Original article

K. KORYBALSKA, E. KAWKA, A. BREBOROWICZ, J. WITOWSKI


ATORVASTATIN DOES NOT IMPAIR ENDOTHELIAL CELL WOUND
HEALING IN AN IN VITRO MODEL OF VASCULAR INJURY



Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland


  Percutaneous coronary intervention (PCI) became standard treatment modality for coronary revascularization. However, it is associated with endothelial cell denudation, which may predispose to in-stent thrombosis and restenosis. Since statins may inhibit vascular cell proliferation, one may fear that their early administration will delay post-PCI re-endothelialization. We have therefore employed an in vitro scratch assay to examine how atorvastatin affects endothelial cell wound healing. Human umbilical vein endothelial cells (HUVEC) were grown to confluence and wounded by scraping. Scratch healing in the presence or absence of atorvastatin was monitored by time-lapse photo-microscopy. In addition cells were assessed for viability (MTT assay), migration (chemotaxis chamber), proliferation (3H-thymidine and bromodeoxyuridine incorporation), and cytokine production (immunoassays). The exposure of HUVEC to atorvastatin resulted in a dose-dependent decrease in cell viability and proliferation. However, this effect was observed only at doses 1 µM, which is well above the concentrations seen in vivo. At clinically relevant doses (0.1 µM) atorvastatin did not impair wound closure, nor did it inhibit cell viability, proliferation, and migration. It did however reduce the constitutive and the stimulated release of cytokines (IL-6, IL-8, MCP-1), adhesion molecules (sICAM-1) and matrix proteins (fibronectin). We conclude that atorvastatin at doses corresponding to concentrations seen in serum during standard therapy does not impair endothelial cell regeneration after injuries mimicking those occurring during PCI. It does, however, inhibits the secretion of pro-inflammatory mediators.

Key words: atorvastatin, endothelial cells, inflammation, vascular injury, percutaneous coronary intervention, statins, wound healing



INTRODUCTION

Percutaneous coronary intervention (PCI) is now routinely used for revascularization of occluded coronary arteries. Recent meta-analyses indicate that intensive short-term pre-treatment with statins may significantly reduce peri-procedural complications and adverse events in patients undergoing PCI (1-4). The mechanism whereby statins exert this effect is not clear, but appears to be independent of cholesterol lowering. More likely it is related to anti-inflammatory properties of statins and their ability to alleviate PCI-associated endothelial cell activation (5). It has been demonstrated that statins improve endothelial cell-dependent coronary blood flow (6, 7) and reduce endothelial cell expression of adhesion molecules (8) and pro-inflammatory cytokines (9). In vitro and animal studies indicate that the beneficial effect of statins may also be related to increased bioavailability of nitric oxide (10, 11).

In addition, statins are known to recruit bone marrow-derived endothelial progenitor cells (EPC) (12). In this respect, a recent study has demonstrated that intensive therapy with atorvastatin initiated before PCI effectively mobilizes EPC and increases their attachment to stent struts (13). This effect is of importance as PCI is associated with endothelial cell injury and prompt re-endothelialization reduces neointimal hyperplasia and subsequent restenosis (14). However, the effect of statins on endothelial cell regeneration may be more complex. This is because healing of endothelial cell wounds occurs not only through EPC incorporation, but also through proliferation and migration of neighboring cells (15). The impact of statins on these processes is less clear and appears to depend critically on the dose applied (16). At low concentrations statins stimulate endothelial cell proliferation and migration, while high concentrations act to the contrary (17). In the clinical setting both effects could be viewed as beneficial, since stimulation of cell proliferation would accelerate endothelial cell recovery from PCI-induced injuries, but on the other hand, the inhibition of endothelial cell growth within atherosclerotic lesions would decrease the risk of plaque rupture. To strike the right balance between these effects, both timing and dosage of therapy need to be carefully optimized so that the plaque stability is improved without compromising post-PCI re-endothelialization.

Most clinical experience on peri-procedural statin administration was gained from studies with atorvastatin (4). Pre-treatment with atorvastatin showed benefits both in patients with stable angina undergoing elective PCI (18) and in patients with acute coronary syndromes undergoing early PCI (19). We have therefore focused on atorvastatin and set out to examine how acute exposure to atorvastatin at clinically relevant doses affects vascular endothelial cell wound healing independent of EPC. To this end we analyzed recovery of cultured endothelial cells from scratch injuries mimicking those occurring during PCI.


MATERIAL AND METHODS

Unless indicated otherwise, all reagents were from Sigma-Aldrich. Cell culture plastics were from Nunc and Costar. The study protocol was accepted by local Ethical Commettee at Poznan Medical University.

Cell culture

The experiments were performed using human umbilical vein endothelial cells (HUVEC) of the EA.hy926 line (kindly provided by Dr. CJ Edgell, University of North Carolina, Chapel Hill, USA) (20). Cells were routinely maintained in Earl’s-buffered M199 culture medium, supplemented with amphotericin (2.5 µg/mL), gentamycin (50 µg/mL), L-glutamine (2 mM), hydrocortisone (0.4 µg/mL), and 10% v/v fetal calf serum (Invitrogen).

Atorvastatin exposure

Atorvastatin was kindly donated by Pfizer and dissolved in dimethyl sulfoxide (DMSO) (21, 22). Atorvastatin concentrations of 0.01–0.1 µM that were used throughout the study corresponded to the levels found in serum after oral administration of 40 mg atorvastatin (23). Final DMSO concentration in test media was 0.2% (v/v) and the same concentration was applied to the controls. Preliminary experiments have determined that this DMSO concentration did not impair HUVEC viability, as assessed by the MTT test (data not shown).

Cell proliferation

Cell proliferation was assessed by [3H]-thymidine incorporation. Briefly, cells were plated at a density of 2x104 cells/cm2, allowed to attach for 4 hours and then treated for 24 hours with either atorvastatin or vehicle control in the presence of [3H]-thymidine (1 µCi/ml; Institute of Radioisotopes, Prague, Czech Republic). After the incubation the cells were harvested, precipitated with 10% (w/v) trichloroacetic acid, and dissolved in 0.1 M NaOH. The radioactivity released was measured in a beta liquid scintillation counter (Wallac Perkin Elmer).

Cell migration

Cell migration was assessed with the use of QCMTM Chemotaxis 96-well Cell Migration Assay with 8 µm pore size membranes (Chemicon/Milipore). Cells at 80% confluence were incubated in the presence of either atorvastatin or vehicle control for 24 hours and then rendered quiescent by serum reduction (to 0.1%) for the next 24 hours. After that the cells were harvested, washed, re-suspended in serum-free medium and placed in a migration chamber (5x104 cells/100 µl). Cells were then stimulated for 24 hours with standard 10% serum-containing medium with or without atorvastatin. Migrated cells were detached and treated for 15 minutes with the CyQuant GR dye in the lysis buffer, as per manufacturer’s instructions. Fluorescence of cell lysates was measured with a fluorescence microplate reader (Perkin Elmer) using 480 nm and 520 nm wavelengths for excitation and emission, respectively.

Cell viability

viability of cells following a 24-hour exposure to atorvastatin was assessed with the MTT assay. It measures the metabolic conversion of the MTT salt (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolinum bromide) by active mitochondrial dehydrogenases (24).The assay was performed as described previously (25). Briefly, after exposure to atorvastatin, the cells were treated with MTT (1.25 mg/ml) for 4 hours at 37°C. The formazan product generated was solubilized by the addition of 20% sodium dodecyl sulfate and 50% N,N-dimethylformamide, and quantified by measuring its absorbance at 595 nm.

Wounding healing

Cells were grown to confluence, pre-treated with or without atorvastatin for 24 hours and then scratched with a cell scraper (Nunc). The resulting debris was removed by gentle washing with medium. After that the cells were placed in an incubator coupled to an Axio Observer D1 inverted microscope (Zeiss). Cells were maintained for up to 12 hours in standard culture medium with either atorvastatin or vehicle control. The images of the closing wound were acquired by time-lapse microscopy at 30-minute intervals and analyzed using the AxioVision Rel. 4.6.3 image analysis software (Zeiss).

Cytochemistry

All cytochemical procedures were performed on cells cultured in Lab-Tek Chamber Slides (Nunc). Incorporation of bromodeoxyuridine (BrdU) was visualized using a Zymed® BrdU Staining kit (Invitrogen), as per manufacturer’s instructions.

Cytokine measurements

Release of selected cytokines (IL-6, IL-8, MCP-1), adhesion molecules (sICAM-1), and extracellular matrix proteins (fibronectin) by HUVEC was assessed under basal conditions (constitutive release) and following the stimulation with IL-1ß (1 ng/ml) and TNF- (10 ng/ml). Target molecules were measured with appropriate DuoSet Immunoassay Kits (R&D Systems). The assays were designed and performed according to the manufacturer’s instructions. Fibronectin was measured as previously described (26). The results were normalized per µg of cell protein. Protein concentration in cell lysates was determined with the Bradford method (27) using the Protein Assay Dye Reagent (Bio-Rad).

Statistical analysis

The data were analyzed with repeated measures analysis of variance using GraphPrism™ 5.00 software (GraphPad Software Inc.). A p value of less than 0.05 was considered significant. Results are presented as means ±S.D.


RESULTS

Effect of atorvastatin on endothelial cell proliferation and viability

Exposure of HUVEC to atorvastatin resulted in a dose-dependent inhibition of cell viability as measured by the MTT assay (Fig. 1). After 24 hours the effect was evident for 10 µM atorvastatin, and after prolonged incubation also for 1 µM. Atorvastatin at doses 1 µM inhibited also cell proliferation (Fig. 2). At the highest dose of atorvastatin tested (10 µM) HUVEC proliferation was 64 ±3% of the control. In contrast, atorvastatin at clinically relevant doses of 0.01 and 0.1 µM impaired neither HUVEC viability proliferation. These doses were used in further experiments.

Fig. 1. Effect of atorvastatin on endothelial cell viability. HUVEC were treated with increasing doses of atorvastatin or vehicle control for 1, 2, and 6 days. After the exposure cell viability was measured with the MTT test (n=3). Data were expressed as a percentage of control at the same time point. Asterisks represent a significant difference compared to a respective control.

Fig. 2. Effect of atorvastatin on endothelial cell proliferation. Proliferation of HUVEC treated with increasing doses of atorvastatin was assessed over a 24-hour period by 3H-thymidine incorporation (n=8). Asterisks represent a significant difference compared to control cells.

Effect of atorvastatin on endothelial cell wound closure

Scratch wounds were inflicted on cells pre-treated with or without atorvastatin for 24 hours. The surface area of the wounds generated did not differ between the groups and was 444 ±50 mm2, 402 ±55 mm2, and 419 ±42 mm2 for cells exposed to vehicle control, 0.01 µM and 0.1 µM of atorvastatin, respectively. Cells from all groups repopulated the denuded areas within 12 hours and there was no difference in the kinetics of the process between cells treated with or without atorvastatin (Fig. 3).

Fig. 3. Effect of atorvastatin on endothelial cell wound healing. (A): Exemplary microphotographs of wound closure in control HUVEC; magnification 100x; (B): Kinetics of wound healing in cells pre-treated for 24 hour and then incubated in the presence or absence of atorvastatin (n=13).

To assess whether atorvastatin changed the contribution of cell proliferation to wound repair, endothelial cell monolayers were tested for incorporation of BrdU immediately before and 4 hours after a scratch injury (Fig. 4). The fraction of control cells that stained for BrdU was 35 ±8% before and 31 ±2% after injury. Pre-treatment of HUVEC with atorvastatin at 0.01 µM and 0.1 µM did not significantly change these percentages.

Fig. 4. Effect of atorvastatin on bromodeoxyuridine incorporation by endothelial cells before and after scratch injury. Exemplary microphotographs of BrdU labeling in control cells before (A) and after injury (B); magnification 100x; the arrows indicate typical cells staining positively for BrdU. (C): Cells were pre-treated with atorvastatin for 24 hours and then wounded. Percentage of cells incorporating BrdU was assessed in each group immediately before and 4 hours after injury (n=7).

Effect of atorvastatin on endothelial cell migration

Compared to the control, atorvastatin at doses up to 0.1 µM did not impair HUVEC migration in the absence of stimulation (Fig. 5). Following the stimulation with serum migration of control HUVEC increased to 208±76% of the baseline values. For cells exposed to 0.01 µM and 0.1 µM atorvastatin these values were 210±72% and 181±41%, respectively, and did not differ significantly from the controls.

Fig. 5. Effect of atorvastatin on endothelial cell migration. HUVEC were treated with atorvastatin as described in the Methods and assessed for the capacity to migrate in the presence or absence of stimulation (n=10).

Effect of atorvastatin on endothelial cell cytokine production

Exposure of HUVEC to atorvastatin resulted in a dose-dependent decrease in both the constitutive and the cytokine-stimulated release of IL-6, IL-8, MCP-1, sICAM-1, and fibronectin (Table 1). For MCP-1 and sICAM-1 the effect became evident with atorvastatin at 0.01 µM, for all other mediators it was significant with the dose of 0.1 µM.

Table 1. Effect of atorvastatin on the release of mediators by endothelial cells. Mediators secreted were measured in post-culture supernatants following a 24-hour exposure to atorvastatin (n=7) in the presence or absence of stimulation with IL-1ß (1 ng/ml) and TNF- (10 ng/ml). Asterisks represent a significant difference compared to control cells treated with the vehicle only.


DISSCUSION

Statins display pleiotropic properties and exert their benefits partly through the inhibition of vascular smooth muscle cell (VSMC) proliferation (28). This effect is important for the prevention of post-PCI restenosis. Several studies have demonstrated that statins reduce VSMC proliferation and migration in vitro (28-33). The concentrations of atorvastatin that produced this effect approximated those found in serum after oral administration of average therapeutic doses. However, Axel et al. observed that the same doses may inhibit the growth of endothelial cells to a significantly greater extent compared to VSMC (29). In contrast, Giordano et al. have recently demonstrated that statins at doses that effectively inhibited VSMC proliferation and migration did not impair these processes in endothelial cells (34). Jaschke et al. (35) observed a similar effect in cells treated with cerivastatin. They have also demonstrated that the implantation of stents coated with cerivastatin did not impair re-endothelialization but inhibited the neoinitima formation (35). Our data seem to be in keeping with these observations and confirm that atorvastatin at doses up to 0.1 µM does not delay endothelial cell wound closure and does not hamper endothelial cell proliferation and migration. These findings are of clinical significance as the integrity of the endothelial barrier protects against excessive VSMC growth (36). Failure of the denuded surfaces to re-endothelialize leads to increased accumulation of VSMC and the neointima formation (37). Urbich et al. (16) have titrated the effect of atorvastatin to find that it promoted endothelial cell migration at low doses (0.001–0.01 µM), did not affect it at moderate doses (0.1 µM), but inhibited it at high doses (1 µM). The mechanism underlying this biphasic effect of statins on endothelial cell proliferation and migration has been attributed to their impact on protein prenylation. It appears that at low doses statins inhibit cholesterol synthesis, but do not impair the biosynthesis of farnesyl and geranylgeranyl pyrophosphates that are key intermediates in the pathways controlling cell growth. In contrast, high doses of statins inhibit both the cholesterol synthesis and the synthesis of prenyl radicals, which results in the inhibition of cell proliferation and migration (5, 38).

Similar mechanisms may underlie anti-inflammatory effects of statins toward endothelial cells (5, 39). Statins (including atorvastatin at high doses) were found to inhibit production of MCP-1 and IL-8 (40, 41) and these effects were attributed to the inhibition of protein prenylation (42). On the other hand, Dayoub et al. (43) reported that atorvastatin at doses as high as 1 µM failed to inhibit the release of IL-6 from endothelial cells exposed to lipopolysaccharide. Interestingly, we have observed that even at low concentrations atorvastatin reduced the constitutive and stimulated production of inflammatory cytokines, adhesion molecules, and fibronectin. Adding to the complexity of this issue, it appears that the final effect of statins in vivo may depend on the exact clinical setting. For example, it has been demonstrated that simvastatin decreased the release of pro-inflammatory cytokines by monocytes from patients with isolated hypercholesterolemia but not from those with impaired glucose tolerance (44).

Several studies reported also on down-regulation by stations of endothelial cell ICAM-1 (39). This effect was reversed by mevalonate, and linked to the increased NO production (45). Indeed, it has been demonstrated that statins are capable of increasing the expression and activity of endothelial nitric oxide synthase (eNOS) in HUVEC (46). The inhibition by atorvastatin of fibronectin secretion also may bear clinical significance as fibronectin has been implicated in the formation of atherosclerotic plaques and expansion of VSMC (47). An additional benefit of atorvastatin in the context of acute coronary syndromes could be related to its anti-arrhythmic properties. It has been observed in an experimental rat model that atorvastatin decreased the propensity for ventricular arrhythmias, possibly by stabilizing the cardiomyocyte cell-to-cell junction integrity (48).

Given the clinical background of our study, the application of HUVEC rather than coronary artery endothelial cells may be seen as a limitation. However, gene expression profiling revealed that, apart from some genes expressed preferentially in the arteries, cells from these two locations shared a lot of similarities (49). Some reservations may also be raised against the usage of an immortalized HUVEC line rather than primary HUVEC. It has been demonstrated that the pattern of gene expression in unstimulated and atorvastatin-treated EA.hy926 cells matched that in primary HUVEC, although some rather expected differences in the expression of genes controlling cell cycle were observed (50). Therefore, additional studies that would validate our observations with primary coronary artery endothelial cells are warranted.

In summary, the present study shows that atorvastatin at clinically relevant concentrations does not impair endothelial cell wound healing but is capable of curtailing the production of pro-inflammatory cytokines. Our data indicate that atorvastatin at these doses is safe to use after PCI as it will not delay endothelial cell recovery from injuries. In addition, it may dampen the inflammatory response associated with the procedure.

Abbreviations: BrdU – bromodeoxyuridine; DMSO – dimethyl sulfoxide; eNOS – endothelial nitric oxide synthase; EPC – endothelial progenitor cells; HUVEC – human umbilical vein endothelial cells, IL – interleukin; MCP-1 – monocyte chemoattractant protein-1; MTT – 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolinum bromide; PCI – percutaneous coronary intervention; RFU – relative fluorescence units; sICAM-1 – soluble intercellular adhesion molecule-1; TNF-– tumor necrosis factor-; VSMC – vascular smooth muscle cells

Conflict of interests: None delared.



REFERENCES
  1. Hao PP, Chen YG, Wang JL, et al. Meta-analysis of the role of high-dose statins administered prior to percutaneous coronary intervention in reducing major adverse cardiac events in patients with coronary artery disease. Clin Exp Pharmacol Physiol 2010; 37: 496-500.
  2. Zhang F, Dong L, Ge J. Effect of statins pretreatment on periprocedural myocardial infarction in patients undergoing percutaneous coronary intervention: a meta-analysis. Ann Med 2010; 42: 171-177.
  3. Mood GR, Bavry AA, Roukoz H, Bhatt DL. Meta-analysis of the role of statin therapy in reducing myocardial infarction following elective percutaneous coronary intervention. Am J Cardiol 2007; 100: 919-923.
  4. Patti G, Cannon CP, Murphy SA, et al. Clinical benefit of statin pretreatment in patients undergoing percutaneous coronary intervention: a collaborative patient-level meta-analysis of 13 randomized studies. Circulation 2011; 123: 1622-1632.
  5. Ii M, Losordo DW. Statins and the endothelium. Vascul Pharmacol 2007; 46: 1-9.
  6. Wassmann S, Faul A, Hennen B, Scheller B, Bohm M, Nickenig G. Rapid effect of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition on coronary endothelial function. Circ Res 2003; 93: e98-103.
  7. Hinoi T, Matsuo S, Tadehara F, Tsujiyama S, Yamakido M. Acute effect of atorvastatin on coronary circulation measured by transthoracic Doppler echocardiography in patients without coronary artery disease by angiography. Am J Cardiol 2005; 96: 89-91.
  8. Patti G, Chello M, Pasceri V, et al. Protection from procedural myocardial injury by atorvastatin is associated with lower levels of adhesion molecules after percutaneous coronary intervention: results from the ARMYDA-CAMs (atorvastatin for reduction of myocardial damage during angioplasty-cell adhesion molecules) substudy. J Am Coll Cardiol 2006; 48: 1560-1566.
  9. Pasceri V, Cheng JS, Willerson JT, Yeh ET. Modulation of C-reactive protein-mediated monocyte chemoattractant protein-1 induction in human endothelial cells by anti-atherosclerosis drugs. Circulation 2001; 103: 2531-2534.
  10. Kalinowski L, Dobrucki LW, Brovkovych V, Malinski T. Increased nitric oxide bioavailability in endothelial cells contributes to the pleiotropic effect of cerivastatin. Circulation 2002; 105: 933-938.
  11. Atar S, Ye Y, Lin Y, et al. Atorvastatin-induced cardioprotection is mediated by increasing inducible nitric oxide synthase and consequent S-nitrosylation of cyclooxygenase-2. Am J Physiol Heart Circ Physiol 2006; 290: H1960-H1968.
  12. Walter DH, Zeiher AM, Dimmeler S. Effects of statins on endothelium and their contribution to neovascularization by mobilization of endothelial progenitor cells. Coron Artery Dis 2004; 15: 235-242.
  13. Hibbert B, Ma X, Pourdjabbar A, et al. Pre-procedural atorvastatin mobilizes endothelial progenitor cells: clues to the salutary effects of statins on healing of stented human arteries. PLoS ONE 2011; 6: e16413.
  14. Jukema JW, Verschuren JJ, Ahmed TA, Quax PH. Restenosis after PCI. Part 1: Pathophysiology and risk factors. Nat Rev Cardiol 2011; 9: 53-62.
  15. Minamino T, Komuro I. Vascular aging: insights from studies on cellular senescence, stem cell aging, and progeroid syndromes. Nat Clin Pract Cardiovasc Med 2008; 5: 637-648.
  16. Urbich C, Dernbach E, Zeiher AM, Dimmeler S. Double-edged role of statins in angiogenesis signaling. Circ Res 2002; 90: 737-744.
  17. Weis M, Heeschen C, Glassford AJ, Cooke JP. Statins have biphasic effects on angiogenesis. Circulation 2002; 105: 739-745.
  18. Pasceri V, Patti G, Nusca A, Pristipino C, Richichi G, Di SG. Randomized trial of atorvastatin for reduction of myocardial damage during coronary intervention: results from the ARMYDA (atorvastatin for reduction of myocardial damage during angioplasty) study. Circulation 2004; 110: 674-678.
  19. Patti G, Pasceri V, Colonna G, et al. Atorvastatin pretreatment improves outcomes in patients with acute coronary syndromes undergoing early percutaneous coronary intervention: results of the ARMYDA-ACS randomized trial. J Am Coll Cardiol 2007; 49: 1272-1278.
  20. Edgell CJ, McDonald CC, Graham JB. Permanent cell line expressing human factor VIII-related antigen established by hybridization. Proc Natl Acad Sci USA 1983; 80: 3734-3737.
  21. Dulak J, Loboda A, Jazwa A, et al. Atorvastatin affects several angiogenic mediators in human endothelial cells. Endothelium 2005; 12: 233-241.
  22. Blank N, Schiller M, Krienke S, et al. Atorvastatin inhibits T cell activation through 3-hydroxy-3-methylglutaryl coenzyme A reductase without decreasing cholesterol synthesis. J Immunol 2007; 179: 3613-3621.
  23. Bahrami G, Mohammadi B, Mirzaeei S, Kiani A. Determination of atorvastatin in human serum by reversed-phase high-performance liquid chromatography with UV detection. J Chromatogr B Analyt Technol Biomed Life Sci 2005; 826: 41-45.
  24. Carmichael J, DeGraff WG, Gazdar AF, Minna JD, Mitchell JB. Evaluation of a tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res 1987; 47: 936-942.
  25. Witowski J, Korybalska K, Wisniewska J, et al. Effect of glucose degradation products on human peritoneal mesothelial cell function. J Am Soc Nephrol 2000; 11: 729-739.
  26. Witowski J, Wisniewska J, Korybalska K, et al. Prolonged exposure to glucose degradation products impairs viability and function of human peritoneal mesothelial cells. J Am Soc Nephrol 2001; 12: 2434-2441.
  27. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976; 72: 248-254.
  28. Bellosta S, Arnaboldi L, Gerosa L, et al. Statins effect on smooth muscle cell proliferation. Semin Vasc Med 2004; 4: 347-356.
  29. Axel DI, Riessen R, Runge H, Viebahn R, Karsch KR. Effects of cerivastatin on human arterial smooth muscle cell proliferation and migration in transfilter cocultures. J Cardiovasc Pharmacol 2000; 35: 619-629.
  30. Yang Z, Kozai T, van der Loo B, et al. HMG-CoA reductase inhibition improves endothelial cell function and inhibits smooth muscle cell proliferation in human saphenous veins. J Am Coll Cardiol 2000; 36: 1691-1697.
  31. Porter KE, Naik J, Turner NA, Dickinson T, Thompson MM, London NJ. Simvastatin inhibits human saphenous vein neointima formation via inhibition of smooth muscle cell proliferation and migration. J Vasc Surg 2002; 36: 150-157.
  32. Yasunari K, Maeda K, Minami M, Yoshikawa J. HMG-CoA reductase inhibitors prevent migration of human coronary smooth muscle cells through suppression of increase in oxidative stress. Arterioscler Thromb Vasc Biol 2001; 21: 937-942.
  33. Turner NA, Midgley L, O’Regan DJ, Porter KE. Comparison of the efficacies of five different statins on inhibition of human saphenous vein smooth muscle cell proliferation and invasion. J Cardiovasc Pharmacol 2007; 50: 458-461.
  34. Giordano A, Romano S, Monaco M, et al. Differential effect of atorvastatin and tacrolimus on proliferation of vascular smooth muscle and endothelial cells. Am J Physiol Heart Circ Physiol 2012; 302: H135-H142.
  35. Jaschke B, Michaelis C, Milz S, et al. Local statin therapy differentially interferes with smooth muscle and endothelial cell proliferation and reduces neointima on a drug-eluting stent platform. Cardiovasc Res 2005; 68: 483-492.
  36. Cines DB, Pollak ES, Buck CA, et al. Endothelial cells in physiology and in the pathophysiology of vascular disorders. Blood 1998; 91: 3527-3561.
  37. Haudenschild CC, Schwartz SM. Endothelial regeneration. II. Restitution of endothelial continuity. Lab Invest 1979; 41: 407-418.
  38. Skaletz-Rorowski A, Walsh K. Statin therapy and angiogenesis. Curr Opin Lipidol 2003; 14: 599-603.
  39. Greenwood J, Mason JC. Statins and the vascular endothelial inflammatory response. Trends Immunol 2007; 28: 88-98.
  40. Romano M, Diomede L, Sironi M, et al. Inhibition of monocyte chemotactic protein-1 synthesis by statins. Lab Invest 2000; 80: 1095-1100.
  41. Zineh I, Luo X, Welder GJ, et al. Modulatory effects of atorvastatin on endothelial cell-derived chemokines, cytokines, and angiogenic factors. Pharmacotherapy 2006; 26: 333-340.
  42. Veillard NR, Braunersreuther V, Arnaud C, et al. Simvastatin modulates chemokine and chemokine receptor expression by geranylgeranyl isoprenoid pathway in human endothelial cells and macrophages. Atherosclerosis 2006; 188: 51-58.
  43. Dayoub JC, Ortiz F, Lopez LC, et al. Synergism between melatonin and atorvastatin against endothelial cell damage induced by lipopolysaccharide. J Pineal Res 2011; 51: 324-330.
  44. Krysiak R, Okopien B. Different effects of simvastatin on ex vivo monocyte cytokine release in patients with hypercholesterolemia and impaired glucose tolerance. J Physiol Pharmacol 2010; 61: 725-732.
  45. Xenos ES, Stevens SL, Freeman MB, Cassada DC, Goldman MH. Nitric oxide mediates the effect of fluvastatin on intercellular adhesion molecule-1 and platelet endothelial cell adhesion molecule-1 expression on human endothelial cells. Ann Vasc Surg 2005; 19: 386-392.
  46. Jantzen F, Konemann S, Wolff B, 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 2007; 58: 503-514.
  47. Astrof S, Hynes RO. Fibronectins in vascular morphogenesis. Angiogenesis 2009; 12: 165-175.
  48. Bacova B, Radosinska J, Knezl V, et al. Omega-3 fatty acids and atorvastatin suppress ventricular fibrillation inducibility in hypertriglyceridemic rat hearts: implication of intracellular coupling protein, connexin-43. J Physiol Pharmacol 2010; 61: 717-723.
  49. Chi JT, Chang HY, Haraldsen G, et al. Endothelial cell diversity revealed by global expression profiling. Proc Natl Acad Sci USA 2003; 100: 10623-10628.
  50. Boerma M, Burton GR, Wang J, Fink LM, McGehee RE, Jr., Hauer-Jensen M. Comparative expression profiling in primary and immortalized endothelial cells: changes in gene expression in response to hydroxy methylglutaryl-coenzyme A reductase inhibition. Blood Coagul Fibrinolysis 2006; 17: 173-180.

R e c e i v e d : June 1, 2012
A c c e p t e d : August 16, 2012

Author’s address: Prof. Janusz Witowski, Department of Pathophysiology, Poznan University of Medical Science, 6 Swiecickiego Street, 60-781 Poznan, Poland. e-mail: jwitow@ump.edu.pl