanosine triphosphate cyclohydrolase; NO: nitric oxide; O2: oxygen; P: phosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; Poly(A): polyadenylate; ROS: reactive oxygen species; Ser633: serine 633; Ser1177: serine 1177.

(0.33MB).

Statins can also contribute to increased eNOS activity through phosphorylation. Fluvastatin and pitavastatin promote eNOS phosphorylation at Ser1177 and/or Ser633 via phosphoinositide 3-kinase (PI3K)/Akt and protein kinase A (PKA), respectively, increasing eNOS activity in human endothelial cells.39,43 When adenosine monophosphate-activated protein kinase (AMPK) is stimulated by statins, it can also phosphorylate eNOS at Ser1177, increasing its activity.44 Statins can also enhance eNOS activity by inhibiting the expression of caveolin-1,45 which interacts with eNOS in endothelial cells and reduces its activity.46 Furthermore, they can increase eNOS activity and coupling by reducing concentrations of asymmetric dimethylarginine, an eNOS inhibitor.11

Effects of statins on nicotinamide adenine dinucleotide phosphate oxidases

As stated above, Nox are important sources of ROS and contribute to oxidative stress. Production of ROS by the isoforms Nox2 and Nox1 requires the activation and translocation of cytoplasmic proteins such as Rac, p47phox or its homolog NoxO1, and p67phox or its homolog NoxA1, which then interact with the membrane subunits Nox1 or gp91phox (Nox2) and p22phox. The subunit p40phox is also translocated in Nox2. Activation and translocation of the protein Rac is dependent on isoprenylation by GGPP, which is synthesized via the mevalonate pathway.11 Inhibition of this pathway by statins blocks Rac activation and thereby reduces Nox1 and Nox2 activity11 (Figure 5).

NADPH oxidases are composed of membrane subunits such as p22phox and gp91phox (Nox2), and cytosolic subunits including p47phox, p67phox, p40phox and GTP-Rac. Activation of Nox requires activation and membrane translocation of Rac and p47phox, p67phox and p40phox, while activation of Rac involves isoprenylation by GGPF. Inhibition of the formation of GGPF by statins prevents activation of Rac and hence of Nox2. Another mechanism by which statins appear to inhibit Nox2 is by altering the phosphorylation of PKC and the membrane translocation of p47phox and is partly mediated by adiponectin, which inhibits p47phox translocation. ADP: adenosine diphosphate; FPF: farnesyl pyrophosphate; GDP: guanosine diphosphate; GGPP: geranylgeranyl pyrophosphate; GTP: guanosine triphosphate; HMG-CoA: hydroxymethylglutaryl-coenzyme A; mRNA: messenger ribonucleic acid; Nox: NADPH oxidase; O2: oxygen; O2−: superoxide; PKC: protein kinase C.'> Action of statins on nicotinamide adenine dinucleotide phosphate oxidase in endothelial cells. <span class=NADPH oxidases are composed of membrane subunits such as p22phox and gp91phox (Nox2), and cytosolic subunits including p47phox, p67phox, p40phox and GTP-Rac. Activation of Nox requires activation and membrane translocation of Rac and p47phox, p67phox and p40phox, while activation of Rac involves isoprenylation by GGPF. Inhibition of the formation of GGPF by statins prevents activation of Rac and hence of Nox2. Another mechanism by which statins appear to inhibit Nox2 is by altering the phosphorylation of PKC and the membrane translocation of p47phox and is partly mediated by adiponectin, which inhibits p47phox translocation. ADP: adenosine diphosphate; FPF: farnesyl pyrophosphate; GDP: guanosine diphosphate; GGPP: geranylgeranyl pyrophosphate; GTP: guanosine triphosphate; HMG-CoA: hydroxymethylglutaryl-coenzyme A; mRNA: messenger ribonucleic acid; Nox: NADPH oxidase; O2: oxygen; O2−: superoxide; PKC: protein kinase C.' title='Action of statins on nicotinamide adenine dinucleotide phosphate oxidase in endothelial cells. NADPH oxidases are composed of membrane subunits such as p22phox and gp91phox (Nox2), and cytosolic subunits including p47phox, p67phox, p40phox and GTP-Rac. Activation of Nox requires activation and membrane translocation of Rac and p47phox, p67phox and p40phox, while activation of Rac involves isoprenylation by GGPF. Inhibition of the formation of GGPF by statins prevents activation of Rac and hence of Nox2. Another mechanism by which statins appear to inhibit Nox2 is by altering the phosphorylation of PKC and the membrane translocation of p47phox and is partly mediated by adiponectin, which inhibits p47phox translocation. ADP: adenosine diphosphate; FPF: farnesyl pyrophosphate; GDP: guanosine diphosphate; GGPP: geranylgeranyl pyrophosphate; GTP: guanosine triphosphate; HMG-CoA: hydroxymethylglutaryl-coenzyme A; mRNA: messenger ribonucleic acid; Nox: NADPH oxidase; O2: oxygen; O2−: superoxide; PKC: protein kinase C.'/>
Figure 5.

Action of statins on nicotinamide adenine dinucleotide phosphate oxidase in endothelial cells.

NADPH oxidases are composed of membrane subunits such as p22phox and gp91phox (Nox2), and cytosolic subunits including p47phox, p67phox, p40phox and GTP-Rac. Activation of Nox requires activation and membrane translocation of Rac and p47phox, p67phox and p40phox, while activation of Rac involves isoprenylation by GGPF. Inhibition of the formation of GGPF by statins prevents activation of Rac and hence of Nox2. Another mechanism by which statins appear to inhibit Nox2 is by altering the phosphorylation of PKC and the membrane translocation of p47phox and is partly mediated by adiponectin, which inhibits p47phox translocation. ADP: adenosine diphosphate; FPF: farnesyl pyrophosphate; GDP: guanosine diphosphate; GGPP: geranylgeranyl pyrophosphate; GTP: guanosine triphosphate; HMG-CoA: hydroxymethylglutaryl-coenzyme A; mRNA: messenger ribonucleic acid; Nox: NADPH oxidase; O2: oxygen; O2: superoxide; PKC: protein kinase C.

(0.35MB).

Statins’ inhibitory effect on Nox activity appears to affect other subunits of these enzymes. Reduced mRNA expression of p22phox, gp91phox and Nox1, protein expression of p47phox and translocation of p47phox and p67phox have been observed following statin therapy; they also inhibit the expression of the angiotensin II receptor AT1, which mediates the stimulating effect of angiotensin II on Nox activity.11

It has recently been suggested that statins’ inhibition of Nox may be partly mediated by adiponectin, a protein synthesized by adipocytes. In hypercholesterolemic patients, increases in adiponectin levels following treatment with atorvastatin were associated with decreases in the soluble form of gp91phox, platelet ROS production and urinary isoprostanes. In-vitro treatment with adiponectin also inhibited p47phox translocation and soluble gp91phox cleavage, inhibiting Nox activation in platelets.47

Effects of statins on antioxidant systems

Statins can stimulate antioxidant defenses. In-vitro treatment of human endothelial cells with atorvastatin increased expression of catalase and SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD following phosphorylation of Akt at Ser473.41 In tumor cells, fluvastatin increased mRNA and protein expression of SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD and caused a two-fold increase in SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD activity.48 These effects appear to be due to downregulation of DNA damage-binding protein 2 (DDB2), a negative regulator of SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD transcription.48 Fluvastatin also increased catalase activity and GSH concentrations.48 Increased GSH has also been observed in promyelocytic cells treated with rosuvastatin, and appears to result from upregulation of gamma-glutamylcysteine synthetase, the rate-limiting enzyme of GSH.49

Statins also influence other antioxidant enzymes, such as thioredoxin 1 and HO-1. In human endothelial cells, atorvastatin stimulated NO synthesis and hence S-nitrosylation of thioredoxin 1, increasing its activity and reducing intracellular ROS,27 while rosuvastatin increased mRNA and protein expression of HO-1.50

Statins may have a further protective effect through their action on PON1, an enzyme involved in the antioxidant effect of HDL, the activity of which was increased by atorvastatin in hypercholesterolemic individuals.51

Other antioxidant/anti-inflammatory effects of statins

The relationship between oxidative stress and inflammation has been demonstrated in HF and other chronic diseases, in which it contributes to their genesis and progression.52,53 Oxidative stress activates various transcription factors, including NF-κB and AP-1, that induce the expression of proinflammatory cytokines, chemokines and adhesion molecules.53,54 At the same time, the production of large quantities of ROS is a characteristic of activated inflammatory cells.53 Some cytokines also trigger ROS production in vascular endothelial and smooth muscle cells.55,56

Statins affect various molecules that play an important role in both oxidative stress and inflammation. For example, MPO, which is found in neutrophils and monocytes, contributes to ROS formation during the inflammatory process, thereby triggering lipid oxidation and tissue damage.57 MPO interferes with endothelial homeostasis, contributing to the onset and progression of atherosclerosis,57 and is also associated with worsening New York Heart Association (NYHA) functional class.16 There is evidence that statins strongly inhibit MPO mRNA expression in human and murine monocyte-macrophages, an effect dependent on blockade of the mevalonate pathway and hence reduction the formation of GGPP formation.58

Galectin-3, a member of the lectin family of proteins, is another biomarker associated with oxidative stress and inflammation. It is involved in proliferation, chemotaxis, phagocytosis, apoptosis, angiogenesis and myocardial fibrosis, all of which are mechanisms underlying the pathogenesis of cardiovascular disease (CVD). The release of galectin-3 by monocytes and macrophages is regulated by ROS and Nox. Curiously, in vitro, galectin-3 stimulates synthesis of O2 by monocytes and macrophages, and so it may contribute to the vicious cycle of oxidative stress and inflammation.59 In an animal model of atherosclerosis, galectin-3 expression increased in proportion to the degree of plaque extent and inflammation, and atorvastatin treatment markedly reduced intraplaque galectin-3 and macrophage signals.60

It has recently been shown that statins reduce inflammation by stimulating the synthesis of 15-epi-lipoxin A4 (15-epi-LXA4), which has pro-resolving, anti-inflammatory, antioxidant, vasodilatory and antiproliferative properties61–66 (Figure 6). Synthesis of 15-epi-LXA4 from arachidonic acid involves the sequential action of cyclooxygenase 2 (COX-2) and 5-lipoxygenase (5-LOX). Statins induce expression and S-nitrosylation of COX-2, resulting in the synthesis of 15R-hydroxyeicosatetraenoic acid, which is then converted via 5-LOX to 15-epi-LXA4 (Figure 6). Treatment with atorvastatin significantly increased the synthesis of 15-epi-LXA4 in rat myocardium via S-nitrosylation of COX-2.62 In an animal model of airway inflammation, treatment with lovastatin also induced the formation of 15-epi-LXA4 and markedly reduced acute lung inflammation. Furthermore, in-vitro lovastatin increased 15-epi-LXA4 production during interactions between polymorphonuclear cells and human airway epithelial cells primed with cytokines63 (Figure 7).

COX-2 by statins promotes the synthesis of 15-epi-lipoxins, which have protective effects in various types of cell and help reduce inflammation. ↑: increased; ↓: decreased; AA: arachidonic acid; CCR5: C-C chemokine receptor type 5; CR3: complement receptor 3 (CD11B, cluster of differentiation 11B/CD18, integrin beta-2); COX-2: cyclooxygenase-2; HO-1: heme oxygenase-1; ICAM-1: intercellular adhesion molecule 1; MMP-3: matrix metalloproteinase-3; NO: nitric oxide; O2−: superoxide; ONOO− peroxynitrite; PGI2: prostacyclin; ROS: reactive oxygen species; TNF-α: tumor necrosis factor alpha; VEFG: vascular endothelial growth factor; 5-LOX: 5-lipoxygenase; 15R-HETE: 15-hydroxyicosatetraenoic acid; 15-epi-LXA4: 15-epi-lipoxin A4.'> Statins and synthesis of 15-epi-lipoxins: protective effects. S-nitrosylation of <span class=COX-2 by statins promotes the synthesis of 15-epi-lipoxins, which have protective effects in various types of cell and help reduce inflammation. ↑: increased; ↓: decreased; AA: arachidonic acid; CCR5: C-C chemokine receptor type 5; CR3: complement receptor 3 (CD11B, cluster of differentiation 11B/CD18, integrin beta-2); COX-2: cyclooxygenase-2; HO-1: heme oxygenase-1; ICAM-1: intercellular adhesion molecule 1; MMP-3: matrix metalloproteinase-3; NO: nitric oxide; O2−: superoxide; ONOO− peroxynitrite; PGI2: prostacyclin; ROS: reactive oxygen species; TNF-α: tumor necrosis factor alpha; VEFG: vascular endothelial growth factor; 5-LOX: 5-lipoxygenase; 15R-HETE: 15-hydroxyicosatetraenoic acid; 15-epi-LXA4: 15-epi-lipoxin A4.' title='Statins and synthesis of 15-epi-lipoxins: protective effects. S-nitrosylation of COX-2 by statins promotes the synthesis of 15-epi-lipoxins, which have protective effects in various types of cell and help reduce inflammation. ↑: increased; ↓: decreased; AA: arachidonic acid; CCR5: C-C chemokine receptor type 5; CR3: complement receptor 3 (CD11B, cluster of differentiation 11B/CD18, integrin beta-2); COX-2: cyclooxygenase-2; HO-1: heme oxygenase-1; ICAM-1: intercellular adhesion molecule 1; MMP-3: matrix metalloproteinase-3; NO: nitric oxide; O2−: superoxide; ONOO− peroxynitrite; PGI2: prostacyclin; ROS: reactive oxygen species; TNF-α: tumor necrosis factor alpha; VEFG: vascular endothelial growth factor; 5-LOX: 5-lipoxygenase; 15R-HETE: 15-hydroxyicosatetraenoic acid; 15-epi-LXA4: 15-epi-lipoxin A4.'/>
Figure 6.

Statins and synthesis of 15-epi-lipoxins: protective effects.

S-nitrosylation of COX-2 by statins promotes the synthesis of 15-epi-lipoxins, which have protective effects in various types of cell and help reduce inflammation. ↑: increased; ↓: decreased; AA: arachidonic acid; CCR5: C-C chemokine receptor type 5; CR3: complement receptor 3 (CD11B, cluster of differentiation 11B/CD18, integrin beta-2); COX-2: cyclooxygenase-2; HO-1: heme oxygenase-1; ICAM-1: intercellular adhesion molecule 1; MMP-3: matrix metalloproteinase-3; NO: nitric oxide; O2: superoxide; ONOO peroxynitrite; PGI2: prostacyclin; ROS: reactive oxygen species; TNF-α: tumor necrosis factor alpha; VEFG: vascular endothelial growth factor; 5-LOX: 5-lipoxygenase; 15R-HETE: 15-hydroxyicosatetraenoic acid; 15-epi-LXA4: 15-epi-lipoxin A4.

(0.29MB).
BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; GSH: glutathione; GTPCH: guanosine triphosphate cyclohydrolase; HDL: high-density lipoprotein; HO-1: heme oxygenase-1; SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD: manganese-dependent superoxide dismutase; MPO: myeloperoxidase; mRNA: messenger ribonucleic acid; Nox: nicotinamide adenine dinucleotide phosphate oxidase; PON-1: paraoxonase-1; 15-epi-LXA4: 15-epi-lipoxinA4.'> Summary of statins’ antioxidant and anti-inflammatory effects. ↑: increased; ↓: decreased; ADMA: asymmetric dimethylarginine; <span class=BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; GSH: glutathione; GTPCH: guanosine triphosphate cyclohydrolase; HDL: high-density lipoprotein; HO-1: heme oxygenase-1; SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD: manganese-dependent superoxide dismutase; MPO: myeloperoxidase; mRNA: messenger ribonucleic acid; Nox: nicotinamide adenine dinucleotide phosphate oxidase; PON-1: paraoxonase-1; 15-epi-LXA4: 15-epi-lipoxinA4.' title='Summary of statins’ antioxidant and anti-inflammatory effects. ↑: increased; ↓: decreased; ADMA: asymmetric dimethylarginine; BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; GSH: glutathione; GTPCH: guanosine triphosphate cyclohydrolase; HDL: high-density lipoprotein; HO-1: heme oxygenase-1; SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD: manganese-dependent superoxide dismutase; MPO: myeloperoxidase; mRNA: messenger ribonucleic acid; Nox: nicotinamide adenine dinucleotide phosphate oxidase; PON-1: paraoxonase-1; 15-epi-LXA4: 15-epi-lipoxinA4.'/>
Figure 7.

Summary of statins’ antioxidant and anti-inflammatory effects. ↑: increased; ↓: decreased; ADMA: asymmetric dimethylarginine; BH4: tetrahydrobiopterin; eNOS: endothelial nitric oxide synthase; GSH: glutathione; GTPCH: guanosine triphosphate cyclohydrolase; HDL: high-density lipoprotein; HO-1: heme oxygenase-1; SOD-2)' alt='manganese-dependent superoxide dismutase (mitochondrial isoform; SOD-2)'>MnSOD: manganese-dependent superoxide dismutase; MPO: myeloperoxidase; mRNA: messenger ribonucleic acid; Nox: nicotinamide adenine dinucleotide phosphate oxidase; PON-1: paraoxonase-1; 15-epi-LXA4: 15-epi-lipoxinA4.

(0.43MB).
Statins and oxidative stress in humans

Oxidative stress is associated with aging and the development of many diseases, including CVD.14 Several studies over the last 10 years have assessed the effects of statins on markers of oxidative stress and endothelial function in cardiovascular, renal and metabolic disorders.

In HF patients with reduced left ventricular ejection fraction (LVEF), one month of simvastatin or atorvastatin therapy reduced ROS production and systemic concentrations of malondialdehyde (MDA), a marker of lipid peroxidation, increased EC-SOD activity, and improved endothelial function and functional capacity.67,68 In another study, of patients with systolic HF, one month's treatment with rosuvastatin also significantly reduced plasma MPO and oxLDL levels.69

In individuals with CAD undergoing coronary artery bypass grafting, prospective treatment with atorvastatin or pravastatin for four weeks significantly reduced mRNA expression and activity of Rac1 in myocardial tissue samples, as well as angiotensin II-induced Nox activity.70 In a study of patients undergoing cardiac surgery who were monitored until discharge, a strong association was observed between O2 and peroxynitrite production and postoperative complications during hospital stay. Preoperative atorvastatin therapy for three days significantly reduced Nox activity and O2 and peroxynitrite production in myocardial tissue, while ex-vivo incubation of myocardium with atorvastatin induced mevalonate-reversible and Rac1-mediated inhibition of Nox.71 A similar study showed that preoperative administration of atorvastatin increased BH4 availability and reduced both basal and uncoupled eNOS-derived O2 production in samples of internal mammary artery.72 Statin therapy also appears to reduce plasma MPO concentrations in patients with acute coronary syndrome but not in those with stable CAD.73

Statins’ redox effects have been assessed in individuals with hyperlipidemia, in whom they have various protective effects, including reduced ROS production and increased platelet NO synthesis, decreased urinary and platelet isoprostanes, diminished platelet Rac1 activation, inhibiting of serum and platelet Nox2, increasing adiponectin levels and thereby reducing gp91phox and Nox activity, raised serum vitamin E levels, and decreased DNA damage in hypercholesterolemic individuals with the C242T polymorphism of the p22phox gene, which is associated with risk for developing CAD.47,74–78

Oxidative stress is also linked to the pathogenesis of atrial fibrillation (AF). In a study assessing the effects of statins on ROS production in patients who developed AF following cardiac surgery or with permanent AF, atorvastatin reduced Rac1 and Nox activity in right atrial samples from those who developed postoperative AFF, but did not affect ROS, eNOS uncoupling, or BH4 in those with permanent AF. These results suggest that Nox upregulation is an early but transient event in the natural history of AF.79

Statins also appear to have a protective effect in diabetes. Rosuvastatin treatment for six months in patients with diabetic nephropathy improved renal function and significantly reduced serum lipid peroxidation products and urinary 8-hydroxy-2′-deoxyguanosine (8-OHdG), a marker of DNA oxidation.80 In diabetic polyneuropathy, rosuvastatin for 12 months led to improvements in severity, symptoms and nerve conduction parameters, as well as reductions in lipid peroxidation.81

Despite the growing evidence of statins’ antioxidant effects in cardiovascular and metabolic disorders, these effects are not seen in some studies. In patients at high risk for cardiovascular disease, atorvastatin and simvastatin did not significantly alter plasma MDA or MPO concentrations or urinary 8-OHdG,82 while in another study of patients with diabetic nephropathy, atorvastatin did not increase NO availability.83

Statins in chronic heart failure: clinical trials

Most studies on the effects of statins in HF are observational and non-randomized, and focus on HF with reduced LVEF (≤35%). They suggest that statins reduce mortality (Table 2); a reduction in mortality has also been reported in HF with preserved LVEF after treatment with statins.84

Table 2.

Effects of statins in heart failure: non-randomized observational studies (adapted from6–8,84,90).

Study  No. of subjects  Statin  Duration
(months) 
Results 
Horwich et al. 2004  551  Any statin  24  ↓ mortality and ↓ need for urgent cardiac transplantation 
Mozaffarian et al. 2004  1153  Any statin  15  ↓ mortality 
Hognestad et al. 2004  5301  Any statin  25  ↓ mortality 
Ezekowitz et al. 2004  6427  Any statin  12  ↓ mortality 
Sola et al. 2005  446  Any statin  24  ↓ mortality and ↓ hospitalization 
Folkeringa et al. 2006  524  Any statin  31  ↓ mortality 
Anker et al. 2006  5200  Any statin  12–36  ↓ mortality 
Go et al. 2006  24598  Any statin  29  ↓ mortality and ↓ hospitalization 
Foody et al. 2006  54960  Any statin  36  ↓ mortality 
Dickinson et al. 2007  2521  Any statin  46  ↓ mortality 
Tehrani et al.84  270  Any statin  60  ↓ mortality and ↔ total and cardiovascular hospitalization 
Senthil et al. 2011  10510  Any statin  31  ↓ mortality 
Gastelurrutia et al.90  960  Any statin  44  ↓ mortality 

↓: decreased; ↓: increased; ↔: no change.

However, there have been some randomized clinical trials on statins in HF, the largest being Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto Miocardico-Heart Failure (GISSI-HF)85 and Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA).86

CORONA was a prospective randomized clinical trial of rosuvastatin use in 5011 patients with symptomatic HF (NYHA II–IV) of ischemic etiology, with LVEF ≤40% (NYHA III or IV) or ≤35% (NYHA II) and aged ≥60 years.86 Despite improvements in lipid profile and reduced high-sensitivity C-reactive protein, there were no significant differences between the two groups in cardiovascular mortality, MI or stroke.87

GISSI-HF was a prospective randomized trial of 4574 patients aged ≥18 years with HF in NYHA II–IV, irrespective of cause and LVEF, randomized to rosuvastatin or placebo. Although daily rosuvastatin was safe, it did not affect clinical outcomes (time to death or admission to hospital for cardiovascular reasons) during a median follow-up of 46 months.85

Various explanations have been put forward for the results of CORONA and GISSI-HF. It is possible that not all statins have the same effects, since atorvastatin, unlike rosuvastatin, reduces overall mortality and hospitalizations, improves LVEF88 and reduces systemic B-type natriuretic peptide (BNP), which may be due to differences in the pharmacokinetic properties of different statins.89 Furthermore, a meta-analysis found no correlation between dose and results, suggesting that the type of statin may be more important than the dose.88

The characteristics of the study populations, particularly age (mean age in CORONA and GISSI-HF was 73 and 68 years, respectively) and the exclusion of patients previously medicated with statins, may also have affected the results.90

According to another theory, statins may be beneficial in the early stages of HF but do not prevent progressive deterioration of cardiac function in more advanced stages.91 It would thus be useful to identify patient subgroups who would derive greater benefit from these drugs. Patients treated with rosuvastatin and in the lower tertile (<103 pmol/l or <868 pg/ml) of N-terminal brain-type natriuretic peptide (NT-proBNP) appear to have greater reductions in cardiovascular mortality, MI and stroke.92 Similarly, patients treated with rosuvastatin and with galectin-3 ≤19 ng/ml presented lower rates of primary events, total mortality and HF hospitalizations than placebo; those with higher galectin-3 levels did not present similar benefits.93

The European Society of Cardiology (ESC) and the American College of Cardiology (ACC) do not currently recommend statins as adjuvant therapy in HF in the absence of other indications for their use.2,3

Conclusions

In addition to their effects on cholesterol synthesis, statins have pleiotropic properties, notably antioxidant action. These effects appear to result from inhibition of the synthesis of isoprenoid intermediates of the mevalonate pathway.

There is widespread recognition of the role of oxidative stress in the pathogenesis of many diseases, including HF. ROS interfere with various processes that affect cardiac structure and function, contributing to apoptosis and mitochondrial dysfunction in cardiomyocytes, contractile dysfunction, myocardial fibrosis and hypertrophy, endothelial dysfunction and atherosclerosis. Statins reduce ROS levels and increase NO synthesis, thereby improving cardiac redox balance. These effects are mainly mediated by inhibition of pro-oxidant enzymes such as Nox and by stimulation of the expression, activity and stability of eNOS. In addition, statins stimulate other antioxidant enzymes and help control inflammatory processes. Statins thus have effects on various molecules that are central to both oxidative stress and inflammation, notably by stimulating the synthesis of 15-epi-LXA4, which has pro-resolving, anti-inflammatory and antioxidant properties. Their antioxidant effects have been demonstrated in several cardiovascular conditions, including HF, CAD and AF, mainly in the early stages of these diseases (Table 3).

Table 3.

Effects of statins in heart failure with reduced ejection fraction: non-randomized studies (adapted from6–8,69,85,87).

Study  No. of subjects  Statin  Duration
(months) 
Results 
Sola et al. 2006  108  Atorvastatin (20 mg)  12  LVEF and attenuation of adverse LV remodeling 
Vrtovec et al. 2008  110  Atorvastatin (10 mg)  12  ↓ all-cause mortality and ↓ sudden cardiac death 
Wojnicz et al. 2006  74  Atorvastatin (40 mg)  LVEF, ↓ NYHA class and improved quality of life index 
Xie et al. 2010  119  Atorvastatin (10/20 mg)  12  LVEF, ↓ QTc interval 
Yamada et al. 2007  38  Atorvastatin (10 mg)  31  LVEF, ↓ BNP 
Andreou et al.69  60  Rosuvastatin (10 mg)  MPO, oxLDL, NT-proBNP and hsCRP; ↔ fibrinogen, sCD40L and IL6 
Node et al. 2003  51  Simvastatin (5–10 mg)  LVEF, ↓ NYHA class, ↓ BNP 
Gissi et al.85  4574  Rosuvastatin (10 mg)  46  ↔ time to death and ↔ time to death or admission to hospital for cardiovascular reasons 
Kjekshus et al.86  5011  Rosuvastatin (10 mg)  32  ↔ cardiovascular mortality, MI and stroke; ↓ hospitalizations in 15–20% 
Krum et al. 2007  95  Rosuvastatin (10–40 mg)  LVEF 
Hammaad et al. 2005  23  Atorvastatin (40 mg)  ↔ HR variability 
Erbs et al. 2011  42  Rosuvastatin (40 mg)  LVEF, ↑ flow-mediated dilation, ↑ VEGF, ↓ oxLDL 
Bleske et al. 2006  15  Atorvastatin (80 mg)  LVEF, BNP, HR variability 
Bielecka et al. 2009  68  Atorvastatin (10–40 mg)  ↑ functional capacity assessed by 6-min walk test, ↓ NYHA class 
Tousoulis et al. 2005  38  Atorvastatin (20 mg)  ↑ blood flow 
Horwich et al. 2011  26  Atorvastatin (10 mg)  ↔ muscle sympathetic nerve activity, LVEF, BNP and quality of life index 

↓: decreased; ↓: increased; ↔: no change; BNP: B-type natriuretic peptide; MI: myocardial infarction; HR: heart rate; LVEF: left ventricular ejection fraction; hsCRP: high-sensitivity C-reactive protein; IL6: interleukin 6; LV: left ventricular; MPO: myeloperoxidase: NYHA: New York Heart Association; NT-proBNP: N-terminal of B-type natriuretic peptide; oxLDL: oxidized low-density lipoprotein; QTc: corrected QT interval; sCD40L: soluble CD40 ligand; VEGF: vascular endothelial growth factor.

With regard to the effects of statins in reducing cardiovascular events and mortality in HF, although various observational studies have shown reduced mortality with statin therapy, two large clinical trials – CORONA and GISSI-HF – did not demonstrate such a benefit. The ESC and the ACC do not currently recommend statins in HF in the absence of other indications for their use. Given the lack of consensus, further clinical trials are needed to clarify the clinical value of statins in subgroups of HF patients and its relation with their effects on redox state.

Funding

T. Sousa acknowledges support from Fundação para a Ciência e a Tecnologia (FCT) (SFRH/BPD/112005/2015).

Ethical responsibilitiesProtection of people and animals

The authors declare that no experiments were performed on humans or animals for this study.

Data confidentiality

The authors declare that no patient data appear in this article.

Right to privacy and informed consent

The authors declare that no patient data appear in this article.

Conflicts of interest

The authors have no conflicts of interest to declare.

References
[1]
D. Mozaffarian, E.J. Benjamin, A.S. Go, et al.
Heart disease and stroke statistics – 2015 update: a report from the American Heart Association.
Circulation, 131 (2015), pp. e29-e322
[2]
C.W. Yancy, M. Jessup, B. Bozkurt, et al.
2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines.
J Am Coll Cardiol, 62 (2013), pp. e147-e239
[3]
J.J. McMurray, S. Adamopoulos, S.D. Anker, et al.
ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: the Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC.
Eur J Heart Fail, 33 (2012), pp. 1787-1847
[4]
J.M. Hare.
Nitroso-redox balance in the cardiovascular system.
N Engl J Med, 351 (2004), pp. 2112-2114
[5]
Furtado C. Medicamentos do Aparelho Cardiovascular: Uma análise dos padrões de utilização e despesa em Portugal Continental entre 2000 e 2011. Infarmed – Autoridade Nacional do Medicamento e Produtos de Saúde I.P. 2013.
[6]
D. Tousoulis, E. Oikonomou, G. Siasos, et al.
Statins in heart failure—with preserved and reduced ejection fraction. An update.
Pharmacol Ther, 141 (2014), pp. 79-91
[7]
K.O. Bonsu, A. Kadirvelu, D.D. Reidpath.
Statins in heart failure: do we need another trial?.
Vasc Health Risk Manag, 9 (2013), pp. 303-319
[8]
W.H. Tang, G.S. Francis.
Statin treatment for patients with heart failure.
Nat Rev Cardiol, 7 (2010), pp. 249-255
[9]
T.B. Horwich, M.A. Hamilton, W.R. Maclellan, et al.
Low serum total cholesterol is associated with marked increase in mortality in advanced heart failure.
J Card Fail, 8 (2002), pp. 216-224
[10]
J.K. Liao, U. Laufs.
Pleiotropic effects of statins.
Annu Rev Pharmacol Toxicol, 45 (2005), pp. 89-118
[11]
M. Margaritis, K.M. Channon, C. Antoniades.
Statins as regulators of redox state in the vascular endothelium: beyond lipid lowering.
Antioxid Redox Signal, 20 (2014), pp. 1198-1215
[12]
D.P. Jones.
Radical-free biology of oxidative stress.
Am J Physiol Cell Physiol, 295 (2008), pp. C849-C868
[13]
T. Sousa, J. Afonso, F. Carvalho, et al.
Lipid peroxidation and antioxidants in arterial hypertension.
Lipid peroxidation, pp. 345-392
[14]
H. Tsutsui, S. Kinugawa, S. Matsushima.
Oxidative stress and heart failure.
Am J Physiol Heart Circ Physiol, 301 (2011), pp. H2181-H2190
[15]
N. Kaludercic, A. Carpi, R. Menabo, et al.
Monoamine oxidases (MAO) in the pathogenesis of heart failure and ischemia/reperfusion injury.
Biochim Biophys Acta, 1813 (2011), pp. 1323-1332
[16]
W.H. Tang, M.L. Brennan, K. Philip, et al.
Plasma myeloperoxidase levels in patients with chronic heart failure.
Am J Cardiol, 98 (2006), pp. 796-799
[17]
D.I. Brown, K.K. Griendling.
Nox proteins in signal transduction.
Free Radic Biol Med, 47 (2009), pp. 1239-1253
[18]
J. Kuroda, J. Sadoshima.
NADPH oxidase and cardiac failure.
J Cardiovasc Transl Res, 3 (2010), pp. 314-320
[19]
Y. Octavia, H.P. Brunner-La Rocca, A.L. Moens.
NADPH oxidase-dependent oxidative stress in the failing heart: from pathogenic roles to therapeutic approach.
Free Radic Biol Med, 52 (2012), pp. 291-297
[20]
J.M. Kwok, C.C. Ma, S. Ma.
Recent development in the effects of statins on cardiovascular disease through Rac1 and NADPH oxidase.
Vascul Pharmacol, 58 (2013), pp. 21-30
[21]
A. Schramm, P. Matusik, G. Osmenda, et al.
Targeting NADPH oxidases in vascular pharmacology.
Vascul Pharmacol, 56 (2012), pp. 216-231
[22]
E. Yamamoto, K. Kataoka, H. Shintaku, et al.
Novel mechanism and role of angiotensin II induced vascular endothelial injury in hypertensive diastolic heart failure.
Arterioscler Thromb Vasc Biol, 27 (2007), pp. 2569-2575
[23]
M.E. Manni, M. Zazzeri, C. Musilli, et al.
Exposure of cardiomyocytes to angiotensin II induces over-activation of monoamine oxidase type A: implications in heart failure.
Eur J Pharmacol, 718 (2013), pp. 271-276
[24]
C. Villeneuve, C. Guilbeau-Frugier, P. Sicard, et al.
p53-PGC-1alpha pathway mediates oxidative mitochondrial damage and cardiomyocyte necrosis induced by monoamine oxidase-A upregulation: role in chronic left ventricular dysfunction in mice.
Antioxid Redox Signal, 18 (2013), pp. 5-18
[25]
N. Kaludercic, A. Carpi, T. Nagayama, et al.
Monoamine oxidase B prompts mitochondrial and cardiac dysfunction in pressure overloaded hearts.
Antioxid Redox Signal, 20 (2014), pp. 267-280
[26]
M. Reina-Couto, J. Carvalho, M.J. Valente, et al.
Impaired resolution of inflammation in human chronic heart failure.
Eur J Clin Invest, 44 (2014), pp. 527-538
[27]
J. Haendeler, J. Hoffmann, A.M. Zeiher, et al.
Antioxidant effects of statins via S-nitrosylation and activation of thioredoxin in endothelial cells: a novel vasculoprotective function of statins.
Circulation, 110 (2004), pp. 856-861
[28]
N.G. Abraham, A. Kappas.
Heme oxygenase and the cardiovascular-renal system.
Free Radic Biol Med, 39 (2005), pp. 1-25
[29]
E.A. Podrez.
Anti-oxidant properties of high-density lipoprotein and atherosclerosis.
Clin Exp Pharmacol Physiol, 37 (2010), pp. 719-725
[30]
D.B. Sawyer, D.A. Siwik, L. Xiao, et al.
Role of oxidative stress in myocardial hypertrophy and failure.
J Mol Cell Cardiol, 34 (2002), pp. 379-388
[31]
S. Dieterich, U. Bieligk, K. Beulich, et al.
Gene expression of antioxidative enzymes in the human heart: increased expression of catalase in the end-stage failing heart.
Circulation, 101 (2000), pp. 33-39
[32]
A.T. Baumer, M. Flesch, X. Wang, et al.
Antioxidative enzymes in human hearts with idiopathic dilated cardiomyopathy.
J Mol Cell Cardiol, 32 (2000), pp. 121-130
[33]
F. Sam, D.L. Kerstetter, D.R. Pimental, et al.
Increased reactive oxygen species production and functional alterations in antioxidant enzymes in human failing myocardium.
J Card Fail, 11 (2005), pp. 473-480
[34]
M. De Lorgeril, P. Salen, M. Accominotti, et al.
Dietary and blood antioxidants in patients with chronic heart failure. Insights into the potential importance of selenium in heart failure.
Eur J Heart Fail, 3 (2001), pp. 661-669
[35]
F.J. Giordano.
Oxygen, oxidative stress, hypoxia, and heart failure.
J Clin Invest, 115 (2005), pp. 500-508
[36]
P. Pacher, J.S. Beckman, L. Liaudet.
Nitric oxide and peroxynitrite in health and disease.
Physiol Rev, 87 (2007), pp. 315-424
[37]
K.M. Naseem.
The role of nitric oxide in cardiovascular diseases.
Mol Aspects Med, 26 (2005), pp. 33-65
[38]
R. Rastaldo, P. Pagliaro, S. Cappello, et al.
Nitric oxide and cardiac function.
Life Sci, 81 (2007), pp. 779-793
[39]
C. Aoki, A. Nakano, S. Tanaka, et al.
Fluvastatin upregulates endothelial nitric oxide synthase activity via enhancement of its phosphorylation and expression and via an increase in tetrahydrobiopterin in vascular endothelial cells.
Int J Cardiol, 156 (2012), pp. 55-61
[40]
Y. Hattori, N. Nakanishi, K. Akimoto, et al.
HMG-CoA reductase inhibitor increases GTP cyclohydrolase I mRNA and tetrahydrobiopterin in vascular endothelial cells.
Arterioscler Thromb Vasc Biol, 23 (2003), pp. 176-182
[41]
H. Ota, M. Eto, M.R. Kano, et al.
Induction of endothelial nitric oxide synthase, SIRT1, and catalase by statins inhibits endothelial senescence through the Akt pathway.
Arterioscler Thromb Vasc Biol, 30 (2010), pp. 2205-2211
[42]
I. Kosmidou, J.P. Moore, M. Weber, et al.
Statin treatment and 3′ polyadenylation of eNOS mRNA.
Arterioscler Thromb Vasc Biol, 27 (2007), pp. 2642-2649
[43]
J. Wang, Z. Xu, I. Kitajima, et al.
Effects of different statins on endothelial nitric oxide synthase and AKT phosphorylation in endothelial cells.
Int J Cardiol, 127 (2008), pp. 33-39
[44]
W. Sun, T.S. Lee, M. Zhu, et al.
Statins activate AMP-activated protein kinase in vitro and in vivo.
Circulation, 114 (2006), pp. 2655-2662
[45]
A. Brouet, P. Sonveaux, C. Dessy, et al.
Hsp90 and caveolin are key targets for the proangiogenic nitric oxide-mediated effects of statins.
Circ Res, 89 (2001), pp. 866-873
[46]
C. Mineo, P.W. Shaul.
Regulation of eNOS in caveolae.
Adv Exp Med Biol, 729 (2012), pp. 51-62
[47]
R. Carnevale, P. Pignatelli, S. Di Santo, et al.
Atorvastatin inhibits oxidative stress via adiponectin-mediated NADPH oxidase down-regulation in hypercholesterolemic patients.
Atherosclerosis, 213 (2010), pp. 225-234
[48]
A.K. Kanugula, P.N. Gollavilli, S.B. Vasamsetti, et al.
Statin-induced inhibition of breast cancer proliferation and invasion involves attenuation of iron transport: intermediacy of nitric oxide and antioxidant defence mechanisms.
FEBS J, (2014),
[49]
N. Schupp, U. Schmid, A. Heidland, et al.
Rosuvastatin protects against oxidative stress and DNA damage in vitro via upregulation of glutathione synthesis.
Atherosclerosis, 199 (2008), pp. 278-287
[50]
N. Grosser, K. Erdmann, A. Hemmerle, et al.
Rosuvastatin upregulates the antioxidant defense protein heme oxygenase-1.
Biochem Biophys Res Commun, 325 (2004), pp. 871-876
[51]
A. Nagila, T. Permpongpaiboon, S. Tantrarongroj, et al.
Effect of atorvastatin on paraoxonase1 (PON1) and oxidative status.
Pharmacol Rep, 61 (2009), pp. 892-898
[52]
N. Khaper, S. Bryan, S. Dhingra, et al.
Targeting the vicious inflammation-oxidative stress cycle for the management of heart failure.
Antioxid Redox Signal, 13 (2010), pp. 1033-1049
[53]
N.D. Vaziri.
Causal link between oxidative stress, inflammation, and hypertension.
Iran J Kidney Dis, 2 (2008), pp. 1-10
[54]
R. Zenz, R. Eferl, C. Scheinecker, et al.
Activator protein 1 (Fos/Jun) functions in inflammatory bone and skin disease.
Arthritis Res Ther, 10 (2008), pp. 201
[55]
S. Corda, C. Laplace, E. Vicaut, et al.
Rapid reactive oxygen species production by mitochondria in endothelial cells exposed to tumor necrosis factor-alpha is mediated by ceramide.
Am J Respir Cell Mol Biol, 24 (2001), pp. 762-768
[56]
J. Kaur, G.S. Dhaunsi, R.B. Turner.
Interleukin-1 and nitric oxide increase NADPH oxidase activity in human coronary artery smooth muscle cells.
Med Princ Pract, 13 (2004), pp. 26-29
[57]
N. Anatoliotakis, S. Deftereos, G. Bouras, et al.
Myeloperoxidase: expressing inflammation and oxidative stress in cardiovascular disease.
Curr Top Med Chem, 13 (2013), pp. 115-138
[58]
A.P. Kumar, W.F. Reynolds.
Statins downregulate myeloperoxidase gene expression in macrophages.
Biochem Biophys Res Commun, 331 (2005), pp. 442-451
[59]
J. Madrigal-Matute, J.S. Lindholt, C.E. Fernandez-Garcia, et al.
Galectin-3, a biomarker linking oxidative stress and inflammation with the clinical outcomes of patients with atherothrombosis.
[60]
Y.J. Lee, Y.S. Koh, H.E. Park, et al.
Spatial and temporal expression, and statin responsiveness of galectin-1 and galectin-3 in murine atherosclerosis.
Korean Circ J, 43 (2013), pp. 223-230
[61]
M. Spite, C.N. Serhan.
Novel lipid mediators promote resolution of acute inflammation: impact of aspirin and statins.
Circ Res, 107 (2010), pp. 1170-1184
[62]
Y. Birnbaum, Y. Ye, Y. Lin, et al.
Augmentation of myocardial production of 15-epi-lipoxin-a4 by pioglitazone and atorvastatin in the rat.
Circulation, 114 (2006), pp. 929-935
[63]
A. Planaguma, M.A. Pfeffer, G. Rubin, et al.
Lovastatin decreases acute mucosal inflammation via 15-epi-lipoxin A4.
Mucosal Immunol, 3 (2010), pp. 270-279
[64]
L. Jozsef, C. Zouki, N.A. Petasis, et al.
Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes.
Proc Natl Acad Sci U S A, 99 (2002), pp. 13266-13271
[65]
T. Carlo, H. Kalwa, B.D. Levy.
15-Epi-lipoxin A4 inhibits human neutrophil superoxide anion generation by regulating polyisoprenyl diphosphate phosphatase 1.
FASEB J, 27 (2013), pp. 2733-2741
[66]
V. Nascimento-Silva, M.A. Arruda, C. Barja-Fidalgo, et al.
Aspirin-triggered lipoxin A4 blocks reactive oxygen species generation in endothelial cells: a novel antioxidative mechanism.
Thromb Haemost, 97 (2007), pp. 88-98
[67]
S.H. Deo, J.P. Fisher, L.C. Vianna, et al.
Statin therapy lowers muscle sympathetic nerve activity and oxidative stress in patients with heart failure.
Am J Physiol Heart Circ Physiol, 303 (2012), pp. H377-H385
[68]
D. Greig, H. Alcaino, P.F. Castro, et al.
Xanthine-oxidase inhibitors and statins in chronic heart failure: effects on vascular and functional parameters.
J Heart Lung Transplant, 30 (2011), pp. 408-413
[69]
I. Andreou, D. Tousoulis, A. Miliou, et al.
Effects of rosuvastatin on myeloperoxidase levels in patients with chronic heart failure: a randomized placebo-controlled study.
Atherosclerosis, 210 (2010), pp. 194-198
[70]
C. Maack, T. Kartes, H. Kilter, et al.
Oxygen free radical release in human failing myocardium is associated with increased activity of rac1-GTPase and represents a target for statin treatment.
Circulation, 108 (2003), pp. 1567-1574
[71]
C. Antoniades, M. Demosthenous, S. Reilly, et al.
Myocardial redox state predicts in-hospital clinical outcome after cardiac surgery effects of short-term pre-operative statin treatment.
J Am Coll Cardiol, 59 (2012), pp. 60-70
[72]
C. Antoniades, C. Bakogiannis, P. Leeson, et al.
Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase coupling.
Circulation, 124 (2011), pp. 335-345
[73]
G. Ndrepepa, S. Braun, A. Schomig, et al.
Impact of therapy with statins, beta-blockers and angiotensin-converting enzyme inhibitors on plasma myeloperoxidase in patients with coronary artery disease.
Clin Res Cardiol, 100 (2011), pp. 327-333
[74]
P. Pignatelli, R. Carnevale, D. Pastori, et al.
Immediate antioxidant and antiplatelet effect of atorvastatin via inhibition of Nox2.
Circulation, 126 (2012), pp. 92-103
[75]
P. Pignatelli, R. Carnevale, S. Di Santo, et al.
Rosuvastatin reduces platelet recruitment by inhibiting NADPH oxidase activation.
Biochem Pharmacol, 84 (2012), pp. 1635-1642
[76]
R. Cangemi, L. Loffredo, R. Carnevale, et al.
Early decrease of oxidative stress by atorvastatin in hypercholesterolaemic patients: effect on circulating vitamin E.
Eur Heart J, 29 (2008), pp. 54-62
[77]
M.J. Shin, E.Y. Cho, Y. Jang, et al.
A beneficial effect of simvastatin on DNA damage in 242T allele of the NADPH oxidase p22phox in hypercholesterolemic patients.
Clin Chim Acta, 360 (2005), pp. 46-51
[78]
Z. Wu, Y. Lou, W. Jin, et al.
Relationship of the p22phox (CYBA) gene polymorphism C242T with risk of coronary artery disease: a meta-analysis.
[79]
S.N. Reilly, R. Jayaram, K. Nahar, et al.
Atrial sources of reactive oxygen species vary with the duration and substrate of atrial fibrillation: implications for the antiarrhythmic effect of statins.
Circulation, 124 (2011), pp. 1107-1117
[80]
M. Abe, N. Maruyama, K. Okada, et al.
Effects of lipid-lowering therapy with rosuvastatin on kidney function and oxidative stress in patients with diabetic nephropathy.
J Atheroscler Thromb, 18 (2011), pp. 1018-1028
[81]
J. Hernandez-Ojeda, L.M. Roman-Pintos, A.D. Rodriguez-Carrizalez, et al.
Effect of rosuvastatin on diabetic polyneuropathy: a randomized, double-blind, placebo-controlled Phase IIa study.
Diabetes Metab Syndr Obes, 7 (2014), pp. 401-407
[82]
P.G. Scheffer, R.K. Schindhelm, V.M. van Verschuer, et al.
No effect of atorvastatin and simvastatin on oxidative stress in patients at high risk for cardiovascular disease.
Neth J Med, 71 (2013), pp. 359-365
[83]
F.H. Mose, T. Larsen, J.M. Jensen, et al.
Effect of atorvastatin on renal NO availability and tubular function in patients with stage II-III chronic kidney disease and type 2 diabetes.
Scand J Clin Lab Invest, 74 (2014), pp. 8-19
[84]
F. Tehrani, R. Morrissey, A. Phan, et al.
Statin therapy in patients with diastolic heart failure.
Clin Cardiol, 33 (2010), pp. E1-E5
[85]
H.F.I. Gissi, L. Tavazzi, A.P. Maggioni, et al.
Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial.
Lancet, 372 (2008), pp. 1231-1239
[86]
J. Kjekshus, P. Dunselman, M. Blideskog, et al.
A statin in the treatment of heart failure? Controlled rosuvastatin multinational study in heart failure (CORONA): study design and baseline characteristics.
Eur J Heart Fail, 7 (2005), pp. 1059-1069
[87]
J. Kjekshus, E. Apetrei, V. Barrios, et al.
Rosuvastatin in older patients with systolic heart failure.
N Engl J Med, 357 (2007), pp. 2248-2261
[88]
M.J. Lipinski, C.A. Cauthen, G.G. Biondi-Zoccai, et al.
Meta-analysis of randomized controlled trials of statins versus placebo in patients with heart failure.
Am J Cardiol, 104 (2009), pp. 1708-1716
[89]
H. Takagi, T. Umemoto.
Atorvastatin, not rosuvastatin, improves cardiac function in heart failure: a meta-analysis of randomized trials.
Int J Cardiol, 155 (2012), pp. 296-299
[90]
P. Gastelurrutia, J. Lupon, M. de Antonio, et al.
Statins in heart failure: the paradox between large randomized clinical trials and real life.
Mayo Clin Proc, 87 (2012), pp. 555-560
[91]
V.G. Athyros, A. Karagiannis, D.P. Mikhailidis.
Statins and heart failure.
J Am Coll Cardiol, 55 (2010), pp. 1644-1645
author reply 6
[92]
J.G. Cleland, J.J. McMurray, J. Kjekshus, et al.
Plasma concentration of amino-terminal pro-brain natriuretic peptide in chronic heart failure: prediction of cardiovascular events and interaction with the effects of rosuvastatin: a report from CORONA (Controlled Rosuvastatin Multinational Trial in Heart Failure).
J Am Coll Cardiol, 54 (2009), pp. 1850-1859
[93]
L. Gullestad, T. Ueland, J. Kjekshus, et al.
Galectin-3 predicts response to statin therapy in the Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA).
Eur Heart J, 33 (2012), pp. 2290-2296
[94]
Y. Shitara, Y. Sugiyama.
Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions.
Pharmacol Ther, 112 (2006), pp. 71-105
[95]
A.S. Antonopoulos, M. Margaritis, C. Shirodaria, et al.
Translating the effects of statins: from redox regulation to suppression of vascular wall inflammation.
Thromb Haemost, 108 (2012), pp. 840-848
[96]
J.K. Liao.
Isoprenoids as mediators of the biological effects of statins.
J Clin Invest, 110 (2002), pp. 285-288
[97]
A.C. Pinho-Gomes, S. Reilly, R.P. Brandes, et al.
Targeting inflammation and oxidative stress in atrial fibrillation: role of 3-hydroxy-3-methylglutaryl-coenzyme a reductase inhibition with statins.
Antioxid Redox Signal, 20 (2014), pp. 1268-1285
[98]
X. Xu, L. Zhang, J. Liang.
Rosuvastatin prevents pressure overload-induced myocardial hypertrophy via inactivation of the Akt, ERK1/2 and GATA4 signaling pathways in rats.
Mol Med Rep, 8 (2013), pp. 385-392
[99]
C. Ulivieri, C.T. Baldari.
Statins: from cholesterol-lowering drugs to novel immunomodulators for the treatment of Th17-mediated autoimmune diseases.
Pharmacol Res, (2014),
[100]
T.B. Horwich, H.R. Middlekauff.
Potential autonomic nervous system effects of statins in heart failure.
Heart Fail Clin, 4 (2008), pp. 163-170

These authors contributed equally to this work.

Please cite this article as: Costa S, Reina-Couto M, Albino-Teixeira A, Sousa T. Estatinas e stresse oxidativo na insuficiência cardíaca crónica. Rev Port Cardiol. 2016;35:41–57.

Copyright © 2015. Sociedade Portuguesa de Cardiologia
Idiomas
Revista Portuguesa de Cardiologia (English edition)
Article options
Tools
en pt

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

By checking that you are a health professional, you are stating that you are aware and accept that the Portuguese Journal of Cardiology (RPC) is the Data Controller that processes the personal information of users of its website, with its registered office at Campo Grande, n.º 28, 13.º, 1700-093 Lisbon, telephone 217 970 685 and 217 817 630, fax 217 931 095, and email revista@spc.pt. I declare for all purposes that the information provided herein is accurate and correct.