Sie sind auf Seite 1von 6

Cellular Antioxidant Effects of Atorvastatin In Vitro and In Vivo

Sven Wassmann, Ulrich Laufs, Kirsten Mller, Christian Konkol, Katja Ahlbory, Anselm T. Bumer, Wolfgang Linz, Michael Bhm, Georg Nickenig
Abstract3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) may exert direct effects on vascular cells and beneficially influence endothelial dysfunction. Because reactive oxygen species (ROS) may lead to vascular damage and dysfunction, we investigated the effect of atorvastatin on ROS production and the underlying mechanisms in vitro and in vivo. Cultured rat aortic vascular smooth muscle cells were incubated with 10 mol/L atorvastatin. Angiotensin IIinduced and epidermal growth factorinduced ROS production were significantly reduced by atorvastatin (dichlorofluorescein fluorescence laser microscopy). Atorvastatin downregulated mRNA expression of the NAD(P)H oxidase subunit nox1, whereas p22phox mRNA expression was not significantly altered (reverse transcriptionpolymerase chain reaction, Northern analysis). Membrane translocation of rac1 GTPase, which is required for the activation of NAD(P)H oxidase, was inhibited by atorvastatin (Western blot). mRNA expression of superoxide dismutase isoforms and glutathione peroxidase was not modified by atorvastatin, whereas catalase expression was upregulated at mRNA and protein levels, resulting in an increased enzymatic activity. Effects of atorvastatin on ROS production and nox1, rac1, and catalase expression were inhibited by L-mevalonate but not by 25-hydroxycholesterol. In addition, spontaneously hypertensive rats were treated with atorvastatin for 30 days. ROS production in aortic segments was significantly reduced in statin-treated rats (lucigenin chemiluminescence). Treatment with atorvastatin reduced vascular mRNA expression of p22phox and nox1 and increased aortic catalase expression. mRNA expression of superoxide dismutases, glutathione peroxidase, and NAD(P)H oxidase subunits gp91phox, p40phox, p47phox, and p67phox remained unchanged. Translocation of rac1 from the cytosol to the cell membrane was also reduced in vivo. Thus, atorvastatin exerts cellular antioxidant effects in cultured rat vascular smooth muscle cells and in the vasculature of spontaneously hypertensive rats mediated by decreased expression of essential NAD(P)H oxidase subunits and by upregulation of catalase expression. These effects of atorvastatin may contribute to the vasoprotective effects of statins. (Arterioscler Thromb Vasc Biol. 2002;22:300-305.) Key Words: statins reactive oxygen species NAD(P)H oxidase spontaneously hypertensive rats vascular smooth muscle cells

s demonstrated by multiple interventional trials,1 4 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) effectively reduce the cardiovascular events of patients with coronary heart disease and hypercholesterolemia. The antiatherogenic potential of these drugs is not exclusively dependent on their lipid-lowering properties, which led to the proposition that statins may exert pleiotropic effects on vascular and cardiac cells independent of cholesterol synthesis.5,6 It has been shown that statins may induce apoptosis, inhibit cell cycle progression, reduce the proliferation of vascular smooth muscle cells (VSMCs),79 decrease inflammatory processes,10 12 increase the production of NO in endothelial cells by the upregulation of endothelial cell NO synthase (ecNOS) expression,13 reduce the production of endothelin-1,14 and beneficially influence

the coagulation processes.15 These effects are dependent on mevalonic acid (the product of the enzyme reaction inhibited by statins), which serves as precursor of numerous isoprenoid metabolites.16 The increased release and production of reactive oxygen species (ROS) is thought to be one of the key events in the pathogenesis of endothelial dysfunction and atherosclerosis.17 ROS elicit direct cellular damage and mitogenicity, serve as intracellular second messengers, and scavenge vasoprotective NO.18 The predominant source of ROS production in the vessel wall is the NAD(P)H oxidase system.19,20 The latter is composed of various subunits, such as rac1, p22phox, gp91phox, nox1 (formerly termed mox1), p40phox, p47phox, and p67phox.2124 Previous studies have suggested that the modulation of subunit expression is decisively important for

Received October 25, 2001; revision accepted November 14, 2001. From the Medizinische Klinik und PoliklinikInnere Medizin III (S.W., U.L., K.M., C.K., K.A., M.B., G.N.), Universittskliniken des Saarlandes, Homburg/Saar, Germany; Klinik III fr Innere Medizin (A.T.B.), Universitt zu Kln, Cologne, Germany; and Aventis Pharma Deutschland GmbH (W.L.), DG Cardiovascular Diseases, Frankfurt/Main, Germany. Correspondence to Dr Georg Nickenig, Medizinische Klinik und PoliklinikInnere Medizin III, Universittskliniken des Saarlandes, 66421 Homburg/Saar, Germany. E-mail nickenig@med-in.uni-saarland.de 2002 American Heart Association, Inc. Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org

300

Wassmann et al
the overall activity of NAD(P)H oxidase.24 26 Besides ROSgenerating enzymes, antioxidative defense systems are important for the oxidative stress that ultimately results. The superoxide dismutase (SOD) isoforms, glutathione peroxidase (GPX), and catalase (CAT) are enzymes residing within the vasculature that finally lead to the elimination of free radicals by the generation of water and oxygen.27,28 Because alterations of either the production or elimination of ROS are known to accelerate the progression of endothelial dysfunction and atherosclerosis, we hypothesized that statins could potentially interact with the gene products involved in these processes. Therefore, the influence of statins on ROS production and the expression of NAD(P)H oxidase subunits as well as radical-scavenging enzymes were investigated in cultured VSMCs and in spontaneously hypertensive rats (SHR).

Statins and ROS

301

Methods
The Methods section can be accessed online (please see http://atvb.ahajournals.org).

Results
Effect of Atorvastatin on Production of ROS in VSMCs
To evaluate the effect of atorvastatin on intracellular ROS production, VSMCs were preincubated for 12 hours with vehicle, 10 mol/L atorvastatin, atorvastatin plus L-mevalonate (200 mol/L), or atorvastatin plus 25-hydroxycholesterol (5 g/mL), followed by a 3-hour coincubation with 1 mol/L angiotensin II. ROS production was quantified by dichlorofluorescein fluorescence laser microscopy. Data analysis of 4 separate experiments is illustrated in Figure 1A. Stimulation with angiotensin II led to a marked increase of ROS production (240 21% of control, P 0.05 versus control). Preincubation with atorvastatin for 12 hours significantly reduced angiotensin IIinduced ROS production to 111 10% of control (P 0.05 versus angiotensin II). L-Mevalonate completely reversed the effect of atorvastatin on angiotensin IIinduced free radical production (246 17% of control, P 0.05 versus control), whereas 25-hydroxycholesterol had no effect (114 9% of control, P 0.05 versus angiotensin II). Incubation with atorvastatin, mevalonate, or hydroxycholesterol alone had no effect on basal ROS production (Figure 1C). Angiotensin IImediated ROS production was completely prevented by coincubation with diphenylene iodonium (DPI, 10 mol/L), an inhibitor of flavoprotein-containing oxidoreductases such as NAD(P)H oxidase (Figure 1C). In addition, VSMCs were preincubated for 12 hours with vehicle, 10 mol/L atorvastatin, atorvastatin plus L-mevalonate (200 mol/L), or atorvastatin plus 25-hydroxycholesterol (5 g/mL), followed by a 3-hour coincubation with 20 ng/mL epidermal growth factor (EGF). The EGFenhanced ROS production (236 20% of control, P 0.05 versus control) was also inhibited by preincubation with atorvastatin (95 8% of control, P 0.05 versus EGF), as shown in Figure 1B. Again, this was inhibited by L-mevalonate (233 33% of control, P 0.05 versus control), whereas hydroxycholesterol had no effect (101 11% of control, P 0.05 versus EGF).

Figure 1. Effect of atorvastatin (ator) on the production of ROS in VSMCs. A and B, VSMCs were preincubated for 12 hours with vehicle, ator (10 mol/L), ator plus L-mevalonate (meva, 200 mol/L), or ator plus 25-hydroxycholesterol (chol, 5 g/mL), followed by a 3-hour coincubation with either 1 mol/L angiotensin II (ang II, A) or 20 ng/mL EGF (B). C, In a set of control experiments, VSMCs were treated for 15 hours with vehicle (con), ator (10 mol/L), meva (200 mol/L), or chol (5 g/mL). In addition, VSMCs were incubated with ang II for 3 hours (1 mol/L), with ang II (3 hours) plus DPI (10 mol/L, 1 hour), or with DPI alone (1 hour). ROS production was quantied by dichlorouorescein uorescence laser microscopy. Values are mean SEM (n 4). *P 0.05 vs con; **P 0.05 vs ang II/EGF.

Effect of Atorvastatin on NAD(P)H Oxidase Subunit Expression in VSMCs


NAD(P)H oxidase is a multicomponent enzyme complex that is thought to be a major source of ROS in the vessel wall and can be activated through angiotensin II type 1 receptor activation by angiotensin II. To examine whether statin

treatment leads to an altered expression of essential subunits of the NAD(P)H oxidase in VSMCs, the cells were treated for 0 to 24 hours with 10 mol/L atorvastatin or vehicle, and total cellular RNA was extracted at the indicated time points. p22phox mRNA levels were assessed by Northern analysis. Figure 2A shows a representative autoradiograph, indicating that compared with the corresponding GAPDH mRNA levels, p22phox mRNA expression was not significantly altered by atorvastatin. In addition, bovine aortic endothelial cells (BAECs) were treated for 0 to 24 hours with 10 mol/L atorvastatin or vehicle; subsequently, Northern blot experiments were performed to assess p22phox mRNA expression in this cell type. A representative autoradiograph is shown in Figure 2B. The densitometric analysis of 3 separate experiments revealed that p22phox mRNA expression was slightly upregulated to 114 9% of control after 8 hours but was finally decreased to 72 5% of control after 24-hour incubation with atorvastatin (P 0.05 versus control). Nox1 mRNA expression in VSMCs after treatment with 10 mol/L atorvastatin for 0 to 24 hours was measured by semiquantitative reverse transcription (RT)polymerase chain reaction (PCR). Figure 2C shows a representative agarose gel of the amplified nox1 and GAPDH DNA frag-

302

Arterioscler Thromb Vasc Biol.

February 2002
cytosol to the cell membrane, we assessed rac1 protein expression in the membrane and cytosolic fraction of VSMCs treated for 12 and 24 hours with 10 mol/L atorvastatin or vehicle. As shown in representative immunoblots in Figure 2E, statin treatment led to a marked time-dependent decrease of membrane rac1 protein expression, whereas cytosolic rac1 expression (and total cell rac1 expression, data not shown) was increased. Furthermore, VSMCs were incubated for 24 hours with vehicle, atorvastatin (10 mol/L), L-mevalonate (200 mol/L), 25-hydroxycholesterol (5 g/mL), or geranylgeranyl pyrophosphate (GGPP, 10 mol/L) or with atorvastatin plus mevalonate, hydroxycholesterol, or GGPP. The effect of atorvastatin on rac1 membrane protein expression was inhibited by mevalonate and by GGPP but not by hydroxycholesterol, as shown in a representative immunoblot in Figure 2F. Finally, the effect of angiotensin II (1 mol/L, 3 hours) in the presence or absence of atorvastatin (10 mol/L, 15 hours) on rac1 protein expression in the membrane fraction of VSMCs was investigated. Treatment with angiotensin II enhanced rac1 expression, whereas coincubation with atorvastatin not only reversed this effect but further decreased rac1 expression below control levels, as demonstrated in Figure 2F.

Figure 2. Effect of ator on p22phox, nox1, and rac1 expression in VSMCs. VSMCs or BAECs were treated with 10 mol/L ator or vehicle (con), and total cellular RNA was extracted at the indicated time points. p22phox mRNA levels were assessed by Northern analysis, whereas nox1 mRNA expression was measured by semiquantitative RT-PCR. A and B, Autoradiographs (representative of 3 separate experiments) of p22phox mRNA and corresponding GAPDH mRNA in VSMCs (A) and of p22phox mRNA and corresponding ribosomal RNA in BAECs (B). C, Agarose gel (representative of 4 separate experiments) of the amplied nox1 and GAPDH DNA fragments in VSMCs. VSMCs were treated for 8 hours with con, ator (10 mol/L), meva (200 mol/L), or chol (5 g/mL), as indicated; RNA was isolated; and subsequent RT-PCR was performed. D, Agarose gels (representative of 4 separate experiments) of the amplied nox1 and GAPDH DNA fragments. Rac1 GTPase protein expression in VSMCs was assessed by Western blot experiments. E, Representative immunoblot of rac1 protein expression in the membrane and cytosolic fraction of VSMCs treated for 12 and 24 hours with 10 mol/L ator or con. F, Representative immunoblots of rac1 membrane protein expression of VSMCs treated for 24 hours with con, ator (10 mol/L), meva (200 mol/L), chol (5 g/mL), or GGPP (10 mol/L), as indicated, or with con, ang II (1 mol/L, 3 hours), or ang II (3 hours) plus ator (10 mol/L, 15 hours). Immunoblots are representative of 3 separate experiments.

Effect of Atorvastatin on Expression and Activity of Antioxidative Enzymes in VSMCs


As a counterpart to ROS-generating enzymes, several antioxidative enzymes are involved to preserve the balance of ROS availability within vascular cells. Decreased production of free radicals in VSMCs after statin treatment could also be mediated by increased expression of the radical-scavenging enzymes manganese SOD (mnSOD), extracellular SOD (ecSOD), copper-zinc SOD (czSOD), GPX, or CAT. Therefore, VSMCs were incubated for 0 to 24 hours with 10 mol/L atorvastatin or vehicle, total cellular RNA was extracted at the indicated time points, and Northern analysis was performed. As demonstrated in representative Northern blots in Figure 3A, statin treatment did not significantly alter the mRNA expression of mnSOD, ecSOD, GPX, czSOD, or GAPDH (representative of 3 separate experiments). In contrast, mRNA expression of CAT was significantly upregulated by atorvastatin, beginning after 4 hours of incubation (180 14% of control) and reaching a maximum of 227% 6% of control after 12 hours (densitometric analysis of 3 experiments, P 0.05 versus control). A representative Northern blot is shown in Figure 3B. In addition, VSMCs were treated for 8 hours with vehicle, atorvastatin (10 mol/L), L-mevalonate (200 mol/L), or 25-hydroxycholesterol (5 g/mL) or with atorvastatin plus mevalonate or hydroxycholesterol. As indicated in a representative autoradiograph in Figure 3C, the statin-mediated effect on CAT mRNA expression was reversed by mevalonate but not by hydroxycholesterol. The enhanced expression of CAT mRNA was translated to an increase of CAT protein expression (n 3) and CAT enzyme activity (4.1 1.0 [control] versus 6.7 0.6 [atorvastatin] U/mg protein, P 0.05 versus control; n 4) in VSMCs, as demonstrated in Figure 3D and 3E. Enzyme activity of the SODs and GPX was not altered by statin treatment (data not shown).

ments. Densitometric analysis showed that nox1 mRNA expression was decreased to 26 5% of control after 8 hours (P 0.05 versus control, n 4). Nox1 mRNA was not detected by Northern analysis (data not shown). In addition, VSMCs were treated for 8 hours with vehicle, atorvastatin (10 mol/L), L-mevalonate (200 mol/L), 25-hydroxycholesterol (5 g/mL), or a combination of these substances. Figure 2D shows representative agarose gels of the amplified nox1 and GAPDH PCR fragments. The inhibitory effect of atorvastatin on nox1 mRNA expression was completely reversed by mevalonate but not by hydroxycholesterol. Because it is known that the activation of the NAD(P)H oxidase requires translocation of the GTPase rac1 from the

Wassmann et al

Statins and ROS

303

Figure 4. Effect of ator on vascular ROS production in SHR. SHR received standard chow or standard chow supplemented with ator (50 mg/kg per day) for 30 days. Vascular production of ROS was assessed by lucigenin chemiluminescence assays (5 mol/L lucigenin) in isolated aortic segments of con- and statintreated SHR. RLU indicates relative light units. Values are mean SEM (n 6). *P 0.05 vs con.

Effect of Atorvastatin on Vascular Expression of NAD(P)H Oxidase Subunits and Antioxidative Enzymes in SHR
To examine the mechanisms underlying the decreased vascular ROS production in vivo, we assessed the expression of NAD(P)H oxidase subunits and antioxidative enzymes in RNA of aortic homogenates of control and statin-treated SHR by means of semiquantitative RT-PCR (n 3 per group). Densitometric analysis demonstrated that the expression of nox1 mRNA (53 14% of control, P 0.05 versus control) and p22phox mRNA (57 9% of control, P 0.05 versus control) was reduced in the aortas of SHR treated with the statin, whereas GAPDH expression remained unchanged. Other subunits of the NAD(P)H oxidase, namely, gp91phox, p40phox, p47phox, and p67phox, were detected in aortic tissue of SHR, but there was no significant effect of atorvastatin on the mRNA expression levels of these genes, as shown in Figure 5A. In agreement with the in vitro findings, rac1 GTPase protein expression was decreased in the membrane fraction of aortic homogenates of statin-treated SHR, whereas cytosolic rac1 protein expression was increased (n 3 per group), as demonstrated in a representative immunoblot in Figure 5B. Finally, treatment with atorvastatin did not exert a significant effect on the vascular mRNA expression of the antioxidative enzymes mnSOD, ecSOD, GPX, and czSOD in vivo. However, consistent with the findings in VSMCs, vascular CAT mRNA expression was increased in the aortas of SHR treated with atorvastatin (157 15% of control, P 0.05 versus control; n 3 per group), as shown in Figure 5C.

Figure 3. Effect of ator on expression of GPX and SODs and on CAT expression and activity in VSMCs. A and B, mnSOD, ecSOD, czSOD, GPX, CAT, and the corresponding GAPDH mRNA expression, as assessed by Northern analysis in VSMCs incubated with 10 mol/L ator or vehicle (con) for 0 to 24 hours. C, Representative Northern blot of VSMCs treated for 8 hours with con, ator (10 mol/L), meva (200 mol/L), or chol (5 g/mL), as indicated. D, Representative immunoblot of CAT protein expression in VSMCs incubated with either con or 10 mol/L ator. Autoradiographs are representative of 3 separate experiments. CAT enzyme activity was assessed by spectrophotometric quantication of H2O2 conversion in proteins of VSMCs incubated with either con or 10 mol/L ator. Values are mean SEM (n 4). *P 0.05 vs con.

Effect of Atorvastatin on Vascular Production of ROS in SHR


To investigate whether decreased free radical production in vitro can also be observed in vivo, 18-week-old SHR received standard chow or standard chow supplemented with atorvastatin for 30 days. After treatment, vascular production of ROS was assessed by lucigenin chemiluminescence assays in isolated aortic segments of control and statin-treated SHR. Figure 4 illustrates that treatment with atorvastatin induced a significant decrease of superoxide production in the vessel wall (11.6 1.7 [control] versus 7.2 1.2 [atorvastatin] relative light units per milligram aortic tissue, P 0.05 versus control; n 6 per group). In a set of control experiments, preincubation of the vessel segments with SOD (150 U/mL) or DPI (10 mol/L) decreased the superoxide-mediated lucigenin chemiluminescence signals to background levels (data not shown).

Discussion
ROS such as superoxide and hydrogen peroxide are involved in the pathogenesis of atherosclerosis.17,18 This is based on a variety of effects exerted by ROS on vascular cells. Free radicals induce VSMC mitogenicity, endothelial dysfunction, endothelial cell apoptosis, and the adhesion of macrophages to endothelial cells; they also serve as intracellular signaling molecules and scavenge vasoprotective NO.17,18,29 One of the predominant sources of ROS is NAD(P)H oxidase, a multicomponent enzyme system residing in white blood cells and in VSMCs and endothelial cells, the composition of which is dependent on the harboring cell type.19 24 Whereas, for example, gp91phox is expressed in endothelial cells and macrophages, nox1 substitutes gp91phox in VSMCs.23,24 Previous reports have suggested that the interaction of nox1 with other subunits is essential for the overall activity of

304

Arterioscler Thromb Vasc Biol.

February 2002
statin-induced cellular depletion of intermediates of cholesterol biosynthesis can impair the action of rac1.32,33 Our data indicate that stimulation with angiotensin II leads to the enhancement of rac1 expression in the cell membrane, indicating that rac1 membrane translocation may be an important step for angiotensin IIinduced ROS production in VSMCs. In contrast, statins decrease the geranylgeranylation-dependent translocation of rac1 from the cytosol to the cell membrane in the presence or absence of angiotensin II in these cells. This observation is extended to the in vivo situation, inasmuch as long-term statin treatment of SHR leads to a similar impairment of rac1 membrane translocation in the vessel wall. In this animal model, the expression levels of 2 other essential components of the NAD(P)H oxidase, namely, p22phox and nox1, were decreased by atorvastatin. These findings indicate that NAD(P)H oxidase is inhibited at various strategic points in vivo and suggest that the antioxidant properties of statins are mediated by the interaction with different genes engaged in the production of ROS in the vessel wall. Nox1 expression was decreased in VSMCs treated with atorvastatin; this finding is consistent with the findings in SHR. Expression of p22phox was not affected in VSMCs. However, in cultured aortic endothelial cells, statin treatment led to a decreased mRNA expression of p22phox. This statin effect on endothelial cells may contribute to the decreased p22phox expression found in SHR aortas. Because the inhibitory effect of atorvastatin in endothelial cells is only moderate, the overall effect of the statin on p22phox expression in SHR cannot be completely explained by the in vitro findings in VSMCs and endothelial cells, indicating that in vivo other cofactors may participate in this statin effect. The ultimate oxidative stress within vascular cells is determined by ROS production and corresponding elimination processes.27,28 The latter are realized by the radicalscavenging enzymes GPX, the SOD isoforms, and CAT. Whereas atorvastatin had no influence on the expression of GPX and SODs, CAT expression and activity were profoundly upregulated in vitro and in vivo. Physiologically, the upregulation of CAT can be observed after an increase of hydrogen peroxide concentrations. However, reduced superoxide production by NAD(P)H oxidase after decreased expression of essential subunits leads to reduced concentrations of hydrogen peroxide when SOD levels are not altered.24,26 Because CAT is used in the elimination of hydrogen peroxide,27,28 increased levels of CAT further reduce the concentrations of this radical, thereby accelerating the turnover of superoxide to hydrogen peroxide. Finally, this leads to a decrease of the overall intracellular free radical load in VSMCs. Therefore, upregulation of CAT may represent another antioxidative action of statins. The effects of atorvastatin on free radical production, nox1 and CAT expression, and membrane translocation of rac1 GTPase could be reversed by mevalonate but not by hydroxycholesterol, indicating that the statin effect is dependent on HMG-CoA reductase inhibition but not on reduced cholesterol synthesis and is, therefore, dependent on impaired isoprenylation processes. It has been shown that statins upregulate ecNOS expression in endothelial cells.13 In VSMCs, ecNOS cannot be found, whereas the inducible isoform of NO synthase (iNOS) is expressed. iNOS produces only relevant concentrations of

Figure 5. Effect of ator on vascular expression of NAD(P)H oxidase subunits and antioxidative enzymes in SHR. A, mRNA of aortic homogenates of control (con)- and statin-treated (ator) SHR was isolated and analyzed by semiquantitative RT-PCR with respect to nox1, p22phox, gp91phox, p40phox, p47phox, p67phox, and GAPDH mRNA expression. Values are mean SEM (densitometric analysis, n 3). *P 0.05 vs con. B, Rac1 GTPase protein expression was assessed by Western blots in membrane and cytosolic fractions of proteins isolated from aortic homogenates of con- or ator-treated SHR. Immunoblot is representative of 3 separate experiments. C, Vascular mRNA expression of mnSOD, ecSOD, czSOD, GPX, CAT, and GAPDH was assessed by semiquantitative RT-PCR in aortic tissue isolated from SHR treated with vehicle (con) or ator. Values are mean SEM (densitometric analysis, n 3). *P 0.05 vs con.

NAD(P)H oxidase in VSMCs.23,24 In analogy, modulation of p22phox expression is also closely connected with NAD(P)H oxidase activity.25,26 Activation and translocation of rac1 GTPase from the cytosolic compartment to the cell membrane is another prerequisite of NAD(P)H oxidase activation, as shown in white blood cells.30,31 The role of other components of NAD(P)H oxidase, such as p40phox, p47phox, and p67phox, is less clear; however, the assembly of all subunits is required for superoxide production by this enzyme.21,22 Given the importance of ROS production for atherosclerosis and the predominant role of NAD(P)H oxidase for free radical load in the vasculature, it is important to determine whether therapeutic interventions other than the blockade of the reninangiotensin system may influence this multicomponent enzyme. Statins are drugs that are widely and successfully used in cardiovascular diseases; besides their lipid-lowering properties, these drugs demonstrate a variety of pleiotropic effects on vascular cells independent of plasma cholesterol levels.5,6 In endothelial cells and macrophages, it has been shown that

Wassmann et al
NO after induction by, for example, proinflammatory cytokines or lipopolysaccharides. In unstimulated VSMCs, as used in the present study, relevant levels of NO are not expected. Previous studies have demonstrated that statins may lead to an inhibition of iNOS induction and expression in blood vessels.34,35 Therefore, it seems unlikely that incubation of VSMCs with statins leads to increased NO levels, which could account for the decreased free radical load of VSMCs after statin treatment. The aforementioned molecular effects of statins on rac1, p22phox, nox1, and CAT lead to reduced production of ROS in vitro and in vivo. It may be assumed that these mechanisms of action may participate in the well-established beneficial effects of statins on endothelial function,36,37 inasmuch as a reduction of free radical load in the vasculature may lead to improved endothelium-dependent vasorelaxation and to reduced adhesion of macrophages to the vessel wall.17,18 Because ROS are involved in the pathogenesis of atherosclerosis,17,18,29 it may be speculated that reduced production and increased elimination of ROS within the vessel wall may resemble a pleiotropic cholesterol-independent effect of statins that may contribute to the atheroprotective properties of these drugs.

Statins and ROS

305

Acknowledgments
This work was supported by the Deutsche Forschungsgemeinschaft and a Gdecke research grant. The excellent technical assistance of Katja Rsler and Sybille Karl is greatly appreciated.

References
1. Levine GN, Keaney JF, Vita JA. Cholesterol reduction in cardiovascular disease. N Engl J Med. 1995;332:512521. 2. Scandinavian Simvastatin Survival Study Group. Randomized trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet. 1994;344:13831389. 3. Shepherd J, Cobbe SM, Ford I, Isles CG, Lorimer AR, McFarlane PW, McKillop JH, Packard CJ. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia: West of Scotland Coronary Prevention Study Group. N Engl J Med. 1995;333:13011307. 4. West of Scotland Coronary Prevention Study Group. Influence of pravastatin and plasma lipids on clinical events in the West of Scotland Coronary Prevention Study (WOSCOPS). Circulation. 1998;97:1440 1445. 5. Vaughan CJ, Murphy MB, Buckley BM. Statins do more than just lower cholesterol. Lancet. 1996;348:10791082. 6. Lefer AM, Scalia R, Lefer DJ. Vascular effects of HMG CoA reductase inhibitors (statins) unrelated to cholesterol lowering: new concepts for cardiovascular disease. Cardiovasc Res. 2001;49:281287. 7. Guijarro C, Blanco-Colio LM, Ortego M, Alonso C, Ortiz A, Plaza JJ, Diaz C, Hernandez G, Egido J. 3-Hydroxy-3-methylglutaryl coenzyme A reductase and isoprenylation inhibitors induce apoptosis of vascular smooth muscle cells in culture. Circ Res. 1998;83:490500. 8. Laufs U, Marra D, Node K, Liao JK. 3-Hydroxy-3-methylglutaryl-CoA reductase inhibitors attenuate vascular smooth muscle proliferation by preventing rho GTPase-induced down-regulation of p27(Kip1). J Biol Chem. 1999;274:2192621931. 9. Negre-Aminou P, van Vliet AK, van Erck M, van Thiel GCF, van Leeuwen REW, Cohen LH. Inhibition of proliferation of human smooth muscle cells by various HMG-CoA reductase inhibitors; comparison with other human cell types. Biochim Biophys Acta. 1997;1345:259 268. 10. Bustos C, Hernandez-Presa MA, Ortega M, Tunon J, Ortega L, Perez F, Diaz C, Hernandez G, Egido J. HMG-CoA reductase inhibition by atorvastatin reduces neointimal inflammation in a rabbit model of atherosclerosis. J Am Coll Cardiol. 1998;32:20572064. 11. Kothe H, Dalhoff K, Rupp J, Muller A, Kreuzer J, Maass M, Katus HA. Hydroxymethylglutaryl coenzyme: a reductase inhibitors modify the inflammatory response of human macrophages and endothelial cells infected with Chlamydia pneumoniae. Circulation. 2000;101:1760 1763. 12. Takeuchi S, Kawashima S, Rikitake Y, Ueyama T, Inoue N, Hirata K, Yokoyama M. Cerivastatin suppresses lipopolysaccharide-induced ICAM-1 expression through inhibition of Rho GTPase in BAEC. Biochem Biophys Res Commun. 2000;269:97102.

13. Laufs U, La Fata V, Plutzky J, Liao JK. Upregulation of endothelial nitric oxide synthase by HMG-CoA reductase inhibitors. Circulation. 1998;97: 11291135. 14. Hernandez-Perera O, Perez-Sala D, Navarro-Antolin J, Sanchez-Pascuala R, Hernandez G, Diaz C, Lamas S. Effects of the 3-hydroxy-3methylglutaryl-CoA reductase inhibitors, atorvastatin and simvastatin, on the expression of endothelin-1 and endothelial nitric oxide synthase in vascular endothelial cells. J Clin Invest. 1998;101:27112719. 15. Essig M, Nguyen G, Prie D, Escoubet B, Sraer JD, Friedlander G. 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors increase fibrinolytic activity in rat aortic endothelial cells: role of geranylgeranylation and rho proteins. Circ Res. 1998;83:683690. 16. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343:425 430. 17. Harrison DG. Endothelial function and oxidant stress. Clin Cardiol. 1997; 20(suppl):II-11II-17. 18. Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res. 2000;87:840 844. 19. Griendling KK, Minieri CA, Ollerenshaw JD, Alexander RW. Angiotensin II stimulates NADH and NADPH oxidase activity in cultured vascular smooth muscle cells. Circ Res. 1994;74:11411148. 20. Rajagopalan S, Kurz S, Muenzel T, Tarpey M, Freeman BA, Griendling KK, Harrison DG. Angiotensin II-mediated hypertension in the rat increases vascular superoxide production via membrane NADH/NADPH oxidase activation: contribution to alterations of vasomotor tone. J Clin Invest. 1996;97: 19161923. 21. Babior BM. NADPH oxidase: an update. Blood. 1999;93:14641476. 22. Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res. 2000;86:494501. 23. Suh YA, Arnold RS, Lassegue B, Shi J, Xu X, Sorescu D, Chung AB, Griendling KK, Lambeth JD. Cell transformation by the superoxidegenerating oxidase mox1. Nature. 1999;401:79 82. 24. Lassgue B, Sorescu D, Szcs K, Yin Q, Akers M, Zhang Y, Grant SL, Lambeth JD, Griendling KK. Novel gp91phox homologues in vascular smooth muscle cells: nox1 mediates angiotensin IIinduced superoxide formation and redox-sensitive signaling pathways. Circ Res. 2001;88:888 894. 25. Fukui T, Ishizaka N, Rajagopalan S, Laursen JB, Capers Q IV, Taylor WR, Harrison DG, de Leon H, Wilcox JN, Griendling KK. p22phox mRNA expression and NADPH oxidase activity are increased in aortas from hypertensive rats. Circ Res. 1997;80:4571. 26. Ushio-Fukai M, Zafari AM, Fukui T, Ishizaka N, Griendling KK. p22phox is a critical component of the superoxide-generating NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in vascular smooth muscle cells. J Biol Chem. 1996;271:2331723321. 27. Gutteridge JM, Halliwell B. Free radicals and antioxidants in the year 2000: a historical look to the future. Ann N Y Acad Sci. 2000;899:136147. 28. Andreoli TE. Free radicals and oxidative stress. Am J Med. 2000;108: 650651. 29. Darley-Usmar VM, McAndrew J, Patel R, Moellering D, Lincoln TM, Jo H, Cornwell T, Digerness S, White CR. Nitric oxide, free radicals and cell signalling in cardiovascular disease. Biochem Soc Trans. 1997;25:925929. 30. Dusi S, Domini M, Rossi F. Mechanisms of NADPH oxidase activation in human neutrophils: p67phox is required for the translocation of rac1 but not rac2 from cytosol to the membranes. Biochem J. 1995;308:991994. 31. Rinckel LA, Faris SL, Hitt ND, Kleinberg ME. Rac1 disrupts p67phox/ p40phox binding: a novel role for rac in NADPH oxidase activation. Biochem Biophys Res Commun. 1999;263:118122. 32. Wagner AH, Kohler T, Rckschloss U, Just I, Hecker M. Improvement of nitric oxide-dependent vasodilatation by HMG-CoA reductase inhibitors through attenuation of endothelial superoxide anion formation. Arterioscler Thromb Vasc Biol. 2000;20:6166. 33. Bockoch GM, Prossnitz V. Isoprenoid metabolism is required for stimulation of the respiratory burst oxidase of HL-60 cells. J Clin Invest. 1992;89: 402408. 34. Vaughan CJ, Delanty N. Neuroprotective properties of statins in cerebral ischemia and stroke. Stroke. 1999;30:19691973. 35. Park JK, Muller DN, Mervaala EM, Dechend R, Fiebeler A, Schmidt F, Bieringer M, Schafer O, Lindschau C, Schneider W, et al. Cerivastatin prevents angiotensin II-induced renal injury independent of blood pressureand cholesterol-lowering effects. Kidney Int. 2000;58:14201430. 36. ODriscoll G, Green D, Taylor RR. Simvastatin, a HMG-coenzyme: a reductase inhibitor, improves endothelial function within 1 month. Circulation. 1997;95:11261131. 37. Tsunekawa T, Hayashi T, Kano H, Sumi D, Matsui-Hirai H, Thakur NK, Egashira K, Iguchi A. Cerivastatin, a hydroxymethylglutaryl coenzyme A reductase inhibitor, improves endothelial function in elderly diabetic patients within 3 days. Circulation. 2001;104:376379.

Das könnte Ihnen auch gefallen