AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Review Article
Volume 51, Issue 3, Pages 120-126
Shin-Yuan Fang 1.2 , Jun-Neng Roan 2.3 , Chwan-Yau Luo 2 , Yu-Chuan Tsai 1 , Chen-Fuh Lam 1.4
2888 Views


Abstract

3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor (statins) is one of the most commonly prescribed agents for controlling hyperlipidemia. Apart from their lipid-lowering property, statins are well known for their pleiotropic effects, such as improvement of vascular endothelial dysfunction, attenuation of inflammatory responses, stabilization of atherosclerotic plaques, inhibition of vascular smooth muscle proliferation, and modulation of procoagulant activity and platelet function. The vasculoprotective effect of statins is mainly mediated by inhibition of the mevalonate pathway and oxidized low-density lipoprotein generation, thereby enhancing the biosynthesis of endothelium-derived nitric oxide. Accumulating clinical evidence strongly suggests that administration of statins reduces overall mortality, the development myocardial infarction and atrial fibrillation, and length of hospital stay after a major cardiac/noncardiac surgery. This review updates the clinical pharmacology and therapeutic applications of statins during major operations, and highlights the anesthesia considerations for perioperative statin therapy.

Keywords

endothelium; hydroxymethylglutaryl-CoA reductase inhibitors; mevalonic acid: mevalonate; nitric oxide; terpenes: isoprenoids;


1. Introduction

The biosynthesis of cholesterol mainly occurs in the liver and intestine by condensation of acetyl-CoA and acetoacetyl-CoA in the cytoplasm and endoplasmic reticulum. The formation of the precursor compound, hydroxymethylglutaryl-coenzyme A (HMG-CoA), is catalyzed by HMG-CoA reductase to form mevalonate (Fig. 1).12 Following a series of catalyzing reactions, mevalonate is converted into isoprenoid derivatives to finally yield squalene and cholesterol. In this process, HMG-CoA reductase is the rate-limiting enzyme, and therefore, a number of drugs were developed by inhibiting the action of HMG-CoA reductase in order to reduce the generation of cholesterol.2 Statins are the pharmaceutical agents that can competitively inhibit the enzymatic activity of HMG-CoA reductase and thus effectively reduce the blood levels of cholesterol (Fig. 1).3 Table 1 lists the HMG-CoA reductase inhibitors (statins) that are currently available in Taiwan. Hypercholesterolemia is a major risk factor for the development of cardiovascular diseases, coronary artery disease, and stroke secondary to the prothrombotic and atherosclerotic effects. On average, administrations of statins reduce serum concentrations of total cholesterol and low-density lipoprotein cholesterol (LDL-C) by 17–35% and 24–49%, respectively.4Furthermore, levels of triglyceride are reduced by 13% and high-density lipoprotein are increased by 5% following treatment with statins.5 Instead of discussing the well-known lipid-lowering effect of statins, this brief review focuses mainly on the pleiotropic vascular protection and their applications in perioperative medicine.

Fig. 1.
Download full-size image
Fig. 1. Biosynthesis of cholesterol through the mevalonate pathway. Generation of mevalonate and the isoprenoid intermediates, particularly geranylgeranyl pyrophosphate (GPP) inactivates biological function of endothelial nitric oxide synthase (eNOS). Administration of statins suppresses the mevalonate pathway and also directly enhances the enzymatic activity of eNOS. DPP = dimethylallyl pyrophosphate; FPP = farmesyl pyrophosphate; HMG-CoA = 3-hydroxy-3-methylglutaryl-coenzyme A; IPP = isopentenyl pyrophosphate.

2. Mevalonate pathway and endothelial function

The synthesis of cholesterol generates mevalonate and other intermediate compounds, such as isopentenyl pyrophosphates (Fig. 1). In cultured endothelial cells, the upregulation of endothelial nitric oxide synthase (eNOS) and biosynthesis of NO following incubation with HMG-CoA inhibitors were completely reversed in the presence of L-mevalonate,678 probably by inhibition of the Akt-mediated eNOS phosphorylation.9 The formation of isoprenoid derivatives (or the so-called isoprenes), particularly geranylgeranyl pyrophosphate (GGPP), prenylates the activity of Rho guanosine triphosphatase (GTPase) and activates the small GTP-binding protein Rho by post-translational modification (Fig. 1).10 Geranylgeranylation of Rho proteins destabilizes the transcription of eNOS and inhibits its activation.10 Activation of Rho A/Rho kinase pathway downregulates eNOS expression and enzymatic activity on the vascular endothelium (Figs. 1 and 2).

Fig. 2.
Download full-size image
Fig. 2. Diagrammatic illustration of the pleiotropic protective effect of statins on the biological function of endothelial nitric oxide synthase (eNOS) in endothelial cells. Excessive superoxide anions (O2–) are generated by the induction of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in the oxidative physiological status. Low-density lipoprotein (LDL) is oxidized by O2– to form oxidized LDL (oxLDL), which in turn decreases the biosynthesis of NO, thereby attenuating the formation of second messenger cyclic guanosine monophosphate (cGMP) in the adjacent vascular smooth muscle cells (VSMCs). Reduced intracellular concentration of cGMP in the VSMCs impairs the arterial relaxation response.

3. Oxidized LDL-C and endothelial function

Arterial expression of oxidized LDL receptor (LOX-1) is upregulated by enhanced angiotensin II, endothelin, cytokines, and nonuniform shear stress in patients with hypertension, dyslipidemia, and diabetes mellitus.11 LDL-C is oxidized by the overproduction of superoxide anions in these highly oxidized states and is internalized by the activation of LOX-1 on the membrane of endothelial cells (Fig. 2).12 Oxidized LDL dephosphorylates Akt impairing the enzymatic activity of eNOS and inhibiting the migration of endothelial cells.13Other previous studies also demonstrated that oxidized LDL displaces eNOS protein with caveolae14 and Hsp90.15 The enzymatic activity is reduced in the uncoupled eNOS, and it generates more superoxide anions rather than NO.15By contrast, oxidized LDL activates the renin–angiotensin system through LOX-1, and accelerates atherogenesis and endothelial dysfunction.1617Oxidation of LDL-C also stimulates the expression of preproendothelin messenger RNA (mRNA) in the endothelial cells, leading to a time- and concentration-dependent release of the endothelin.18 Increase of endothelin causes arterial vasospasm, promotes vascular smooth muscle proliferation and progression of atherosclerosis.1819

4. Pleiotropic effect of statins on vascular function

Competitive inhibition of HMG-CoA reductase by administration of statins effectively reduced the production of mevalonate and isoprenoids. Treatment with cerivastatin and rosuvastatin significantly enhanced NO bioavailability and upregulated eNOS expression in cultured endothelial cells by blocking mevalonate and isoprenoid synthesis.2021 Administration of statins inhibited the formation of superoxide anions by preventing the isoprenylation of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and thus improved vascular endothelial function in isolated rat aorta.22 In diabetic animals, atorvastatin promoted vascular endothelial function by inhibiting the action of Rac-1, and this protective effect was significantly abolished by the addition of GGPP, indicating that the reduction in isoprenoid formation is an important molecular mechanism responsible for the vascular antioxidant effect of atorvastatin in patients with diabetes mellitus.23 Nishimatsu et al24 demonstrated that fluvastatin potentiated the endothelium-dependent relaxation response in obese Zucker rats by restoring eNOS expression, enhancing Akt phosphorylation, and inhibiting Rho kinase activity in the arterial system.

In addition to reducing serum levels of LDL-C, sustained evidence supports the direct effect of statins in the suppression of LOX-1 expression2526 and LDL-mediated oxidative stress272829 in the cardiovascular system of animals and humans. We have recently reported that prolonged administration of rosuvastatin suppressed the expression of NADPH oxidase, reduced superoxide generation, attenuated proinflammatory reactions, and thereby enhanced blood flow in the arteriovenous fistula of rats with streptozotocin-induced diabetes.3031 Furthermore, simvastatin relaxed the tonic contraction response induced by endothelin-1 in a concentration-dependent manner and inhibited the endothelin-1-mediated DNA synthesis of Rho proteins.32

Statin therapy increases mobilization and differentiation of bone marrow-derived endothelial progenitor cells through the PI3K/Akt pathway.33 In our laboratory, we also found that treatment with rosuvastatin increased the number of circulating endothelial progenitor cells and improved vascular endothelial function in diabetic rats.30 Augmentation in the numbers and function of endothelial progenitor cells provides therapeutic and preventive beneficial effects in a wide range of cardiovascular diseases.34

Collectively, statins may improve cardiovascular outcomes of patients with normal plasma cholesterol level, suggesting that the pleiotropic vascular protective effect of statins is independent from their lipid-lowering property. The improvement in cardiovascular outcomes is well correlated with improved endothelial function by upregulating eNOS expression and NO bioavailability following treatment with statins. At subcellular levels, statins stabilize the eNOS mRNA, enhance eNOS enzymatic activity through the PI3K/Akt signaling pathway, reduce inflammatory responses in the vasculature, inhibit Rho isoprenylation, and suppress oxidized LDL-induced endothelin-1 expression (Figs. 1 and 2). Readers are referred to several precise review articles for more detailed mechanistic discussion on the pleiotropic vascular protective effect of statins.3536

5. Cardiovascular outcomes of perioperative statin use

5.1. Outcomes in cardiac surgery

The first prospective randomized study reported in 1999 demonstrated that a 4-week treatment with simvastatin (20 mg/day) significantly improved the events of postoperative thrombocytosis, myocardial infarction, and renal insufficiency in hypercholesterolemic patients after coronary artery bypass grafting (CABG).37 A most recent comprehensive meta-analysis summarized 54 observational and randomized controlled trials (inclusion of 91,491 patients) to identify the effects of preoperative statin therapy on the major adverse outcomes after cardiac surgery.38 Their analysis suggested that preoperative statin therapy resulted in 31% odds reduction for early all-cause mortality [odds ratio (OR) = 0.69; 95% confidence interval (CI) = 0.59–0.81] and a substantial reduction in postoperative atrial fibrillation (OR = 0.71; 95% CI = 0.61–0.82). Administration of statin also significantly reduced the incidence of postoperative stroke and period of intensive care unit (ICU) stay. The beneficial effects of preoperative use of statins in patients receiving valvular heart surgery are less conclusive. A retrospective study analyzed the outcomes of patients undergoing valve operation.39 This cohort study showed that the odds for the composite end points of death, stroke, and renal failure were lower in patients taking statins (unadjusted OR = 1.90; 95% CI = 0.95–3.76; p = 0.068). However, the patients included in the statin group had higher comorbidities (i.e., stroke, diabetes, cerebrovascular disease, and dyslipidemia). Therefore, prospective randomized controlled trials are warranted to further elucidate the actual effects of perioperative statin use in valvular heart surgery.40

5.2. Outcomes in vascular surgery

Poldermans et al41 conducted an important case-controlled study to determine the effect of statin on the survival outcome among 2816 patients who underwent major vascular surgery. Their results provided the first evidence demonstrating that statin therapy significantly reduced the mortality (8% vs. 25%) during the perioperative phase with an adjusted odds ratio of 0.22 (95% CI = 0.10–0.47). These findings were supported by a retrospective study, which included 570 patients who underwent elective open infrarenal abdominal aortic aneurysm surgery.42 The incidence of perioperative mortality and myocardial infarction within 30 days of surgery was lower in statin users (3.7% vs. 11.0%; OR = 0.31; 95% CI = 0.13–0.74; p = 0.01). In symptomatic patients admitted for carotid endarterectomy, administration of statins was associated with reduced in-hospital mortality and in-hospital ischemic stroke or death (adjusted OR = 0.25; 95% CI = 0.07 to 0.90–0.55; 95% CI = 0.32–0.95, respectively).43 The most solid evidence of cardiovascular-protective effect of statins in noncardiac vascular surgery came from the landmark randomized controlled trial reported by the Dutch Echocardiographic Cardiac Risk Evaluation Applying Stress Echocardiography Study Group in 2009.44 Incidence of postoperative myocardial ischemia was reduced in patients treated with fluvastatin [10.8% vs. 19.0%; hazard ratio (HR) = 0.55; 95% CI = 0.34–0.88; p = 0.01]. Cardiovascular death or myocardial infarction occurred in 12 patients (4.8%) in the fluvastatin group and in 25 patients (10.1%) in the placebo group (HR = 0.47; 95% CI = 0.24–0.94; p = 0.03).

Most recently, a large-scale systemic meta-analysis study assessed the available randomized controlled trials reporting the effect of perioperative statins in statin-naïve patients undergoing cardiac/noncardiac surgery.45 Fifteen randomized controlled trials involving 2292 patients were included in this analysis. Postoperative development of atrial fibrillation was decreased in the statin-treatment group [relative risk (RR) = 0.56; 95% CI = 0.45–0.69; number needed to treat (NNT) = 6]. In cardiac and noncardiac surgeries, perioperative administration of statin reduced the risk of myocardial infarction (RR = 0.53; 95% CI = 0.38–0.74; NNT = 23) and the mean length of hospital stay (standardized mean difference = −0.32; 95% CI = −0.53 to −0.11). Statin use also tended to reduce the risk of death (RR = 0.62; 95% CI = 0.34–1.14) and length of ICU stay (standardized mean difference = −0.08; 95% CI = −0.25–0.10). The key results of this important meta-analysis are listed in Table 2. Furthermore, the authors also carried out a sensitivity analysis by excluding cardiac surgery (11 of 15 eligible studies) and found that statin treatment continued to demonstrate reduction of risk of myocardial infarction in patients who underwent noncardiac surgery (Table 2).

6. Anesthesia consideration of perioperative statin use

6.1. Withdrawal of perioperative statin therapy

The National Registry of Myocardial Infarction recommends that statin therapy within 24 hours of hospitalization for acute myocardial infarction is associated with reduced early complications and in-hospital death, and discontinuation of statin therapy is associated with increased mortality.46 Similar findings were found in a prospective cohort study comparing the continuation and discontinuation of statin therapy during the perioperative period in patients undergoing infrarenal aortic surgery.47 Withdrawal of statin therapy after the operation (>4 days) significantly increased the risk of postoperative myocardial necrosis (OR = 2.9; 95% CI = 1.6–5.5). Therefore, the American College of Cardiology/American Heart Association (ACC/AHA) strongly recommends that patients who are currently under statin therapy should continue taking statins during the perioperative period of noncardiac and vascular surgery.48 Likewise, expert opinions also suggest that early resumption of statins should be exercised after all major operations.49 The adverse responses secondary to acute statin withdrawal may be caused by overshoot translocation and activation of Rho, thereby downregulating eNOS enzymatic activity and reducing baseline production of NO.50

7. ACC/AHA recommendations

The ACC/AHA Joint Task Force published guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery in 2007, and has made clear recommendations for statin use in patients undergoing noncardiac surgery (Table 3).48 This clinical practice guideline also suggests that consideration should be given to starting statin therapy in surgical patients who meet the National Cholesterol Education Program criteria.

8. Doses and regimens of perioperative statins

Based on current clinical data, the hydrophilic rosuvastatin is considered as the most potent statin for the lipid-lowering and pleiotropic effects, followed by atorvastatin.51 Extended-release statin formulas are preferable during the perioperative period to cover the first 1–2 days after the procedure when oral intake may not be feasible.52 Individual studies also indicated that higher doses of statin use seem to provide better clinical outcomes in patients with coronary artery disease or after CABG surgery.5354 However, the cost effectiveness of increased therapeutic dose of perioperative statins should be justified individually according to the health of patients and types of statins.40 Although expert opinions suggest that 4–6 weeks of preoperative statin therapy is desired,55 the appropriate timing for commencing statin treatment prior to surgery remains inconclusive.52 It is recommended that perioperative statin use should be started early with an appropriate high therapeutic dose and continued for the long term after surgical procedures.55

9. Adverse effects of statin therapy

Based on the large clinical trials conducted, administration of statin appears to be safe, except for a small excess risk of hepatic transaminase elevations.56 However, anesthesiologists should be aware of the fact that the most common adverse events related to statin therapy are myopathy/rhabdomyolysis and elevated liver enzymes, with incidences of 0.04–0.07% and 1.18%, respectively.57 Patients with advanced age (>80 years), small body frame, coexisting with chronic diseases (such as chronic renal failure and severe liver impairment), multiple medications, and chronic alcoholism are at an increased risk for the development of statin-related adverse effects.58

10. Perioperative drug–drug interactions with statins

Statins are metabolized by the cytochrome P450 (mainly by CYP3A4 and CYP2C9; Table 1). Therefore, they interact with a wide array of medicines or compounds at the pharmacokinetic level, particularly during drug transformation in the liver.3Concurrent administration of statins with CYP450 inhibitors may thus elevate their concentration in blood and increase the risk of toxicity. The most common concern about perioperative use of statins is the co-administration with succinylcholine, which could be deleterious in causing muscle injury. Turan et al59 included 70 patients who were under chronic statin therapy (>3 months) and delivered succinylcholine (1.5 mg/kg) for general anesthesia in 38 patients. Their results showed that plasma concentrations of myoglobin were higher and fasciculations in statin users were more intense than that in nonusers. However, plasma potassium and creatine kinase concentrations were similar in statin users and nonusers, as was muscle pain. Therefore, the authors concluded that the effect of succinylcholine given to patients taking statins is likely to be small and probably of limited clinical consequence.59 In clinical practice, adverse responses due to perioperative drug interaction with statins are probably overlooked, but one should be aware that statin-induced muscle and hepatic toxicity could be amplified in high-risk patients.

11. Future perspectives

It is clear that the mechanisms responsible for the tissue-protective effects of statins are pleiotropic and multifactorial. In clinical settings, the evidence on the exclusive vascular protection of statins (independent from their lipid-lowering property) is relatively insufficient, as therapeutic indications of statins usually do not include vascular protection (Table 1). Clinical studies investigating the pleiotropic effect of statins in patients without dyslipidemia are therefore warranted in order to extend their clinical applications. Currently, our research group is conducting a randomized controlled trial in diabetic patients with a normal blood lipid profile to determine the perioperative vascular protection of rosuvastatin on the function of arteriovenous fistula, which is created for hemodialysis (ClinicalTrials.gov, protocol record 101-2314-B-006-045). This ongoing clinical trial is expected to manifest the vascular protective effects of rosuvastatin in the arteriovenous fistula of diabetic patients, as well as to enhance our ability to prevent and manage vascular access failure in diabetic patients with end-stage renal disease.

The optimal dose and treatment period for perioperative administration of statins are still a subject of debate. Dose-ranging and time-course studies are necessary to provide a more accurate use of different statins during the perioperative period. In addition, more clinical evidence is required to support the use of statins as part of acute-phase therapy prior to a major operation. Because surgical patients are most likely to suffer from prolonged fasting prior to and after a major operation, the development of parenteral preparations for statins would be highly desirable to enhance the bioavailability of perioperative statin delivery.40

12. Conclusion

Apart from the lipid-lowering property, statins mediate important pleiotropic effects in vascular protection, and are apparently applicable to other systemic organ protection (renal, neurological, and psychosomatic systems) during the perioperative period. Perioperative statin use reduces overall mortality, the development of myocardial infarction and atrial fibrillation, and duration of hospital stay after a major cardiac and noncardiac surgery. Patients who are currently undergoing long-term statin treatment should continue it perioperatively, because discontinuation of statins may result in adverse cardiovascular events. Perioperative use of statins is generally well tolerated, but attention should be paid to patients with old age, small body size, and advanced stage comorbidities.

Acknowledgments

This work is supported in part by research grants from the Multidisciplinary Center of Excellence for Clinical Trial and Research Department of Health (Grant No. DOH101-TD-111-102 to CFL) and from the National Science Council of Taiwan National Institutes of Health (Grant No. 98-2314-B-006-055-MY2 to CFL), Taiwan.


References

1
D.W. Russell
Cholesterol biosynthesis and metabolism
Cardiovasc Drugs Ther, 6 (1992), pp. 103-110
2
M.H. Moghadasian
Clinical pharmacology of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors
Life Sci, 65 (1999), pp. 1329-1337
3
S. Bellosta, R. Paoletti, A. Corsini
Safety of statins: focus on clinical pharmacokinetics and drug interactions
Circulation, 109 (23 suppl 1) (2004), pp. III50-III57
4
J.E. Edwards, R.A. Moore
Statins in hypercholesterolaemia: a dose-specific meta-analysis of lipid changes in randomised, double blind trials
BMC Fam Pract, 4 (2003), p. 18
5
J.C. LaRosa, J. He, S. Vupputuri
Effect of statins on risk of coronary disease: a meta-analysis of randomized controlled trials
JAMA, 282 (1999), pp. 2340-2346
6
M. Endres, U. Laufs, Z. Huang, T. Nakamura, P. Huang, M.A. Moskowitz, et al.
Stroke protection by 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors mediated by endothelial nitric oxide synthase
Proc Natl Acad Sci USA, 95 (1998), pp. 8880-8885
7
L. Kalinowski, L.W. Dobrucki, V. Brovkovych, T. Malinski
Increased nitric oxide bioavailability in endothelial cells contributes to the pleiotropic effect of cerivastatin
Circulation, 105 (2002), pp. 933-938
8
M. Eto, T. Kozai, F. Cosentino, H. Joch, T.F. Lüscher
Statin prevents tissue factor expression in human endothelial cells: role of Rho/Rho-kinase and Akt pathways
Circulation, 105 (2002), pp. 1756-1759
9
J. Wang, T. Tokoro, K. Matsui, S. Higa, I. Kitajima
Pitavastatin at low dose activates endothelial nitric oxide synthase through PI3K-AKT pathway in endothelial cells
Life Sci, 76 (2005), pp. 2257-2268
10
U. Laufs, J.K. Liao
Post-transcriptional regulation of endothelial nitric oxide synthase mRNA stability by Rho GTPase
J Biol Chem, 273 (1998), pp. 24266-24271
11
J.L. Mehta, J. Chen, P.L. Hermonat, F. Romeo, G. Novelli
Lectin-like, oxidized low-density lipoprotein receptor-1 (LOX-1): a critical player in the development of atherosclerosis and related disorders
Cardiovasc Res, 69 (2006), pp. 36-45
12
T. Sawamura, N. Kume, T. Aoyama, H. Moriwaki, H. Hoshikawa, Y. Aiba, et al.
An endothelial receptor for oxidized low-density lipoprotein
Nature, 386 (1997), pp. 73-77
13
E. Chavakis, E. Dernbach, C. Hermann, U.F. Mondorf, A.M. Zeiher, S. Dimmeler
Oxidized LDL inhibits vascular endothelial growth factor-induced endothelial cell migration by an inhibitory effect on the Akt/endothelial nitric oxide synthase pathway
Circulation, 103 (2001), pp. 2102-2107
14
A. Blair, P.W. Shaul, I.S. Yuhanna, P.A. Conrad, E.J. Smart
Oxidized low density lipoprotein displaces endothelial nitric-oxide synthase (eNOS) from plasmalemmal caveolae and impairs eNOS activation
J Biol Chem, 274 (1999), pp. 32512-32519
15
D.W. Stepp, J. Ou, A.W. Ackerman, S. Welak, D. Klick, K.A. Pritchard Jr.
Native LDL and minimally oxidized LDL differentially regulate superoxide anion in vascular endothelium in situ
Am J Physiol Heart Circ Physiol, 283 (2002), pp. H750-H759
16
R.A. Catar, G. Müller, J. Heidler, G. Schmitz, S.R. Bornstein, H. Morawietz
Low-density lipoproteins induce the renin-angiotensin system and their receptors in human endothelial cells
Horm Metab Res, 39 (2007), pp. 801-805
17
J. Chen, D. Li, R. Schaefer, J.L. Mehta
Cross-talk between dyslipidemia and renin-angiotensin system and the role of LOX-1 and MAPK in atherogenesis studies with the combined use of rosuvastatin and candesartan
Atherosclerosis, 184 (2006), pp. 295-301
18
C.M. Boulanger, F.C. Tanner, M.L. Béa, A.W. Hahn, A. Werner, T.F. Lüscher
Oxidized low density lipoproteins induce mRNA expression and release of endothelin from human and porcine endothelium
Circ Res, 70 (1992), pp. 1191-1197
19
M.E. Ivey, N. Osman, P.J. Little
Endothelin-1 signalling in vascular smooth muscle: pathways controlling cellular functions associated with atherosclerosis
Atherosclerosis, 199 (2008), pp. 237-247
20
L. Kalinowski, I.T. Dobrucki, T. Malinski
Cerivastatin potentiates nitric oxide release and enos expression through inhibition of isoprenoids synthesis
J Physiol Pharmacol, 53 (2002), pp. 585-595
21
F. Jantzen, S. Könemann, B. Wolff, S. Barth, A. Staudt, H.K. Kroemer, et al.
Isoprenoid depletion by statins antagonizes cytokine-induced down-regulation of endothelial nitric oxide expression and increases NO synthase activity in human umbilical vein endothelial cells
J Physiol Pharmacol, 58 (2007), pp. 503-514
22
A.H. Wagner, T. Köhler, U. Rückschloss, I. Just, M. Hecker
Improvement of nitric oxide-dependent vasodilatation by HMG-CoA reductase inhibitors through attenuation of endothelial superoxide anion formation
Arterioscler Thromb Vasc Biol, 20 (2000), pp. 61-69
23
C. Vecchione, M.T. Gentile, A. Aretini, G. Marino, R. Poulet, A. Maffei, et al.
A novel mechanism of action for statins against diabetes-induced oxidative stress
Diabetologia, 50 (2007), pp. 874-880
24
H. Nishimatsu, E. Suzuki, H. Satonaka, R. Takeda, M. Omata, T. Fujita, et al.
Endothelial dysfunction and hypercontractility of vascular myocytes are ameliorated by fluvastatin in obese Zucker rats
Am J Physiol Heart Circ Physiol, 288 (2005), pp. H1770-H1776
25
O. Hofnagel, B. Luechtenborg, H. Eschert, G. Weissen-Plenz, N.J. Severs, H. Robenek
Pravastatin inhibits expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) in Watanabe heritable hyperlipidemic rabbits: a new pleiotropic effect of statins
Arterioscler Thromb Vasc Biol, 26 (2006), pp. 604-610
26
T. Matsumoto, M. Fujita, T. Sawamura, A. Kakino, Y. Sato, Y. Fujita, et al.
Pitavastatin reduces lectin-like oxidized low-density lipoprotein receptor-1 ligands in hypercholesterolemic humans
Lipids, 45 (2010), pp. 329-335
27
Y. Rikitake, S. Kawashima, S. Takeshita, T. Yamashita, H. Azumi, M. Yasuhara, et al.
Anti-oxidative properties of fluvastatin, an HMG-CoA reductase inhibitor, contribute to prevention of atherosclerosis in cholesterol-fed rabbits
Atherosclerosis, 154 (2001), pp. 87-96
28
J.A. Beckman, J.K. Liao, S. Hurley, L.A. Garrett, D. Chui, D. Mitra, et al.
Atorvastatin restores endothelial function in normocholesterolemic smokers independent of changes in low-density lipoprotein
Circ Res, 95 (2004), pp. 217-223
29
M. Sugiyama, M. Ohashi, H. Takase, K. Sato, R. Ueda, Y. Dohi
Effects of atorvastatin on inflammation and oxidative stress
Heart Vessels, 20 (2005), pp. 133-136
30
J.N. Roan, S.Y. Fang, S.W. Chang, C.H. Hsu, C.C. Huang, M.H. Chiou, et al.
Rosuvastatin improves vascular function of arteriovenous fistula in a diabetic rat model
J Vasc Surg, 56 (2012), pp. 1381-1389 e1
31
S.Y. Fang, J.N. Roan, Y. Lin, C.H. Hsu, S.W. Chang, C.C. Huang, et al.
Rosuvastatin suppresses oxidative response in the venous limb of arteriovenous fistula and enhances fistula blood flow in diabetic rats
J Vasc Res (2013) in press
Article  
32
F. Mraiche, J. Cena, D. Das, B. Vollrath
Effects of statins on vascular function of endothelin-1
Br J Pharmacol, 144 (2005), pp. 715-726
33
S. Dimmeler, A. Aicher, M. Vasa, C. Mildner-Rihm, K. Adler, M. Tiemann, et al.
HMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI 3-kinase/Akt pathway
J Clin Invest, 108 (2001), pp. 391-397
34
Y. Liu, J. Wei, S. Hu, L. Hu
Beneficial effects of statins on endothelial progenitor cells
Am J Med Sci, 344 (2012), pp. 220-226
35
Q. Zhou, J.K. Liao
Pleiotropic effects of statins. Basic research and clinical perspectives
Circ J, 74 (2010), pp. 818-826
36
P. Balakumar, S. Kathuria, G. Taneja, S. Kalra, N. Mahadevan
Is targeting eNOS a key mechanistic insight of cardiovascular defensive potentials of statins?
J Mol Cell Cardiol, 52 (2012), pp. 83-92
37
J.T. Christenson
Preoperative lipid-control with simvastatin reduces the risk of postoperative thrombocytosis and thrombotic complications following CABG
Eur J Cardiothorac Surg, 15 (1999), pp. 394-399
38
E.W. Kuhn, O.J. Liakopoulos, S. Stange, A.C. Deppe, I. Slottosch, Y.H. Choi, et al.
Preoperative statin therapy in cardiac surgery: a meta-analysis of 90 000 patients
Eur J Cardiothorac Surg (2013) in press [Epub ahead of print (PMID 23562936)]
Article  
39
L.M. Fedoruk, H. Wang, M.R. Conaway, I.L. Kron, K.C. Johnston
Statin therapy improves outcomes after valvular heart surgery
Ann Thorac Surg, 85 (2008), pp. 1521-1526
40
E.W. Kuhn, O.J. Liakopoulos, Y.H. Choi, T. Wahlers
Current evidence for perioperative statins in cardiac surgery
Ann Thorac Surg, 92 (2011), pp. 372-379
41
D. Poldermans, J.J. Bax, M.D. Kertai, B. Krenning, C.M. Westerhout, A.F. Schinkel, et al.
Statins are associated with a reduced incidence of perioperative mortality in patients undergoing major noncardiac vascular surgery
Circulation, 107 (2003), pp. 1848-1851
42
M.D. Kertai, E. Boersma, C.M. Westerhout, J. Klein, H. van Urk, J.J. Bax, et al.
A combination of statins and beta-blockers is independently associated with a reduction in the incidence of perioperative mortality and nonfatal myocardial infarction in patients undergoing abdominal aortic aneurysm surgery
Eur J Vasc Endovasc Surg, 28 (2004), pp. 343-352
43
J. Kennedy, H. Quan, A.M. Buchan, W.A. Ghali, T.E. Feasby
Statins are associated with better outcomes after carotid endarterectomy in symptomatic patients
Stroke, 36 (2005), pp. 2072-2076
44
O. Schouten, E. Boersma, S.E. Hoeks, R. Benner, H. van Urk, M.R. van Sambeek, et al.
Fluvastatin and perioperative events in patients undergoing vascular surgery
N Engl J Med, 36 (2009), pp. 980-989
45
V. Chopra, D.H. Wesorick, J.B. Sussman, T. Greene, M. Rogers, J.B. Froehlich, et al.
Effect of perioperative statins on death, myocardial infarction, atrial fibrillation, and length of stay: a systematic review and meta-analysis
Arch Surg, 147 (2012), pp. 181-189
46
G.C. Fonarow, R.S. Wright, F.A. Spencer, P.D. Fredrick, W. Dong, N. Every, et al.
Effect of statin use within the first 24 hours of admission for acute myocardial infarction on early morbidity and mortality
Am J Cardiol, 96 (2005), pp. 611-616
47
Y. Le Manach, G. Godet, P. Coriat, C. Martinon, M. Bertrand, M.H. Fléron, et al.
The impact of postoperative discontinuation or continuation of chronic statin therapy on cardiac outcome after major vascular surgery
Anesth Analg, 104 (2007), pp. 1326-1333
48
L.A. Fleisher, J.A. Beckman, K.A. Brown, H. Calkens, E.L. Chaikof, K.E. Fleischmann, et al.
ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (writing committee to revise the 2002 guidelines on perioperative cardiovascular evaluation for noncardiac surgery): developed in collaboration with the American Society of Echocardiography, American Society of Nuclear Cardiology, Heart Rhythm Society, Society of Cardiovascular Anesthesiologists, Society for Cardiovascular Angiography and Interventions, Society for Vascular Medicine and Biology, and Society for Vascular Surgery
Circulation, 116 (2007), pp. 1971-1996
49
N. Fallouh, V. Chopra
Statin withdrawal after major noncardiac surgery: risks, consequences, and preventative strategies
J Hosp Med, 7 (2012), pp. 573-579
50
Y. Le Manach, P. Coriat, C.D. Collard, B. Riedel
Statin therapy within the perioperative period
Anesthesiology, 108 (2008), pp. 1141-1146
51
K.I. Paraskevas, D.P. Mikhailidis, F.J. Veith
Optimal statin type and dosage for vascular patients
J Vasc Surg, 53 (2011), pp. 837-844
52
K.I. Paraskevas, F.J. Veith, C.D. Liapis, D.P. Mikhailidis
Perioperative/periprocedural effects of statin treatment for patients undergoing vascular surgery or endovascular procedures: an update
Curr Vasc Pharmacol, 11 (2013), pp. 112-120
53
C.P. Cannon, E. Braunwald, C.H. McCabe, D.J. Rader, J.L. Rouleau, R. Belder, et al.
Intensive versus moderate lipid lowering with statins after acute coronary syndromes
N Engl J Med, 350 (2004), pp. 1495-1504
54
The Post Coronary Artery Bypass Graft Trial Investigators
The effect of aggressive lowering of low-density lipoprotein cholesterol levels and low-dose anticoagulation on obstructive changes in saphenous-vein coronary-artery bypass grafts
N Engl J Med, 336 (1997), pp. 153-162
Article  
55
B.M. Biccard
A peri-operative statin update for non-cardiac surgery. Part I: the effects of statin therapy on atherosclerotic disease and lessons learnt from statin therapy in medical (non-surgical) patients
Anaesthesia, 63 (2008), pp. 52-64
56
A. Kashani, C.O. Phillips, J.M. Foody, Y. Wang, S. Mangalmurti, D.T. Ko, et al.
Risks associated with statin therapy: a systematic overview of randomized clinical trials
Circulation, 114 (2006), pp. 2788-2797
57
B.M. Biccard, J.W. Sear, P. Foëx
Statin therapy: a potentially useful peri-operative intervention in patients with cardiovascular disease
Anaesthesia, 60 (2005), pp. 1106-1114
58
S.M. Grundy
The issue of statin safety: where do we stand?
Circulation, 111 (2005), pp. 3016-3019
59
A. Turan, M.L. Mendoza, S. Gupta, J. You, A. Gottlieb, W. Chu, et al.
Consequences of succinylcholine administration to patients using statins
Anesthesiology, 115 (2011), pp. 28-35

References

Close