You are on page 1of 14

Eur J Clin Pharmacol (2003) 58: 719–731

DOI 10.1007/s00228-002-0556-0

R EV IE W A RT I C L E

Ulrich Laufs

Beyond lipid-lowering: effects of statins on endothelial nitric oxide

Received: 2 September 2002 / Accepted: 15 December 2002 / Published online: 18 February 2003
Ó Springer-Verlag 2003

Abstract Endothelial dysfunction is now recognised as Keywords Atherosclerosis Æ Endothelial dysfunction Æ


an important process in the pathogenesis of atheroscle- Nitric oxide
rosis. Nitric oxide (NO) release by the endothelium
regulates blood flow, inflammation and platelet aggre-
gation, and consequently its disruption during endo-
thelial dysfunction can decrease plaque stability and Introduction
encourage the formation of atherosclerotic lesions and
thrombi. Inhibitors of 3-hydroxy-3-methylglutaryl Atherosclerosis is the underlying cause of many car-
coenzyme A reductase (statins) are often utilised in the diovascular diseases, such as stroke, myocardial infarc-
prevention of coronary heart disease due to their efficacy tion and peripheral artery disease. There is increasing
at lowering lipid levels. However, statins may also pre- support for the premise that atherosclerosis is the result
vent atherosclerotic disease by non-lipid or pleiotropic of a chronic inflammation perpetuated by a dysfunc-
effects, for example, improving endothelial function by tional endothelium [1]. Atherosclerotic lesions develop
promoting the production of NO. There are various as a result of monocyte migration and subsequent foam-
mechanisms whereby statins may alter NO release, such cell formation, combined with vascular smooth-muscle
as inhibiting the production of mevalonate and impor- cell proliferation. As the endothelium regulates vascular
tant isoprenoid intermediates, thereby preventing the tone, endothelial permeability, platelet aggregation, and
isoprenylation of the small GTPase Rho, which nega- leucocyte activation and extravasation, it has been
tively regulates the expression of endothelial nitric oxide implicated in the development of atherosclerosis [2].
synthase (eNOS). Furthermore, statins may also in- Indeed, endothelial dysfunction can promote athero-
crease eNOS activity via post-translational activation of sclerosis by affecting production of vasoactive media-
the phosphatidylinositol 3-kinase/protein kinase Akt tors, such as nitric oxide (NO) and prostacyclin (PGI2),
(PI3 K/Akt) pathway and/or through an interaction which regulate these processes. The most important
with the molecular chaperone heat-shock protein 90 and well-known endothelium-derived mediator is NO.
(HSP90). Data suggest that statins may vary in their Disrupted production and release of endothelium-
efficacy for enhancing the release of NO, and the derived NO is a major consequence of endothelial dys-
mechanisms dictating these differences are not yet clear. function [3].
By increasing NO production, statins may interfere with The aim of this review was to examine the role of NO
atherosclerotic lesion development, stabilise plaque, in- in the development of atherosclerosis and how statins
hibit platelet aggregation, improve blood flow and pro- may improve endothelial function.
tect against ischaemia. Therefore, the ability of statins to
improve endothelial function through the release of NO
may partially account for their beneficial effects at re- Endothelial dysfunction and the role
ducing the incidence of cardiovascular events. of NO in atherosclerosis

Endothelial dysfunction occurs early in the development


U. Laufs of atherosclerosis as a result of exposure to cardiovas-
Medizinische Klinik und Poliklinik der Universität des Saarlandes, cular risk factors, such as age, smoking, diabetes, hy-
Innere Medizin III, 66421, Homburg/Saar, Germany
E-mail: ulrich@laufs.com
pertension or dyslipidaemia [1, 2]. A dysfunctional
Tel.: +49-6841-1623000 endothelium predisposes blood vessels to processes
Fax: +49-6841-1623437 favouring the development of atherosclerosis (Fig. 1).
720

L-citrulline by endothelial nitric oxide synthase (eNOS).


Production and release of NO by endothelial cells occurs
at both a basal level and in response to stimuli by
vasoactive mediators such as endothelin-1, bradykinin
and acetylcholine. In addition, flow of blood over en-
dothelial cells (shear stress) can also stimulate release of
NO [5]. Indeed, it is notable that atherosclerotic lesions
tend to develop at vascular bifurcations, where turbu-
lence disturbs the flow of blood over the endothelium.
Several anti-atherosclerotic processes are mediated by
NO (Fig. 2). NO can inhibit vascular smooth muscle cell
proliferation [6], induce vasodilation [7] and increase
blood flow [8], promote angiogenesis, increase endothe-
lial cell survival [9], reduce platelet aggregation [10],
inhibit inflammation [11] and regulate leucocyte–endo-
thelium interactions [12, 13]. For example, NO can
reduce adhesion of monocytes to human endothelial
cells (Fig. 2) by counteracting the induction of vascular
cell adhesion molecule-1 (VCAM-1) by cytokines [14]
and by downregulating the chemokine monocyte
chemoattractant protein-1 (MCP-1) at a transcriptional
level [15]. Considering the pivotal role played by NO in
regulating atherosclerotic processes, it is of little surprise
that an imbalance in production has been shown to
promote the development of coronary artery disease in
individuals with multiple risk factors [16]. Mice lacking
the gene for eNOS show increased blood pressure,
impaired vascular response to injury and suffer from
larger cerebral infarcts [17, 18, 19]. Furthermore, it has
Fig. 1 Scheme through which cardiovascular risk factors may been reported that chronic inhibition of eNOS induces
promote atherosclerosis via endothelial dysfunction. LDL low- early vascular inflammation and development of ath-
density lipoprotein erosclerosis in rats [20].
Improving endothelial dysfunction and NO release
For example, macrophage accumulation, a key process may represent a new treatment paradigm for the pre-
in atherosclerotic lesion development, may occur when a vention of coronary heart disease (CHD). The 3-hy-
dysfunctional endothelium: (1) disrupts anti-inflamma- droxy-3-methylglutaryl coenzyme A (HMG-CoA)
tory processes, leading to increased monocyte activa- reductase inhibitors (statins), a class of lipid-modifying
tion, adhesion and migration; (2) increases endothelial agents used in the treatment and prevention of athero-
permeability thereby increasing access of monocytes and sclerosis, have been shown to have beneficial effects on
lipoproteins to the vessel wall; and/or (3) reduces these processes.
availability of vasodilators, thereby reducing blood flow
and aiding monocyte adhesion and migration.
Following monocyte migration there are several steps Use of statins for the treatment of atherosclerosis
in the development of an atherosclerotic lesion (Fig. 1).
Once within the vessel wall, macrophages can take up There is a strong association between elevated plasma
and store oxidised low-density lipoprotein (oxLDL), cholesterol levels and atherosclerosis [21, 22]. Conse-
forming a foam cell and creating the characteristic ‘‘fatty quently, physicians recognise the importance of lowering
streak’’ of an early atherosclerotic lesion [118]. Release total cholesterol and LDL cholesterol levels to prevent
of growth factors from these inflammatory cells can CHD. The cholesterol-modifying agents of choice for
cause proliferation of vascular smooth muscle cells prevention of CHD are the statins [23, 24].
within the intima, further increasing lesion size and HMG-CoA reductase catalyses the rate-limiting step
decreasing lumen diameter. Finally, release of matrix of cholesterol biosynthesis, a four-electron reductive
metalloproteases from leucocytes can break down col- deacylation of HMG-CoA to CoA and mevalonate.
lagen and cause atherosclerotic plaque to destabilise and Statins are HMG-CoA reductase inhibitors with inhi-
rupture, leading to thrombus formation and triggering bition constant values in the nanomolar range. All sta-
an acute cardiovascular event [4]. tins share a HMG-like moiety and inhibit the reductase
One consequence of endothelial dysfunction is by the same mechanism. Recently, the structure of the
reduced production of the vasoactive mediator NO, catalytic portion of human HMG-CoA reductase com-
which is created during the conversion of L-arginine to plexed with different statins was determined [25]. The
721

Fig. 2 Diagram indicating the


influence of oxidised low-
density lipoprotein (LDL) on
nitric oxide (NO) production
and its effect on various
atherosclerotic processes.
Dotted lines indicate an
inhibitory effect. eNOS
endothelial nitric oxide
synthase, L-ArgL-arginine,
L-Cit L-citrulline, MCP-1
monocyte chemoattractant
protein-1, ICAM-1 intercellular
adhesion molecule-1, VCAM-1
vascular cell adhesion molecule-
1, oxLDL oxidised low-density
lipoprotein

bulky, hydrophobic compounds of statins occupy the differences account for the diverse inhibition constant
HMG-binding pocket and block access of the substrate values. The currently available statins (atorvastatin,
HMG. The tight binding of statins is due to the large fluvastatin, lovastatin, pravastatin and simvastatin) can
number of van der Waals interactions between inhibitors reduce LDL cholesterol levels by 24–60% (Table 1) [26].
and HMG-CoA reductase. The structurally diverse, New synthetic statins, such as rosuvastatin, are more
rigid, hydrophobic groups of the different statins are effective at reducing LDL cholesterol (Table 1) [27, 116,
accommodated in a shallow non-polar groove that is 117, 119], possibly due to their improved binding to
present only when COOH-terminal residues of HMG- HMG-CoA reductase. The effectiveness of statins at
CoA reductase are disordered. There are subtle differ- reducing cholesterol levels and cardiovascular events has
ences in the modes of binding between the various been reported in large outcome-based clinical trials [28,
statins, with the synthetic compounds atorvastatin and 29, 30, 31, 32, 33].
rosuvastatin having the greatest number of bonding More recently, additional benefits of statins have
interactions with HMG-CoA reductase [25]. These been identified that may be independent of their

Table 1 Comparison of lipophilicity and the selectivity of choles- not available, IC50 inhibition of cholesterol synthesis by 3-hydroxy
terol synthesis for various statins in hepatocytes and endothelial 3-methylglutaryl coenzyme A reductase, HUVEC human umbilical
cells. LDL-C low-density lipoprotein cholesterol, NA information vein endothelial cells, NO nitric oxide

Statin Maximum Reduction Lipophilicityc Plasma Purified IC50 (nM) HUVECe NO releasef
,c
dose in LDL-C at half-life enzyme *
maximum dose
(mg) (%) Log D at (h) Rat hepatocytese (% of A23187-
pH 7.4 stimulated release)**

Atorvastatin 80 55a 1.11 14d 8.2 0.8 5.5 27


Cerivastatin 0.4 34a 1.69 23d 10 2.5 3.1 NA
Fluvastatin 80 34a 1.27 1.2d 27.6 4.8 0.6 NA
Lovastatin 80 41a NA 2d NA NA NA 23
Pravastatin 40 34a 0.84 12d 44.1 5.0 1900 28
Rosuvastatin 40 63b 0.33 20c 5.4 0.3 41 NA
Simvastatin 80 48a 1.6 12d 11.2 5.2 1.0 56
 a
Cerivastatin is no longer available Maron et al. 2000 [26]
b
*Human HMG-CoA reductase catalytic domain Olsson et al. 2001 [27]
c
**NO release is the peak NO concentration after stimulation of McTaggart et al. 2001 [28] and Hanefield 2001 [29]
d
bovine endothelial cells with of 1 lmol/l of statin compared as a Knopp 1999 [30]
e
percentage decrease from the peak concentration after stimulation Buckett et al. 2000 [31]
f
with calcium ionophore, A23187 (1 lmol/l) Dobrucki et al. 2001 [32]
722

cholesterol-lowering effects. Pleiotropic effects, i.e. ac- an important precursor in the biosynthesis of cholesterol
tions not dependent on cholesterol lowering, have been (Fig. 3) and its production is inhibited by statins.
observed in several studies. For example, in the West of However, mevalonate is a substrate for compounds other
Scotland Coronary Prevention Study (WOSCOPS), the than cholesterol [54]. This may indicate a potential
Scandinavian Simvastatin Survival Study (4S) and the mechanism for the cholesterol-independent effects
recent Heart Protection Study (HPS), it was noted that observed with statins. It is now apparent that the
clinical benefit associated with statin therapy was inde- mechanisms whereby statins increase NO production can
pendent of LDL cholesterol levels [33, 34, 35]. Fur- be both cholesterol dependent and cholesterol indepen-
thermore, although LDL cholesterol is not considered a dent (Table 2, Fig. 3).
major risk factor for stroke [36], statin treatment can
reduce the risk of cerebrovascular events [33, 37, 38].
However, the clinical importance of non-cholesterol or Cholesterol-dependent regulation of NO by statins
‘‘pleiotropic’’ effects of statins in humans in relation to
cardiovascular mortality is difficult to determine in a Regulation of NO production and endothelial function
prospective clinical trial because statins effectively re- may be dependent on LDL cholesterol concentration
duce cholesterol levels even in individuals with low and oxidation status [120]. LDL cholesterol apheresis
baseline cholesterol levels. In cynomolgus monkeys, has been shown to rapidly improve endothelial-depen-
cholesterol-independent effects of statins were demon- dent vasodilation [55] and coronary blood flow [56] in
strated using a ‘‘clamped-cholesterol’’ design, where hypercholesterolaemic patients. While this could argu-
dietary cholesterol was regulated to maintain equal ably be a result of decreased blood viscosity and a cor-
changes in lipoprotein levels in control and pravastatin- responding improvement in shear stress-mediated NO
treated animals [39]. In addition, a similar study has release, in vitro experiments indicate other possible
shown a cholesterol-independent reduction in the mechanisms. In cultured human endothelial cells, eNOS
development of atherosclerosis with rosuvastatin in mRNA and protein was downregulated by oxidised
APOE*-Leiden transgenic mice [40]. One explanation LDL cholesterol [57] as well as by atherogenic concen-
for the pleiotropic effects of statins is their ability to trations of human native LDL [58]. Therefore, by re-
influence endothelial function, and NO production in ducing LDL cholesterol levels, statins may prevent this
particular. downregulation of eNOS. Indeed, such an effect has
been reported with simvastatin treatment in human en-
dothelial cells in culture [53].
Effects of statins on NO synthesis Oxidation status of LDL also appears to be an im-
portant factor in the regulation of eNOS. It has been
Increased release of NO from bovine endothelial cells in reported that expression of eNOS mRNA in human and
culture has been demonstrated with lovastatin, ator- bovine endothelial cells was decreased by oxLDL in a
vastatin, pravastatin, simvastatin and rosuvastatin [32, concentration-dependent manner [57, 59]. Simvastatin,
41]. Enhanced NO release by statins appears to be as- atorvastatin and lovastatin treatment attenuated this
sociated with dose-dependent increases in eNOS in hu- reduction in eNOS expression in the presence of oxLDL
man endothelial cells in culture [42, 43]. This effect of [59, 60]. Therefore, the reduction of LDL and hence
statins on eNOS expression has been observed in several oxLDL, may be a mechanism for statins to regulate
species, including human cells in culture [42, 43, 44] and eNOS in vivo. However, in cell-culture experiments,
in mice [45], rats [46], rabbits [47], dogs [48] and primates statins do not decrease the concentration of the exter-
in vivo [39]. The NO synthase inhibitor, L-nitro arginine nally added cholesterol, indicating that additional path-
methyl ester (L-NAME), inhibited the increased gener- ways are also likely to contribute to eNOS upregulation.
ation of NO with rosuvastatin [49], pravastatin and Oxidised LDL appears to regulate eNOS through its
simvastatin [13, 50] in rats and rat tissues in vitro, con- action on caveolin-1, a primary coat protein of cellular
firming an effect on synthesis by eNOS. Both basal and invaginations called caveolae that are necessary for the
stimulated NO release is increased with statin treatment. assembly and trafficking of cholesterol [61]. Caveolin-1
For example, basal and stimulated release of NO was has been shown to bind to eNOS in caveolae and act as a
increased by 45% and 107%, respectively, from cultured negative regulator of the enzyme [61, 62]. Exposure of
bovine aortic endothelial cells treated with atorvastatin bovine endothelial cells to hypercholesterolaemic serum
[51]. These studies show that statins increase the bio- has been shown to upregulate caveolin-1 abundance and
availability of NO. promotes association of caveolin-1 and eNOS into in-
The mechanisms whereby statins improve NO pro- hibitory complexes, thereby decreasing NO production
duction are being elucidated. It has been suggested that [63]. From these data, it is possible to reason that statins
this beneficial effect of statins may be a result of reduced may improve eNOS activity by reducing cholesterol and,
formation or availability of mevalonate within endothe- consequently, abundance and expression of caveolin-1.
lial cells [52]. Indeed, statin-induced increases in eNOS Indeed, statins have been shown to reduce caveolin-1
expression are reversed by addition of mevalonate to abundance in cultured bovine endothelial cells [51] and
human endothelial cells in culture [42, 53]. Mevalonate is mice in vivo [64] and thereby decrease its inhibitory
723

Fig. 3 Mevalonate-dependent
inhibition of endothelial nitric
oxide synthase (eNOS) may
occur through cholesterol-
dependent and/or independent
mechanisms. Acetyl-CoA acetyl
coenzyme A, HMG-CoA
3-hydroxy-3-methylglutaryl
coenzyme A, NO nitric oxide,
oxLDL oxidised low density
lipoprotein, PP pyrophosphate

action on both basal and agonist-stimulated eNOS ac-


tivity [51]. Feron and colleagues have reported that basal Cholesterol-independent regulation of NO by statins
NO release was potentiated by statin treatment more
when cholesterol levels were low, whereas stimulated In addition to their effects on NO through cholesterol
release was greater when cholesterol levels were high synthesis, statins can upregulate eNOS expression in-
[51]. This varying ability of statin treatment to influence dependent of plasma cholesterol levels (Fig. 3) [67]. In-
caveolin-1 and eNOS with cholesterol level may be hibition of mevalonate synthesis by statins prevents the
suggestive of a combination of cholesterol-dependent synthesis of important isoprenoid intermediates such as
and independent effects. Thus, statins may regulate NO geranylgeranylpyrophosphate (GGPP). Rho GTPases,
production through lowering cholesterol and decreasing members of the Ras superfamily of GTP-binding pro-
abundance of caveolin-1. teins, consist of at least 14 proteins that can be separated
Receptors for LDL also mediate cholesterol-depen- into Rho, Rac and Cdc42 [68]. Rho proteins cycle be-
dent regulation of eNOS. For example, the oxLDL tween an active GTP-bound state and an inactive GDP-
class-B scavenger receptor, CD36, has been shown to bound state (Fig. 4). Translocation of inactive Rho in
mediate the effects of oxLDL on caveolae composition the cytosol to the membrane where it is active is de-
and eNOS activation in cultured human microvascular pendent on geranylgeranylation [69]. The active form of
endothelial cells [65]. Antibodies for CD36 prevented Rho can induce changes in the actin cytoskeleton and
oxLDL-induced redistribution of eNOS and increased influence intracellular transport, gene transcription and
acetylcholine-induced eNOS activation. Upregulation of mRNA stability [70]. Rho can negatively regulate eNOS
the lectin-like oxidised LDL receptor 1 (LOX-1) by ox- expression [69, 71] and it is the inhibition of Rho
LDL was associated with a reduction in eNOS expres- isoprenylation by statins that was shown to be respon-
sion in human coronary artery endothelial cells in sible for the upregulation of eNOS mRNA expression
culture [66]. Thus, statins could improve eNOS expres- within human endothelial cells in culture [72]. Indeed,
sion by reducing LDL. Treatment of human coronary the effects of statins on Rho isoprenylation and eNOS
artery endothelial cells in culture with simvastatin or expression can be reversed by GGPP. The inhibition of
atorvastatin reduced the oxLDL-induced LOX-1 upre- Rho isoprenylation by statins has been shown to in-
gulation and increased eNOS expression. However, as crease eNOS mRNA stability in cultured human endo-
statins are unable to influence extracellular cholesterol in thelial cells [44, 72, 71]. While statins prevent the
these cell culture experiments, cholesterol-independent activation of Rho by geranylgeranylation, it has been
effects may also have an important role to play in in- noted that a negative feedback signal from the actin
creasing eNOS expression. cytoskeleton results in increased Rho gene transcription
724

Table 2 Mechanism of action of statins on nitric oxide pathways. LDL low-density lipoprotein, LOX-1 lectin-like oxidised LDLreceptor-1, oxLDL oxidised LDL,NO nitric oxide, and accumulation. Therefore, if statin treatment is

Feron et al. 1999 [63]; Everson & Smart 2001 [61]; Sessa 2001 [62]
ceased, the increase in active Rho may induce a rebound

Liao et al. 1995 [57], Vidal et al. 1998 [58], Hernandez-Perera

Laufs & Liao 1998 [72], 2000 [69]; Takemoto et al. 2002 [71]
inhibition of NO [73].

Oxidative stress, NO and statins

Dimmeler et al. 1999 [91]; Fulton et al. 1999 [92]

Feron et al. 2001 [51]; Brouet et al. 2001 [95]


Endres et al. 1998 [42]; Scalia et al. 2001 [52]
Oxygen radicals impair endothelial function and accel-

Wagner et al. 2000 [84]; Wassman 2002 [85]


Feron et al. 2001 [51]; Pelat et al. 2002 [64]
erate the progression of atherosclerotic lesions by
et al. 1998 [59]; Mehta et al. 2001 [66]

promoting lipid oxidation, the expression of proinflam-


matory genes and by oxidative inactivation of endothe-
Laufs & Liao 1998 [72], 2000 [69]

lial NO [74, 75, 76]. Superoxide radicals can react with

Garcia-Cardena et al. 1998 [94]


NO to form peroxynitrite (ONOO)). As ONOO) does
not activate guanylyl cyclase as effectively as NO, the
bioavailability of NO is limited [77]. ONOO) has also
Kureishi et al. 2000 [90]
Hattori et al. 2002 [83]
Wang et al. 1998 [74]

been shown to transform LDL cholesterol into its pro-


atherogenic oxidised form [75, 78]. In addition to the
inactivation of NO by superoxide, there is evidence to
suggest that oxLDL cholesterol can also inhibit the ac-
Reference

tivity of NO in cultured bovine aortic endothelial cells


eNOS endothelial nitric oxide synthase, NADPH nicotinamide adenine dinucleotide phosphate, HSP heat shock protein

[79]. There are several sources of superoxide radicals


within vascular cells, including NADH/NADPH oxid-
ases, xanthine oxidase and NOS [80]. When the avail-
LDL cholesterol increases the abundance ofcaveolin-1 (an inhibitor of eNOS)

ability of its co-factor tetrahydrobiopterin is limited,


production may determine their effectiveness at improving NO production

eNOS can uncouple and generate superoxide anions [81,


The ability of statins to influence circulating or endothelial mevalonate

82, 121, 122]. Interestingly, statins have been shown to


potentiate the synthesis of tetrahydrobiopterin in rat
Statins potentiate the synthesis of tetrahydrobiopterin (a cofactor

Mevalonate, a precurser of cholesterol and isoprenoid synthesis,

aortic smooth muscle cells in culture [83], which may


Rho negatively regulates eNOS mRNA expression and stability

Statins decrease production of superoxide by NADPH oxidase

Statins promote the phosphorylation and activation of eNOS

shift the balance away from NOS-generated superoxide


for eNOS that reduces production of superoxide by eNOS)
Oxidative stress and oxLDL can inhibit the activityof NO

production to the generation of NO. Statins such as


Protein kinase Akt phosphorylates and activates eNOS

atorvastatin appear to have further indirect anti-oxidant


Statins promote the recruitment of HSP90 to eNOS

effects by preventing isoprenylation of the GTP-binding


OxLDL/native LDL downregulates eNOS mRNA

can limit the effect of statins on NO production

protein p21 Rac, which reduces NADPH oxidase-


Statins may decrease the abundance of caveolin

HSP90 binds to, and rapidly activates, eNOS

dependent superoxide formation by rat endothelial cells


[84] and smooth muscle cells [85] in culture and in vivo
[46, 85]. Fluvastatin treatment in hypercholesterolaemic
rabbits has been shown to reduce superoxide production
Statins inhibit Rho isoprenylation

and improve NO availability in arteries [86]. Thus,


statins may prolong the availability of NO through an
anti-oxidant capacity. The relevance of this anti-oxidant
effect of statins in humans has not yet been clarified
since anti-oxidant vitamin supplements have no effect on
coronary events [87]. Another recent study indicates that
anti-oxidants may limit the beneficial effect of statins on
coronary events [88]. In contrast, a prior study with anti-
oxidants has shown that flow-mediated dilation, a
Details

measure of endothelial function, can be improved when


used in combination with a statin in hypercholestero-
laemic men [89]. Therefore, whether anti-oxidants are
Regulation of NO independent

beneficial and an anti-oxidant activity is a clinically


relevant effect of statins has yet to be demonstrated
satisfactorily.
Additional mechanisms

Mevalonate hypothesis
Cholesterol-dependent

of serum cholesterol

and NO regulation
regulation of NO

Oxidative stress

Additional mechanisms of NO regulation


Mechanism

There are further mechanisms whereby NO release


may be improved by statin treatment. For example,
statins may increase eNOS activity via post-translational
725

Fig. 4 Regulation of small G-


proteins: Rho isoprenylation a
key event in cellular events. PP
pyrophosphate, GG
geranylgeranyl, GEF guanine
nucleotide exchange factors,
GTP guanine triphosphate,
GAP GTPase activating
proteins, HMG-Co A
3-hydroxy-3-methylglutaryl
coenzyme A

activation of the phosphatidylinositol 3-kinase/protein clinical situation, such as an acute coronary syndrome,
kinase Akt (PI3 K/Akt) pathway, as has been reported may increase the rate of myocardial infarction [100].
in cultured human endothelial cells [90]. Phosphoryla-
tion of Akt is an important event in several cellular ac-
tivities. Indeed, production of NO by the endothelium Differences among statins
can be regulated by phosphorylation and activation of
eNOS by Akt [91, 92]. Simvastatin has been shown to Statins differ in their ability to increase NO production
promote phosphorylation and activation of eNOS by in vitro [32, 101]. However, comparative studies of the
Akt, thereby increasing angiogenesis in normocholeste- pleiotropic effects of statins are few. One study of cul-
rolaemic rabbits [90]. Interestingly, the activation of Akt tured bovine aortic endothelial cells has shown that
by statins in total mononuclear cells from human blood simvastatin induced the release of more NO than pra-
is also thought to increase the release of endothelial vastatin, atorvastatin and lovastatin (Table 1) [32]. In
progenitor cells [93], which may play a role in angio- contrast, another study has shown pravastatin to be
genesis and the revascularisation of ischaemic tissue and more effective than simvastatin at releasing NO from
contribute to the benefits of statin therapy in the sec- cultured bovine endothelial cells and producing NO-
ondary prevention of coronary events. mediated vasorelaxation of endothelium-intact rat aortic
Statins may also increase eNOS activity through an rings [50].
interaction with the molecular chaperone, heat shock The reasons why statins differ in their ability to re-
protein 90 (HSP90). Studies have shown that binding of lease NO are unclear. Various factors could influence
HSP90 to eNOS is associated with rapid activation and their effectiveness at increasing NO production. For
release of NO [94]. Statins promote the recruitment of example, it has been shown that regulation of NO syn-
HSP90 to the eNOS protein in cultured bovine endo- thesis by endothelial cells can, at least in part, be cho-
thelial cells [51], an effect that may involve the loss of lesterol dependent. Therefore, the ability of a statin
plasma membrane cholesterol and/or caveolin-1 [62]. to lower LDL cholesterol levels (Table 1) may be rele-
Atorvastatin has been shown to stimulate NO-depen- vant in determining the degree that NO production may
dent angiogenesis independent of eNOS expression. In be improved, particularly in hypercholesterolaemic
cultured human endothelial cells, statin-induced phos- patients.
phorylation of eNOS was directly dependent on the
ability of HSP90 to recruit Akt to the eNOS complex,
and this potentiated NO-mediated angiogenesis [95]. The mevalonate hypothesis
Taken together, the different cholesterol-independent
pathways leading to upregulation of endothelial NO Mevalonate appears to play a major role in the regula-
bioavailability may explain the observation that one of tion of NO production. Statin-induced increases in
the earliest clinical effects of statin treatment is a rapid eNOS within cultured human endothelial cells can be
improvement of NO-dependent vasodilatation [96, 97, reversed by addition of mevalonate [42, 53]. Therefore,
98, 99]. Indeed, increased endothelial NO release has increasing mevalonate concentrations within endothelial
been reported as early as 3 days after initiation of statin cells could limit the NO-mediated benefits of statins and
treatment in patients with elevated LDL cholesterol may explain why there are differences in pleiotropic ef-
levels [97]. Conversely, retrospective analysis suggests ficacy between statins. If circulating mevalonate does
that withdrawal of statin treatment in a vulnerable limit the activity of eNOS in endothelial cells, what
726

would be the source and how would the different statins pravastatin is the least potent statin at inhibiting HMG-
regulate it? Endothelial cell mevalonate concentration CoA reductase from human endothelial cells in culture
may be dependent on extracellular sources or intracel- (Table 1) [31]. Similarly, despite being less lipophilic and
lular production (Fig. 5). It seems most likely that the less potent at inhibiting endothelial HMG-CoA reduc-
liver, being involved in the synthesis of cholesterol, may tase than atorvastatin and simvastatin (Table 1) [28, 31],
be a major source of mevalonate. Hence, it may be hy- rosuvastatin is at least as effective and potent as these
pothesised that circulating mevalonate levels are most agents at increasing eNOS activity in mouse aorta [41].
likely influenced by those statins that easily access, and Thus, the lipophilicity and potency within endothelial
are potent at inhibiting, hepatocyte HMG-CoA reduc- cells does not entirely predict the ability of statins to
tase. Unfortunately, comparative data for the effect of improve NO production/release, and so other unidenti-
statins on circulating mevalonate is scarce. Similarly, fied factors may play a role. Indeed, vascular MCP-1
there is insufficient data currently available on the role of expression can also be downregulated by statins in hy-
circulating mevalonate at limiting the extra-hepatic ef- percholesterolaemic pigs, regardless of their lipophilicity
fects of statins. [103]. It may be that there are specific mechanisms for
Another source of mevalonate that would influence hydrophilic statins to enter endothelial cells. Such a
the pleiotropic activity of statins is the endothelial cell mechanism is present in the liver, where the hepatic or-
itself. Intracellular mevalonate production is likely to be ganic anion transporter (OATP-C) helps hydrophilic
dependent on a statin’s ability to enter endothelial cells statins enter hepatocytes [104], and a similar process in
and its potency for inhibiting endothelial HMG-CoA endothelial cells would explain why the lipophilicity of a
reductase. It has been suggested that the varying lipo- statin does not always predict its pleiotropic efficacy.
philicity of statins affects their uptake into target organs More data on how different statins enter endothelial
and, therefore, their ability to elicit pleiotropic effects cells are needed to address this issue.
[69, 102]. The relative lipophilicity of some commonly It is likely that there is a complex interplay of
available statins and their potency on hepatocyte and mechanisms determining the effectiveness of statins for
endothelial cell HMG-CoA reductase are listed for increasing NO release. Further comparative and mech-
comparison in Table 1. Lipophilic statins, such as lo- anistic studies are required if this pleiotropic effect is to
vastatin and cerivastatin, are considered more likely to be understood fully and predictions made on the utility
enter endothelial cells by passive diffusion than hydro- of different statins for increasing NO in humans.
philic statins, such as pravastatin and rosuvastatin,
which are targeted to the liver. However, in bovine en-
dothelial cells in culture, where external sources of Benefits of statins by improving endothelial function
cholesterol and mevalonate are absent, pravastatin was
at least as effective as more lipophilic statins such as Animal experiments
atorvastatin, lovastatin [32] and simvastatin [50] at
stimulating the release of NO from cultured bovine By increasing NO production, statins can interfere with
endothelial cells. Moreover, it would also seem that atherosclerotic lesion development, thereby improving
vascular health and reducing the risk of atherosclerotic
disease. The beneficial effect of increasing NO release
with statins has been shown in several animal models of
cardiovascular disease. For example, a study with flu-
vastatin has demonstrated regression of atherosclerosis
in rabbits, an effect associated with upregulation of
eNOS mRNA and restoration of endothelial function in
cholesterol-fed rabbits [47]. In addition, simvastatin
provided protection against stroke by increasing cere-
bral blood flow in wild-type mice, although this benefit
was not observed in eNOS ()/)) mice [42], indicating
that NO mediates this effect of statin treatment. Simi-
larly, treatment with the synthetic statins, atorvastatin
or rosuvastatin significantly reduced cerebral infarct
volume after an hour of cerebral artery occlusion in
normocholesterolaemic mice [41, 105]. This protective
effect was mediated in part by upregulation of eNOS
and a subsequent decrease in platelet activation. Other
experiments show protection from ischaemic damage
Fig. 5 Possible mechanisms whereby both lipophilic and hydro- with statin treatment. Following an episode of hindlimb
philic statins may improve endothelial release of nitric oxide (NO).
EC endothelial cell, Acetyl-Co A acetyl coenzyme A, HMG-Co A ischaemia in mice, collateral vessel growth and enhanced
3-hydroxy-3-methylglutaryl coenzyme A, eNOS endothelial nitric blood flow was promoted by cerivastatin, and this was
oxide synthase associated with an increase in eNOS activity [106].
727

Simvastatin has been reported to limit contractile dys- function, treatment with high-dose atorvastatin (80 mg)
function in isolated rat hearts subjected to global myo- increased forearm blood flow within 24 h, whereas se-
cardial ischaemia and reperfusion [13]. This protective rum cholesterol and high-sensitivity C reactive protein
effect was attributed to a NO-mediated attenuation of (hs-CRP) were not decreased until 2 days of treatment.
neutrophil infiltration into the myocardium [13]. Simi- Cessation of statin treatment after 30 days resulted in a
larly, a reduction in myocardial injury following ischa- rebound of forearm blood flow within 24 h of with-
emia/reperfusion in rats in vivo has been observed with drawal, whereas cholesterol and hs-CRP slowly and
rosuvastatin treatment [107]. These latter effects are steadily returned to baseline [111]. Therefore, the po-
likely a result of decreased leucocyte–endothelium in- tential importance of cholesterol-independent effects of
teractions. For example, rosuvastatin reduced expres- statins in humans may also relate to the time course of
sion of P-selectin and, consequently, leucocyte adhesion their effects, for example, after withdrawal of treatment
and migration in rats [49]. This effect with rosuvastatin [101].
was not observed in eNOS-deficient mice [49, 52], indi- Theoretically, it may be possible to observe a rela-
cating that the anti-inflammatory benefits with statin tionship between the cholesterol-independent effect of
treatment are a result of an improvement in endothelial statins on NO and cardiovascular outcomes by com-
function and NO release. paring statin-treated patients with individuals whose
cholesterol was lowered to the same degree with a non-
statin, lipid-lowering agent that does not influence NO.
Human studies In cynomolgus monkeys, who develop atherosclerosis
very similar to humans, cholesterol-independent effects
Statins have been shown to increase NO production in of statins, including upregulation of endothelial NO,
humans. For example, fluvastatin increased the bio- were demonstrated using a ‘‘clamped-cholesterol’’ de-
availability of NO in a randomised, double-blind, pla- sign, where dietary cholesterol was adjusted to equalise
cebo-controlled trial of 29 hypercholesterolaemic cholesterol levels between groups [39, 112]. Inhibitors of
patients [108]. Similarly, simvastatin has also been NO production are unlikely to be useful to differentiate
shown to improve endothelial function, augment basal between NO-mediated and lipid-dependent effects of
and acetylcholine-stimulated vasodilation, over 4 weeks statins in long-term studies since NO plays such an in-
in a double-blind, placebo-controlled, crossover trial tegral role in normal vascular function. Unfortunately,
[98]. Moreover, the NO enhancing effects of statins oc- many potential studies are not currently possible be-
cur at clinically relevant doses [109]. cause of a lack of suitable pharmacological tools. NO
However, it is difficult to determine whether this does however appear to have some clinical relevance in
ability of statin treatment to increase NO production is atherosclerotic disease. For example, there is a reduced
clinically important. In a clinical setting, it would not contribution of NO to vasodilation in patients with
be easy to differentiate between benefits derived from hypercholesterolaemia [113] or risk factors for athero-
improved NO release and the well-reported advantages sclerosis [114]. In addition, restoring vascular NO for-
of lipid lowering, particularly as cholesterol may mation can improve symptoms of intermittent
modulate the production of NO. Indeed, changes in claudication in patients with peripheral arterial disease
forearm blood flow were reported to be inversely re- [115]. A prospective trial to test a potentially negative
lated to alterations in LDL cholesterol levels in humans effect of withdrawal of statin treatment mediated by
[108]. Furthermore, both the inhibition of cholesterol reduced NO production is ethically impossible. Thus,
synthesis and the cholesterol-independent regulation of there is compelling circumstantial evidence that im-
NO by statins are dependent on the production of a proved NO production by statins may be clinically rel-
common precurser, mevalonate (Fig. 3). Consequently, evant, but it may be some time before such an effect is
it is unsurprising that the two effects are often inversely proven to contribute to the reduction in cardiovascular
related. However, despite these difficulties, a short-term events.
study with pravastatin has demonstrated a NO-medi-
ated improvement in forearm blood flow and an in-
crease in urinary nitrite/nitrate production, indicative Conclusions
of an increase in NO production, which occurred in-
dependently of changes in cholesterol levels in normo- Statins improve endothelial dysfunction by enhancing
cholesterolaemic patients with other risk factors for the production of NO, which may partially account for
CHD [110]. In another study, short-term (2 weeks) their beneficial effects in reducing the incidence of car-
lipid-lowering therapy with cerivastatin improved en- diovascular events. The mechanisms of increased NO
dothelial function and NO bioavailability in patients production by statins may be cholesterol dependent and/
with hypercholesterolaemia [97]. This improvement in or cholesterol independent.
endothelium-dependent vasodilation was no longer Statins vary in their efficacy for increasing NO pro-
observed when the NO synthase inhibitor N(G)- duction. The exact mechanisms dictating these differences
monomethyl-L-arginine was co-infused. Similarly, in have yet to be fully elucidated, but may be related to their
normocholesterolaemic men with normal vascular ability to reduce LDL cholesterol, to lower circulating
728

and/or endothelial cell mevalonate or by their lipophilic- 16. Ijem J, Granlie C (2000) More than cholesterol: the com-
ity and ability to access endothelial cells. plexity of coronary artery disease. S D J Med 53:489–491
17. Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz
By improving endothelial function and NO produc- MA, Bevan JA, Fishman MC (1995) Hypertension in mice
tion, statin therapy may limit the development of ath- lacking the gene for endothelial nitric oxide synthase. Nature
erosclerosis and improve cardiovascular clinical 377:239–242
outcomes for patients. Furthermore, in patients with 18. Huang Z, Huang PL, Ma J, Meng W, Ayata C, Fishman MC,
Moskowitz MA (1996) Enlarged infarcts in endothelial nitric
atherosclerosis, increasing NO levels with statins may oxide synthase knockout mice are attenuated by nitro-L-arg-
decrease the likelihood of an acute event by stabilising inine. J Cereb Blood Flow Metab 16:981–987
plaques, increasing blood flow and reducing platelet 19. Huang PL (1998) Disruption of the endothelial nitric oxide
aggregation. synthase gene: effect on vascular response to injury. Am J
Cardiol 82:57S–59S
20. Ni W, Egashira K, Kataoka C, Kitamoto S, Koyanagi M,
Acknowledgements Thanks to G. Allcock for editorial assistance in
Inoue S, Takeshita A (2001) Antiinflammatory and antiarte-
the preparation of this manuscript. U.L. received financial support
riosclerotic actions of HMG-CoA reductase inhibitors in a rat
from and has been a speaker for AstraZeneca, Bayer, MSD,
model of chronic inhibition of nitric oxide synthesis. Circ Res
Novartis, Pfizer and Sankyo. U.L.’s research was supported by the
89:415–421
Deutsche Forschungsgemeinschaft and the Universität des Saar-
21. LaRosa JC, Hunninghake D, Bush D, Criqui MH, Getz GS,
landes.
Gotto AM Jr, Grundy SM, Rakita L, Robertson RM, Weis-
feldt ML, Cleeman JI (1990) The cholesterol facts. A summary
of the evidence relating dietary fats, serum cholesterol, and
coronary heart disease. A joint statement by the American
References Heart Association and the National Heart, Lung and Blood
Institute. The Task Force on Cholesterol Issues, American
1. Libby P, Ridker PM, Maseri A (2002) Inflammation and Heart Association. Circulation 81:1721–1733
atherosclerosis. Circulation 105:1135–1143 22. Coresh J, Kwiterovich PO Jr (1996) Small, dense low-density
2. Kinlay S, Ganz P (2000) Relation between endothelial dys- lipoprotein particles and coronary heart disease risk: a clear
function and the acute coronary syndrome: implications for association with uncertain implications. JAMA 276:914–915
therapy. Am J Cardiol 86[Suppl]:10J–13J 23. Wood D, De Backer G, Faergeman O, Graham I, Mancia G,
3. Liao JK (1998) Endothelium and acute coronary syndromes. Pyörälä K (1998) Prevention of coronary heart disease in
Clin Chem 44:1799–1808 clinical practice: recommendations of the second joint task
4. Libby P (2001) Current concepts of the pathogenesis of the force of European and other societies on coronary prevention.
acute coronary syndromes. Circulation 104:365–372 Eur Heart J 19:1434–1503
5. Joannides R, Haefeli WE, Linder L, Richard V, Bakkali EH, 24. Gotto AM Jr, Kuller LH (2002) Eligibility for lipid-lowering
Thuillez C, Luscher TF (1995) Nitric oxide is responsible for drug therapy in primary prevention: how do the Adult
flow-dependent dilatation of human peripheral conduit ar- Treatment Panel II and Adult Treatment Panel III guidelines
teries in vivo. Circulation 91:1314–1319 compare? Circulation 105:136–139
6. Garg UC, Hassid A (1989) Nitric oxide-generating vasodila- 25. Istvan ES, Deisenhofer J (2001) Structural mechanism for
tors and 8-bromo-cyclic guanosine monophosphate inhibit statin inhibition of HMG-CoA reductase. Science 292:1160–
mitogenesis and proliferation of cultured rat vascular smooth 1164
muscle cells. J Clin Invest 83:1774–1777 26. Maron DJ, Fazio S, Linton MF (2000) Current perspectives
7. Fleming I, Busse R (1999) NO: the primary EDRF. J Mol on statins. Circulation 101:207–213
Cell Cardiol 31:514 27. Olsson AG, Pears J, McKellar J, Mizan J, Raza A (2001)
8. Vallance P, Collier J, Moncada S (1989) Nitric oxide syn- Effect of rosuvastatin on low-density lipoprotein cholesterol in
thesised from L-arginine mediates endothelium dependent patients with hypercholesterolemia. Am J Cardiol 88:504–508
dilatation in human veins in vivo. Cardiovasc Res 23:1053– 28. Scandinavian Simvastatin Survival Study Group (1994)
1057 Randomised trial of cholesterol lowering in 4444 patients with
9. Dimmeler S, Zeiher AM (1999) Nitric oxide – an endothelial coronary heart disease: the Scandinavian Simvastatin Survival
cell survival factor. Cell Death Differ 6:964–968 Study (4S). Lancet 344:1383–1389
10. Radomski MW, Palmer RM, Moncada S (1987) Endogenous 29. Shepherd J, Cobbe SM, Ford I, Isles CG, Lorimer AR,
nitric oxide inhibits human platelet adhesion to vascular MacFarlane PW, McKillop JH, Packard CJ, for the West of
endothelium. Lancet 2:1057–1058 Scotland Coronary Prevention Study Group (1995) Preven-
11. Peng HB, Libby P, Liao JK (1995) Induction and stabilization tion of coronary heart disease with pravastatin in men with
of I kappa B alpha by nitric oxide mediates inhibition of NF- hypercholesterolemia. N Engl J Med 333:1301–1307
kappa B. J Biol Chem 270:14214–14219 30. Sacks FM, Pfeffer MA, Moye LA, Rouleau JL, Rutherford
12. Kubes P, Suzuki M, Granger DN (1991) Nitric oxide: an JD, Cole TG, Brown L, Warnica JW, Arnold JM, Wun C-C,
endogenous modulator of leukocyte adhesion. Proc Natl Acad Davis BR, Braunwald E (1996) The effect of pravastatin on
Sci USA 88:4651–4655 coronary events after myocardial infarction in patients with
13. Lefer AM, Campbell B, Shin YK, Scalia R, Hayward R, Lefer average cholesterol levels. Cholesterol and Recurrent Events
DJ (1999) Simvastatin preserves the ischemic-reperfused Trial investigators. N Engl J Med 335:1001–1009
myocardium in normocholesterolemic rat hearts. Circulation 31. Downs JR, Clearfield M, Weis S, Whitney E, Shapiro DR,
100:178–184 Beere PA, Langendorfer A, Stein EA, Kruyer W, Gotto AM
14. De Caterina R, Libby P, Peng HB, Thannickal VJ, Rajav- Jr (1998) Primary prevention of acute coronary events with
ashisth TB, Gimbrone MA Jr, Shin WS, Liao JK (1995) Nitric lovastatin in men and women with average cholesterol levels:
oxide decreases cytokine-induced endothelial activation. results of AFCAPS/TexCAPS. Air Force/Texas Coronary
Nitric oxide selectively reduces endothelial expression of Atherosclerosis Prevention Study. JAMA 279:1615–1622
adhesion molecules and proinflammatory cytokines. J Clin 32. The Long-term Intervention with Pravastatin in Ischemic
Invest 96:60–68 Disease (LIPID) Study Group (1998) Prevention of cardio-
15. Tsao PS, Wang B, Buitrago R, Shyy JY, Cooke JP (1997) vascular events and death with pravastatin in patients with
Nitric oxide regulates monocyte chemotactic protein-1. coronary heart disease and a broad range of initial cholesterol
Circulation 96:934–940 levels. N Engl J Med 339:1349–1357
729

33. Heart Protection Study Collaborative Group (2002) MRC/ 50. Kaesemeyer WH, Caldwell RB, Huang J, Caldwell RW (1999)
BHF heart protection study of cholesterol lowering with Pravastatin sodium activates endothelial nitric oxide synthase
simvastatin in 20,536 high-risk individuals: a randomised independent of its cholesterol-lowering actions. J Am Coll
placebo-controlled trial. Lancet 360:722 Cardiol 33:234–241
34. West of Scotland Coronary Prevention Study Group (1998) 51. Feron O, Dessy C, Desager JP, Balligand JL (2001) Hydroxy-
Influence of pravastatin and plasma lipids on clinical events in methylglutaryl-coenzyme A reductase inhibition promotes
the West of Scotland coronary prevention study (WOSCOPS). endothelial nitric oxide synthase activation through a decrease
Circulation 97:1440–1445 in caveolin abundance. Circulation 103:113–118
35. Scandinavian Simvastatin Survival Study Group (1995) Base- 52. Scalia R, Stalker TJ, Lefer AM (2001) Mechanisms of the
line serum cholesterol and treatment effect in the Scandinavian anti-inflammatory action of rosuvastatin, a new HMG-CoA
simvastatin survival study (4S). Lancet 345:1274–1275 reductase inhibitor. Atherosclerosis Suppl 2:37
36. Prospective Studies Collaboration (1995) Cholesterol, dia- 53. Martinez-Gonzalez J, Raposo B, Rodriguez C, Badimon L
stolic blood pressure, and stroke: 13,000 strokes in 450,000 (2001) 3-hydroxy-3-methylglutaryl coenzyme a reductase
people in 45 prospective cohorts. Lancet 346:1647–1653 inhibition prevents endothelial NO synthase downregulation
37. Pedersen TR, Kjekshus J, Pyörälä K, Olsson AG, Cook TJ, by atherogenic levels of native LDLs: balance between tran-
Musliner TA, Tobert JA, Haghfelt T (1998) Effect of sim- scriptional and posttranscriptional regulation. Arterioscler
vastatin on ischemic signs and symptoms in the Scandinavian Thromb Vasc Biol 21:804–809
simvastatin survival study (4S). Am J Cardiol 81:333–335 54. Grunler J, Ericsson J, Dallner G (1994) Branch-point
38. Liao JK (2002) Statins and ischemic stroke. Atherosclerosis reactions in the biosynthesis of cholesterol, dolichol, ubiq-
Suppl 3:21–25 uinone and prenylated proteins. Biochim Biophys Acta
39. Williams JK, Sukhova GK, Herrington DM, Libby P (1998) 1212:259–277
Pravastatin has cholesterol-lowering independent effects on 55. Tamai O, Matsuoka H, Itabe H, Wada Y, Kohno K, Imaiz-
the artery wall of atherosclerotic monkeys. J Am Coll Cardiol umi T (1997) Single LDL apheresis improves endothelium-
31:684–691 dependent vasodilation in hypercholesterolemic humans.
40. Havekes LM, van Duyvenvoorde W, Maas MCE, van der Circulation 95:76–82
Boom H, van den Hoogen CM, Emeis JJ, Princen HMG 56. Mellwig KP, Baller D, Gleichmann U, Moll D, Betker S,
(2002) Rosuvastatin reduces atherosclerosis independently of Weise R, Notohamiprodjo G (1998) Improvement of coro-
its cholesterol-lowering effect in APOE*3 Leiden transgenic nary vasodilatation capacity through single LDL apheresis.
mice. Atherosclerosis 3:187 Atherosclerosis 139:173–178
41. Laufs U, Gertz K, Dirnagl U, Bohm M, Nickenig G, Endres 57. Liao JK, Shin WS, Lee WY, Clark SL (1995) Oxidized low-
M (2002) Rosuvastatin, a new HMG-CoA reductase inhibitor, density lipoprotein decreases the expression of endothelial
upregulates endothelial nitric oxide synthase and protects nitric oxide synthase. J Biol Chem 270:319–324
from ischemic stroke in mice. Brain Res 942:23–30 58. Vidal F, Colome C, Martinez-Gonzalez J, Badimon L (1998)
42. Endres M, Laufs U, Huang Z, Nakamura T, Huang P, Atherogenic concentrations of native low-density lipoproteins
Moskowitz MA, Liao JK (1998) Stroke protection by 3-hy- down-regulate nitric-oxide-synthase mRNA and protein levels
droxy-3-methylglutaryl (HMG)-CoA reductase inhibitors in endothelial cells. Eur J Biochem 252:378–384
mediated by endothelial nitric oxide synthase. Proc Natl Acad 59. Hernandez-Perera O, Perez-Sala D, Navarro-Antolin J,
Sci U S A 95:8880–8885 Sanchez-Pascuala R, Hernandez G, Diaz C, Lamas S (1998)
43. Mueck AO, Seeger H, Wallwiener D (2001) Further evidence Effects of the 3-hydroxy-3-methylglutaryl-CoA reductase in-
for direct vascular actions of statins: effect on endothelial hibitors, atorvastatin and simvastatin, on the expression of
nitric oxide synthase and adhesion molecules. Exp Clin endothelin-1 and endothelial nitric oxide synthase in vascular
Endocrinol Diabetes 109:181–183 endothelial cells. J Clin Invest 101:2711–2719
44. Laufs U, Fata VL, Liao JK (1997) Inhibition of 3-hydroxy- 60. Laufs U, La Fata V, Plutzky J, Liao JK (1998) Upregulation
3-methylglutaryl (HMG)-CoA reductase blocks hypoxia- of endothelial nitric oxide synthase by HMG CoA reductase
mediated down-regulation of endothelial nitric oxide synthase. inhibitors. Circulation 97:1129–1135
J Biol Chem 272:31725–31729 61. Everson WV, Smart EJ (2001) Influence of caveolin, choles-
45. Yamada M, Huang Z, Dalkara T, Endres M, Laufs U, terol, and lipoproteins on nitric oxide synthase: implications
Waeber C, Huang PL, Liao JK, Moskowitz MA (2000) for vascular disease. Trends Cardiovasc Med 11:246–250
Endothelial nitric oxide synthase-dependent cerebral blood 62. Sessa WC (2001) Can modulation of endothelial nitric oxide
flow augmentation by L-arginine after chronic statin treat- synthase explain the vasculoprotective actions of statins?
ment. J Cereb Blood Flow Metab 20:709–717 Trends Mol Med 7:189–191
46. Wassmann S, Laufs U, Baumer AT, Muller K, Ahlbory K, 63. Feron O, Dessy C, Moniotte S, Desager JP, Balligand JL
Linz W, Itter G, Rosen R, Bohm M, Nickenig G (2001) (1999) Hypercholesterolemia decreases nitric oxide production
HMG-CoA reductase inhibitors improve endothelial dys- by promoting the interaction of caveolin and endothelial nitric
function in normocholesterolemic hypertension via reduced oxide synthase. J Clin Invest 103:897–905
production of reactive oxygen species. Hypertension 37:1450– 64. Pelat M, Dessy C, Feron O, Balligand J-L (2002) HMG-CoA
1457 reductase inhibition decreases caveolin-1 independently of
47. Kano H, Hayashi T, Sumi D, Esaki T, Asai Y, Thakur NK, plasma lipid lowering in heart and blood vessels of apoE ()/))
Jayachandran M, Iguchi A (1999) A HMG-CoA reductase mice. Int J Clin Pract Suppl 124:4
inhibitor improved regression of atherosclerosis in the rabbit 65. Uittenbogaard A, Shaul PW, Yuhanna IS, Blair A, Smart EJ
aorta without affecting serum lipid levels: possible relevance of (2000) High density lipoprotein prevents oxidized low density
up-regulation of endothelial NO synthase mRNA. Biochem lipoprotein-induced inhibition of endothelial nitric-oxide
Biophys Res Commun 259:414–419 synthase localization and activation in caveolae. J Biol Chem
48. Mital S, Zhang X, Zhao G, Bernstein RD, Smith CJ, Fulton 275:11278–11283
DL, Sessa WC, Liao JK, Hintze TH (2000) Simvastatin 66. Mehta JL, Li DY, Chen HJ, Joseph J, Romeo F (2001)
upregulates coronary vascular endothelial nitric oxide pro- Inhibition of LOX-1 by statins may relate to upregulation of
duction in conscious dogs. Am J Physiol Heart Circ Physiol eNOS. Biochem Biophys Res Commun 289:857–861
279:H2649–H2657 67. Chen H, Ikeda U, Shimpo M, Shimada K (2000) Direct effects
49. Stalker TJ, Lefer AM, Scalia R (2001) A new HMG-CoA of statins on cells primarily involved in atherosclerosis.
reductase inhibitor, rosuvastatin, exerts anti-inflammatory Hypertens Res 23:187–192
effects on the microvascular endothelium: the role of meva- 68. Hall A (1998) Rho GTPases and the actin cytoskeleton.
lonic acid. Br J Pharmacol 133:406–412 Science 279:509–514
730

69. Laufs U, Liao JK (2000) Direct vascular effects of HMG-CoA 89. Neunteufl T, Kostner K, Katzenschlager R, Zehetgruber M,
reductase inhibitors. Trends Cardiovasc Med 10:143–148 Maurer G, Weidinger F (1998) Additional benefit of vitamin E
70. Laufs U, Liao JK (2000) Targeting Rho in cardiovascular supplementation to simvastatin therapy on vasoreactivity of
disease. Circ Res 87:526–528 the brachial artery of hypercholesterolemic men. J Am Coll
71. Takemoto M, Sun J, Hiroki J, Shimokawa H, Liao JK (2002) Cardiol 32:711–716
Rho-kinase mediates hypoxia-induced downregulation of 90. Kureishi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer DJ,
endothelial nitric oxide synthase. Circulation 106:57–62 Sessa WC, Walsh K (2000) The HMG-CoA reductase inhib-
72. Laufs U, Liao JK (1998) Post-transcriptional regulation of itor simvastatin activates the protein kinase Akt and promotes
endothelial nitric oxide synthase mRNA stability by Rho angiogenesis in normocholesterolemic animals. Nat Med
GTPase. J Biol Chem 273:24266–24271 6:1004–1010
73. Laufs U, Endres M, Custodis F, Gertz K, Nickenig G, Liao 91. Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R,
JK, Böhm M (2000) Suppression of endothelial nitric oxide Zeiher AM (1999) Activation of nitric oxide synthase in en-
production after withdrawal of statin treatment is mediated by dothelial cells by Akt-dependent phosphorylation. Nature
negative feedback regulation of Rho GTPase gene transcrip- 399:601–605
tion. Circulation 102:3104–3110 92. Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y,
74. Wang HD, Pagano PJ, Du YT, Cayatte AJ, Quinn MT, Walsh K, Franke TF, Papapetropoulos A, Sessa WC (1999)
Brecher P, Cohen RA (1998) Superoxide anion from the ad- Regulation of endothelium-derived nitric oxide production by
ventitia of the rat thoracic aorta inactivates nitric oxide. Circ the protein kinase Akt. Nature 399:597–601
Res 82:810–818 93. Dimmeler S, Aicher A, Vasa M, Mildner-Rihm C, Alder K,
75. Darley-Usmar VM, Hogg N, O’Leary VJ, Wilson MT, Tiemann M, Rutten H, Fichtlscherer S, Martin H, Zeiher AM
Moncada S (1992) The simultaneous generation of superoxide (2001) HMG-CoA reductase inhibitors (statins) increase en-
and nitric oxide can initiate lipid peroxidation in human low dothelial progenitor cells via the PI 3-kinase/Akt pathway. J
density lipoprotein. Free Radic Res Commun 17:920 Clin Invest 108:391–397
76. Nickenig G, Harrison DG (2002) The AT(1)-type angiotensin 94. Garcia-Cardena G, Fan R, Shah V, Sorrentino R, Cirino G,
receptor in oxidative stress and atherogenesis. Part I: oxidative Papapetropoulos A, Sessa WC (1998) Dynamic activation of
stress and atherogenesis. Circulation 105:393–396 endothelial nitric oxide synthase by Hsp90. Nature 392:821–824
77. Gryglewski RJ, Palmer RM, Moncada S (1986). Superoxide 95. Brouet A, Sonveaux P, Dessy C, Moniotte S, Balligand JL,
anion is involved in the breakdown of endothelium-derived Feron O (2001) Hsp90 and caveolin are key targets for the
vascular relaxing factor. Nature 320:454–456 proangiogenic nitric oxide-mediated effects of statins. Circ Res
78. Hogg N, Darley-Usmar VM, Graham A, Moncada S (1993) 89:866–873
Peroxynitrite and atherosclerosis. Biochem Soc Trans 21:358– 96. Anderson TJ, Meredith IT, Yeung AC, Frei B, Selwyn AP,
362 Ganz P (1995) The effect of cholesterol-lowering and antiox-
79. Chin JH, Azhar S, Hoffman BB (1992) Inactivation of endo- idant therapy on endothelium-dependent coronary vasomo-
thelial-derived relaxing factor by oxidized lipoproteins. J Clin tion. N Engl J Med 332:488–493
Invest 89:10–18 97. John S, Delles C, Jacobi J, Schlaich MP, Schneider M, Sch-
80. Kojda G, Harrison D (1999) Interactions between NO and mitz G, Schmieder RE (2001) Rapid improvement of nitric
reactive oxygen species: pathophysiological importance in oxide bioavailability after lipid-lowering therapy with ceri-
atherosclerosis, hypertension, diabetes and heart failure. vastatin within two weeks. J Am Coll Cardiol 37:1351–1358
Cardiovasc Res 43:562–571 98. O’Driscoll G, Green D, Taylor RR (1997) Simvastatin, an
81. Xia Y, Dawson VL, Dawson TM, Snyder SH, Zweier JL HMG-coenzyme A reductase inhibitor, improves endothelial
(1996) Nitric oxide synthase generates superoxide and nitric function within 1 month. Circulation 95:1126–1131
oxide in arginine-depleted cells leading to peroxynitrite-me- 99. Tsunekawa T, Hayashi T, Kano H, Sumi D, Matsui-Hirai H,
diated cellular injury. Proc Natl Acad Sci U S A 93:6770–6774 Thakur NK, Egashira K, Iguchi A (2001) Cerivastatin, a hy-
82. Stuehr D, Pou S, Rosen GM (2001) Oxygen reduction by droxymethylglutaryl coenzyme a reductase inhibitor, im-
nitric oxide synthases. J Biol Chem 276:14533–14536 proves endothelial function in elderly diabetic patients within
83. Hattori Y, Nakanishi N, Kasai K (2002) Statin enhances 3 days. Circulation 104:376–379
cytokine-mediated induction of nitric oxide synthesis in 100. Heeschen C, Hamm CW, Laufs U, Snapinn S, Böhm M,
vascular smooth muscle cells. Cardiovasc Res 54:649–658 White HD. Platelet and Receptor Inhibition in Ischaemic
84. Wagner AH, Kohler T, Ruckschloss U, Just I, Hecker M Syndrome Management (PRISM) Investigators (2002) With-
(2000) Improvement of nitric oxide-dependent vasodilatation drawal of statins increases event rates in patients with acute
by HMG-CoA reductase inhibitors through attenuation of coronary syndromes. Circulation 105:1446–1452
endothelial superoxide anion formation. Arterioscler Thromb 101. Aengevaeren WR (1999) Beyond lipids the role of the endo-
Vasc Biol 20:61–69 thelium in coronary artery disease. Atherosclerosis 147[Suppl
85. Wassman S, Laufs U, Muller K, Konkol C, Ahlbory K, 1]:S11–S16
Baumer AT, Linz W, Bohm M, Nickenig G (2002) Cellular 102. Fukumoto Y, Libby P, Rabkin E, Hill CC, Enomoto M,
antioxidant effects of atorvastatin in vitro and in vivo. Arte- Hirouchi Y, Shiomi M, Aikawa M (2001) Statins alter smooth
rioscler Thromb Vasc Biol 22:300–305 muscle cell accumulation and collagen content in established
86. Sumi D, Hayashi T, Thankur NK, Jayachandran M, Asai Y, atheroma of Watanabe heritable hyperlipidemic rabbits. Cir-
Kano H, Matsui H, Iguchi A (2001) A HMG-CoA reductase culation 103:993–999
inhibitor possesses a potent anti-atherosclerotic effect other 103. Martinez-Gonzalez J, Alfon J, Berrozpe M, Badimon L (2001)
than serum lipid lowering effects the relevance of endothelial HMG-CoA reductase inhibitors reduce vascular monocyte
nitric oxide synthase and superoxide anion scavenging action. chemotactic protein-1 expression in early lesions from hyper-
Atherosclerosis 155:347–357 cholesterolemic swine independently of their effect on plasma
87. Heart Protection Study Collaborative Group (2002) MRC/ cholesterol levels. Atherosclerosis 159:27–33
BHF heart protection study of antioxidant vitamin supple- 104. Corsini A, Bellosta S, Baetta R, Fumagalli R, Paoletti R, Bernini
mentation in 20,536 high-risk individuals: a randomised F (1999) New insights into the pharmacodynamic and phar-
placebo-controlled trial. Lancet 360:23–33 macokinetic properties of statins. Pharmacol Ther 84:413–428
88. Brown BG, Zhao X-Q, Chait A, Fisher LD, Cheung MC, 105. Laufs U, Gertz K, Huang P, Nickenig G, Bohm M, Dirnagl
Morse JS, Dowdy AA, Marino EK, Bolson EL, Alaupovic P, U, Endres M (2000) Atorvastatin upregulates type III nitric
Frohlich J, Albers JJ (2001) Simvastatin and niacin, antioxi- oxide synthase in thrombocytes, decreases platelet activation,
dant vitamins, or the combination for the prevention of and protects from cerebral ischemia in normocholesterolemic
coronary disease. N Engl J Med 345:1583–1592 mice. Stroke 31:2442–2449
731

106. Sata M, Nishimatsu H, Suzuki E, Sugiura S, Yoshizumi M, 114. Quyyumi AA, Dakak N, Andrews NP, Gilligan DM, Panza
Ouchi Y, Hirata Y, Nagai R (2001) Endothelial nitric oxide JA, Cannon RO III (1995) Contribution of nitric oxide to
synthase is essential for the HMG-CoA reductase inhibitor metabolic coronary vasodilation in the human heart. Circu-
cerivastatin to promote collateral growth in response to isch- lation 92:320–326
emia. FASEB J 15:2530–2532 115. Boger RH, Bode-Boger SM, Thiele W, Creutzig A, Alexander
107. Li W, Asagami T, McTaggart F, Tsao PS (2002) Rosuvastatin K, Frolich JC (1998) Restoring vascular nitric oxide forma-
diminishes myocardial injury after ischemia/reperfusion. Int tion by L-arginine improves the symptoms of intermittent
J Clin Pract Suppl 124:6 claudication in patients with peripheral arterial occlusive dis-
108. John S, Schlaich M, Langenfeld M, Weihprecht H, Schmitz G, ease. J Am Coll Cardiol 32:1336–1344
Weidinger G, Schmieder RE (1998) Increased bioavailability 116. McTaggart F, Buckett L, Davidson R, Holdgate G, McCor-
of nitric oxide after lipid-lowering therapy in hypercholeste- mick A, Schneck R, Smith G, Warwick M (2001) Preclinical
rolemic patients: a randomized, placebo-controlled, double- and clinical pharmacology of rosuvastatin, a new 3-hydroxy-
blind study. Circulation 98:211–216 3-methylglutaryl coenzyme A reductase inhibitor. Am J Car-
109. Lefer AM, Scalia R, Lefer DJ (2001) Vascular effects of HMG diol 87 [Suppl]:28B–32B
CoA-reductase inhibitors (statins) unrelated to cholesterol 117. Hanefield M (2001) Clinical rationale for rosuvastatin, a
lowering: new concepts for cardiovascular disease. Cardiovasc potent new HMG-CoA reductase inhibitor. Int J Clin Pract
Res 49:281–287 55:399–405
110. Masumoto A, Hirooka Y, Hironaga K, Eshima K, Setoguchi 118. Knopp RH (1999) Drug treatment of lipid disorders. N Engl
S, Egashira K, Takeshita A (2001) Effect of pravastatin on J Med 341:498–511
endothelial function in patients with coronary artery disease 119. Buckett L, Ballard P, Davidson R, Dunkley C, Martin L,
(cholesterol-independent effect of pravastatin). Am J Cardiol Stafford J, McTaggart F (2000) Selectivity of ZD4522 for
88:1291–1294 inhibition of cholesterol synthesis in hepatic versus non-he-
111. Laufs U, Wassmann S, Hilgers S, Ribaudo N, Bohm M, patic cells. Atherosclerosis 151:41
Nickenig G (2001) Rapid effects on vascular function after 120. Dobrucki LW, Kalinowski L, Dobrucki IT, Malinski T (2001)
initiation and withdrawal of atorvastatin in healthy, normo- Statin-stimulated nitric oxide release from endothelium. Med
cholesterolemic men. Am J Cardiol 88:1306–1307 Sci Monit 7:622–627
112. Sukhova GK, Williams JK, Libby P (2002) Statins reduce 121. Griendling KK, Harrison DG (2001) Out, damned dot:
inflammation in atheroma of nonhuman primates independent studies of the NADPH oxidase in atherosclerosis. J Clin
of effects on serum cholesterol. Arterioscler Thromb Vasc Biol Invest 108:1423–1424
22:1452–1458 122. Heitzer T, Schlinzig T, Krohn K, Meinertz T, Münzel T
113. Preik M, Kelm M, Schoebel F, Schottenfeld Y, Leschke M, (2001) Endothelial dysfunction, oxidative stress, and risk of
Strauer BE (1996) Selective impairment of nitric oxide cardiovascular events in patients with coronary artery disease.
dependent vasodilation in young adults with hypercholeste- Circulation 104:2673–2678
rolaemia. J Cardiovasc Risk 3:465–471
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

You might also like