Skip main navigation

Statins Inhibit Leukocyte Recruitment

New Evidence for Their Anti-Inflammatory Properties
Originally publishedhttps://doi.org/10.1161/atvb.21.8.1256Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21:1256–1258

    Atherosclerosis and its devastating clinical complications, such as arterial thrombosis; ischemia and infarction of the heart, brain, and other vital organs; ruptured aortic aneurysms; and peripheral vascular insufficiency, continue to account for the majority of morbidity and mortality in the adult population of industrialized nations.1 The process of atherosclerosis is not simply an inevitable degenerative consequence of aging in the large arteries but a progressive disease characterized by chronic inflammation.2–4 Increasing evidence suggests that atherosclerosis is a multifactorial process involving the interplay of lipid metabolism, mononuclear leukocytes, coagulation proteins, cytokines, extracellular matrix, and hemodynamic forces.5 Since their introduction in the late 1980s, statins have revolutionized the treatment of dyslipidemia and demonstrated their ability to reduce and prevent coronary morbidity and mortality in both primary and secondary intervention trials.6 Statins competitively inhibit 3-hydroxyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase, an enzyme crucial to cholesterol biosynthesis. The resulting decrease in hepatic cholesterol concentration leads to a compensatory increase in expression of hepatic LDL receptors, which clear cholesterol-rich LDL and LDL precursors from the circulation. However, mevalonate, the product of HMG-CoA reductase, is the precursor not only for cholesterol but also for many nonsteroidal isoprenoid compounds. The isoprenoids farnesyl pyrophosphate and geranylgeranyl pyrophosphate are known to play an important role in signal transduction pathways by their attachment to signaling proteins, such as Ras and Rho.7

    See p 1165 and 1327

    The effectiveness and rapidity with which statins decrease coronary events already led a few years ago to the speculation that statins might influence vascular biology through mechanisms other than lowered plasma cholesterol.8–10 Indeed, numerous pleiotropic effects of statins on vascular cells have been identified during the past several years that could modulate atherogenesis, plaque rupture, or thrombosis. For example, statins upregulate nitric oxide (NO) expression in endothelial cells by inducing transcriptional activation of the endothelial NO synthase (eNOS) gene11,12 and inhibit induced ischemic cerebral stroke in normal but not in eNOS-deficient mice.13 Statins inhibit smooth muscle cell proliferation in vitro and in vivo and stimulate apoptosis.14–16 Statins affect fibrinolysis by enhancing the expression of tissue plasminogen activator and platelet activator inhibitor-117 and by decreasing the expression of matrix metalloproteinases.18–20 Finally, statins inhibit leukocyte adhesion and the expression of monocyte chemoattractant protein-1 (MCP-1), thereby possibly affecting the recruitment of inflammatory cells into the vascular intima of the atherosclerotic lesion.21–23 Recently, our group has demonstrated that statins also regulate some crucial molecules of the immune system and thus could be seen as immunomodulators.24–26

    In this issue and in the July issue of Arteriosclerosis, Thrombosis, and Vascular Biology, 2 articles describe exciting findings involving cholesterol-independent effects of statins on leukocyte recruitment.27,28 The study by Diomede and colleagues27 compared the effects of sterol and nonsterol derivatives arising from mevalonate biosynthesis on a well-defined in vivo model of local, acute inflammation: the air-pouch model. In 1 condition, mice received within 24 hours 3 repetitive doses of lovastatin, which did not affect levels of circulating cholesterol but reduced hepatic HMG-CoA reductase activity. In contrast, in the second condition, the animals received squalestatin, a selective inhibitor of the synthesis of sterol derivatives only, which reduced circulating cholesterol levels and increased hepatic HMG-CoA reductase activity through a negative-feedback mechanism. Subsequent pouch leukocyte recruitment induced by lipopolysaccharide or carrageenan was inhibited only in the group that received lovastatin and could be reversed by mevalonate administration to these mice. The authors then tested for the production of proinflammatory mediators in the air-pouch exudate. Inhibition of interleukin-6, MCP-1, and RANTES (Regulated upon Activation, Normally T-cell Expressed and Secreted) production could be detected in the exudates of lovastatin-treated mice only and not in those of squalestatin-treated mice. By excluding the anti-inflammatory effects of the inhibition of sterol synthesis through the use of squalestatin, the authors elegantly showed in this study that the biosynthesis of nonsterol derivatives arising from mevalonate is important for the anti-inflammatory effects of statins in vivo. In a recent study, effects of simvastatin were tested in mice deficient for apoE, a model for atherogenesis.29 Without affecting plasma cholesterol levels, daily doses of simvastatin for 6 weeks significantly reduced the development of atherosclerotic plaques. It would be worth testing squalestatin, and thus involvement of sterol derivatives arising from mevalonate, in this model of chronic inflammation. The study by Yoshida and colleagues28 sheds light on the mechanism responsible for the inhibitory effects of statins on leukocyte recruitment. In an in vitro model mimicking low physiological flow conditions, they observed that pretreatment of monocytic U937 cells with cerivastatin reduced adhesion of these cells to an activated endothelial monolayer. This effect, which could be reversed by mevalonate, coincided with decreased expression of cell surface integrins, such as CD11a, CD18, and VLA-4 and a disruption of F-actin organization. It has been recently reported that protein geranylgeranylation, and more specifically, RhoA geranylgeranylation, is required for integrin-dependent adhesion of leukocytes.30 In their study in this issue of Arteriosclerosis, Thrombosis, and Vascular Biology, Yoshida and colleagues28 also show that cerivastatin prevented the translocation of RhoA to the membrane fraction of the cell and that C3 transferase, an inhibitor of RhoA, inhibited adhesion of monocytic U937 cells to an endothelial monolayer. It is thus tempting to speculate that statins modulate integrin affinity by inhibition of geranylgeranylation of RhoA protein. Alternatively, statins may act through altered actin polymerization or through Rho-regulated altered clustering of adhesion molecules, as reported by Wojciak-Stothard et al.30 More recently, confirming the results of the 2 articles in this issue, it has been reported that the new statin rosuvastatin inhibits leukocyte rolling, adherence, and transmigration in a rat mesenteric microvasculature model in vivo.31 Furthermore, as seen for protection from ischemic stroke, the influence of statins on leukocyte recruitment was mediated via NO, because the statins had no effect in eNOS-null mice.13,31

    In concert, the 2 studies published in this issue demonstrate that during acute inflammation, treatment with statins inhibits leukocyte recruitment, independent of their cholesterol-lowering properties. These findings provide further mechanistic insights that could explain, in part, the great beneficial effects of statins described within a relatively short period of treatment, even in patients without hypercholesterolemia in secondary as well as primary prevention clinical trials to reduce morbidity and mortality.6 Because of the relatively high doses of statins required to achieve their anti-inflammatory effects, the development of novel inhibitors of nonsterol mevalonate-derived compounds that dissociate cholesterol-lowering effects from anti-inflammatory activities would be highly desirable. As we learn more about statins as anti-inflammatory agents in vitro and in a growing number of in vivo animal models, we await information that will establish their relevance in acute inflammation in humans. One can speculate that using animal models in in vivo experiments that block farnesylation or geranylgeranylation of cellular proteins will soon bring more information on how statin therapy, beyond its lipid-lowering effects, influences the inflammatory state of atherogenesis and its development. The high degree of patient tolerance of statins opens new drug strategies to inhibit nonsterol mevalonate-derived compounds that would make them potentially interesting agents to use in diseases characterized by acute (eg, sepsis, pancreatitis, pneumonia) or chronic inflammation, such as atherosclerosis or rheumatoid arthritis.

    Footnotes

    Correspondence to François Mach, MD, Cardiology Division, Foundation for Medical Research, 64 Avenue Roseraie, 1211 Geneva 4, Switzerland. E-mail

    References

    • 1 Lopez AD, Murray CC. The global burden of disease, 1990–2020. Nat Med. 1998; 4: 1241–1243.CrossrefMedlineGoogle Scholar
    • 2 Ross R. Atherosclerosis: an inflammatory disease. N Engl J Med. 1999; 340: 115–126.CrossrefMedlineGoogle Scholar
    • 3 Glass CK, Witztum JL. Atherosclerosis: the road ahead. Cell. 2001; 104: 503–516.CrossrefMedlineGoogle Scholar
    • 4 Lusis AJ. Atherosclerosis. Nature. 2000; 407: 233–241.CrossrefMedlineGoogle Scholar
    • 5 Libby P. Changing concepts of atherogenesis. J Intern Med. 2000; 247: 349–358.CrossrefMedlineGoogle Scholar
    • 6 Maron DJ, Fazio S, Linton MF. Current perspectives on statins. Circulation. 2000; 101: 207–213.LinkGoogle Scholar
    • 7 Casey PJ. Protein lipidation in cell signaling. Science. 1995; 268: 221–225.CrossrefMedlineGoogle Scholar
    • 8 Vaughan CJ, Murphy MB, Buckley BM. Statins do more than just lower cholesterol. Lancet. 1996; 348: 1079–1082.CrossrefMedlineGoogle Scholar
    • 9 Bellosta S, Ferri N, Bernini F, Paoletti R, Corsini A. Non-lipid-related effects of statins. Ann Med. 2000; 32: 164–176.CrossrefMedlineGoogle Scholar
    • 10 Davignon J, Laaksonen R. Low-density lipoprotein-independent effects of statins. Curr Opin Lipidol. 1999; 10: 543–559.CrossrefMedlineGoogle Scholar
    • 11 Laufs U, La Fata V, Plutzky J, Liao JK. Upregulation of endothelial nitric oxide synthase by HMG CoA reductase inhibitors. Circulation. 1998; 97: 1129–1135.CrossrefMedlineGoogle Scholar
    • 12 Laufs U, Liao JK. Post-transcriptional regulation of endothelial nitric oxide synthase mRNA stability by Rho GTPase. J Biol Chem. 1998; 273: 24266–24271.CrossrefMedlineGoogle Scholar
    • 13 Endres M, Laufs U, Huang Z, Nakamura T, Huang P, Moskowitz MA, Liao JK. Stroke protection by 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors mediated by endothelial nitric oxide synthase. Proc Natl Acad Sci U S A. 1998; 95: 8880–8885.CrossrefMedlineGoogle Scholar
    • 14 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: 21926–21931.CrossrefMedlineGoogle Scholar
    • 15 Soma MR, Donetti E, Parolini C, Mazzini G, Ferrari C, Fumagalli R, Paoletti R. HMG CoA reductase inhibitors: in vivo effects on carotid intimal thickening in normocholesterolemic rabbits. Arterioscler Thromb. 1993; 13: 571–578.CrossrefMedlineGoogle Scholar
    • 16 Guijarro C, Blanco-Colio LM, Ortego M, Alonso C, Ortiz A, Plaza JJ, Diaz C, Hernandez G, Edigo J. 3-Hydroxy-3-methylglutaryl coenzyme A reductase and isoprenylation inhibitors induce apoptosis of vascular smooth muscle cells in culture. Circ Res. 1998; 83: 490–500.CrossrefMedlineGoogle Scholar
    • 17 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: 683–690.CrossrefMedlineGoogle Scholar
    • 18 Aikawa M, Rabkin E, Voglic SJ, Shing H, Nagai R, Schoen FJ, Libby P. Lipid lowering promotes accumulation of mature smooth muscle cells expressing smooth muscle myosin heavy chain isoforms in rabbit atheroma. Circ Res. 1998; 83: 1015–1026.CrossrefMedlineGoogle Scholar
    • 19 Aikawa M, Rabkin E, Okada Y, Voglic SJ, Clinton SK, Brinckerhoff CE, Sukhova GK, Libby P. Lipid lowering by diet reduces matrix metalloproteinase activity and increases collagen content of rabbit atheroma: a potential mechanism of lesion stabilization. Circulation. 1998; 97: 2433–2444.CrossrefMedlineGoogle Scholar
    • 20 Fukumoto Y, Libby P, Rabkin E, Hill CC, Enomoto M, Hirouchi Y, Shiomi M, Aikawa M. Statins alter smooth muscle cell accumulation and collagen content in established atheroma of Watanabe heritable hyperlipidemic rabbits. Circulation. 2001; 103: 993–999.CrossrefMedlineGoogle Scholar
    • 21 Weber C, Erl W, Weber KS, Weber PC. HMG-CoA reductase inhibitors decrease CD11b expression and CD11b-dependent adhesion of monocytes to endothelium and reduce increased adhesiveness of monocytes isolated from patients with hypercholesterolemia. J Am Coll Cardiol. 1997; 30: 1212–1217.CrossrefMedlineGoogle Scholar
    • 22 Liu L, Moesner P, Kovach NL, Bailey R, Hamilton AD, Sebti SM, Harlan JM. Integrin-dependent leukocyte adhesion involves geranylgeranylated protein(s). J Biol Chem. 1999; 274: 33334–33340.CrossrefMedlineGoogle Scholar
    • 23 Romano M, Diomede L, Sironi M, Massimiliano L, Sottocorno M, Polentarutti N, Guglielmotti A, Albani D, Bruno A, Fruscella P, Salmona M, Vecchi A, Pinza M, Mantovani A. Inhibition of monocyte chemotactic protein-1 synthesis by statins. Lab Invest. 2000; 80: 1095–1100.CrossrefMedlineGoogle Scholar
    • 24 Kwak B, Mulhaupt F, Myit S, Mach F. Statins as a newly recognized type of immunomodulator. Nat Med. 2000; 6: 1399–1403.CrossrefMedlineGoogle Scholar
    • 25 Kwak B, Mulhaupt F, Veillard N, Pelli G, Mach F. The HMG-CoA reductase inhibitor simvastatin inhibits IFN-γ induced MHC class II expression in human vascular endothelial cells. Swiss Med Wkly. 2001; 131: 41–46.MedlineGoogle Scholar
    • 26 Palinsky W. Immunomodulation: a new role for statins? Nat Med. 2000; 6: 1311–1312.CrossrefMedlineGoogle Scholar
    • 27 Diomede L, Albani D, Sottocorno M, Polentarutti N, Donati MB, Bianchi M, Fruscella P, Salmona M. The in vivo anti-inflammatory effect of statins is mediated by nonsterol mevalonate products. Arterioscler Thromb Vasc Biol. 2001; 21: 1327–1332.CrossrefMedlineGoogle Scholar
    • 28 Yoshida M, Sawada T, Ishii H, Gerszten RE, Rosenzweig A, Gimbrone MA Jr, Yasukochi Y, Numano F. HMG-CoA reductase inhibitor modulates monocyte endothelial interaction under physiological flow condition in vitro: involvement of Rho GTPase-dependent mechanism. Arterioscler Thromb Vasc Biol. 2001; 21: 1165–1171.CrossrefMedlineGoogle Scholar
    • 29 Sparrow CP, Burton CA, Hernandez M, Mundt S, Hassing H, Patel S, Rosa R, Hermanowski-Vosatka A, Wang PR, Zhang D, Peterson L, Detmers PA, Chao YS, Wright SD. Simvastatin has anti-inflammatory and antiatherosclerotic activities independent of plasma cholesterol lowering. Arterioscler Thromb Vasc Biol. 2001; 21: 115–121.CrossrefMedlineGoogle Scholar
    • 30 Wojciak-Stothard B, Williams L, Ridley AJ. Monocyte adhesion and spreading on human endothelial cells is dependent on Rho-regulated receptor clustering. J Cell Biol. 1999; 145: 1293–1307.CrossrefMedlineGoogle Scholar
    • 31 Stalker TJ, Lefer AM, Scalia R. A new HMG-CoA reductase inhibitor, rosuvastatin, exerts anti-inflammatory effects on the microvascular endothelium: the role of mevalonic acid. Br J Pharmacol. 2001; 133: 406–412.CrossrefMedlineGoogle Scholar