Abstract
Statins, long known to be beneficial in conditions where dyslipidemia occurs by lowering serum cholesterol levels, also have been proposed for use in neurodegenerative conditions, including Alzheimer disease. However, it is not clear that the purported effectiveness of statins in neurodegenerative disorders is directly related to cholesterol-lowering effects of these agents; rather, the pleiotropic functions of statins likely play critical roles.
Moreover, it is becoming more apparent with additional studies that statins can have deleterious effects in preclinical studies and lack effectiveness in various recent clinical trials. This perspective paper outlines pros and cons of the use of statins in neurodegenerative disorders, with particular emphasis on Alzheimer disease.
1. Basic pharmacology of statins
Statins are a family of drugs with pleiotropic functions. To this class belong 8 drugs: mevastatin and lovastatin, which were the first developed and studied in humans; pravastatin and simvastatin, which can be considered as derivatives of the parental lovastatin; and atorvastatin, fluvastatin, rosuvastatin and pitavastatin, which are distinct synthetic compounds [1]. Due to their main mechanism of action, namely the inhibition of the hydroxyl-methyl-glutaryl-CoA (HMG-CoA) reductase, statins are widely used for the treatment of dyslipidemias [1]. By inhibiting HMG-CoA reductase, statins block the conversion of HMG-CoA into mevalonate, the first step in cholesterol biosynthesis [1,2]. As a result of statin administration, low-density lipoprotein (LDL)-cholesterol synthesis decreases in hepatocytes and this reflects a reduced cholesterol blood level. In addition to this effect, statins have been shown to reduce triglyceride and increase HDL-cholesterol plasma levels. Taken together, the composite effect of statins in reducing triglycerydes and LDL-cholesterol, coupled with the increase in HDL-cholesterol, put these drugs in the arena of cardiovascular agents, due to their ability to counteract hyperlipidemias, the major cause of atherosclerosis which, in turn, is a common pathogenetic mechanism for coronary artery disease, ischemic cerebrovascular disease and peripheral vascular disease [1,2].
Although statins share the same main mechanism of action, their pharmacokinetic profile is quite different (Table 1). All statins are well absorbed by the intestine when given by orally, even though they undergo marked first-pass effects by the liver, which reduces the systemic biovailability (5–30%) [1]. With the exception of simvastatin and lovastatim, which are pro-drugs and require hepatic activation, other statins are administered as β-hydroxy-acids. Upon administration, statins reach peak plasma concentration, ranging from 10–448 ng/ml, within 0.5–4 h. In the plasma, statins are bound to albumin (43–99%) and this binding accounts for their variable half-life [1]. Atorvastatin and rosuvastatin are the statins with the longest half-life (15–30 and 20.8 h, respectively), whereas fluvastatin, lovastatin, pravastatin and simvastatin have half-lives around 0.5–3 h [1]. All statins are metabolized by the liver through the isoforms 3A4 (atorvastatin, lovastatin and simvastatin) and 2C9 (fluvastatin and rosuvastatin) of the cytochrome-P-450 (CYP) system, whereas pravastatin undergoes sulfation. The primary route of elimination is fecal, and only a minor fraction of statins is eliminated via urine [1,2].
Table 1.
Pharmacokinetic parameters of statins.
| Tmax (h) |
Cmax (ng/mL) |
B (%) |
L | Pb (%) |
M | T1/2 (h) |
E (%) |
||
|---|---|---|---|---|---|---|---|---|---|
| Atorvastatin | ![]() |
2–3 | 27–66 | 12 | Yes | 80–90 | CYP3A4 | 15–30 | Urinary 2 Fecal 70 |
| Fluvastatin | ![]() |
0.5–1 | 448 | 19–29 | Yes | >99 | CYP2C9 | 0.5–2.3 | Urinary 6 Fecal 90 |
| Fluvastatin XL | 4 | 55 | 6 | Yes | >99 | CYP2C9 | 4.7 | Urinary 6 Fecal 90 |
|
| Lovastatin | ![]() |
2–4 | 10–20 | 5 | Yes | >95 | CYP3A4 | 2.9 | Urinary 10 Fecal 83 |
| Pitavastatin | ![]() |
1.2 | 41 | ~ 80 | Yes | 96 | CYP2C9 | 11 | Urinary < 2 Fecal 90 |
| Pravastatin | ![]() |
0.9–1.6 | 45–55 | 18 | No | 43–55 | Sulfation | 1.3–2.8 | Urinary 20 Fecal 71 |
| Rosuvastatin | ![]() |
3 | 37 | 20 | No | 88 | CYP2C9 | 20.8 | Urinary 10 Fecal 90 |
| Simvastatin | ![]() |
1.3–2.4 | 10–34 | 5 | Yes | 94–98 | CYP3A4 | 2–3 | Urinary 13 Fecal 58 |
The main adverse effects of statins are hepatotoxicity and myopathy. A transient elevation of serum transaminases (up to 3-times the baseline value) is a common outcome of statin therapy [2]. However, the incidence of this side effect is low and dose-dependent and does not imply the contraindication of statins in individuals with concomitant liver diseases such as hepatitis C [2]. Myalgia is often associated with statin use and is paralleled by an increase in plasma creatine kinase up to 10 times [2]. Rhabdomyolisis is quite rare, and the risk to develop this side effect of statins is correlated to the dose and plasma concentration [2]. About 30 cases of serious hepatic failure and 42 cases of death due to rhabdomyolisis associated with statin administration were reported to the FDA over the last 15 years [2,3]. In order to reduce the incidence of hepatotoxicity and myopathy, statins should not be associated with inhibitors of CYP3A4 such as azole antifungals, erythromycin, ritonavir and grapefruit juice. Also the association statins and fibrates should be avoided, in particular gemfibrozil [2].
2. Pleiotropic effects of statins
The debate about statin treatment regards the mechanism of action by which statins mediate their potentially beneficial effects. In particular, are these benefits due to the well known ability of statins to lower cholesterol or to their so called pleiotropic effects [4–6]? Statins can modulate several cellular pathways, independent of their ability to inhibit HMG-CoA reductase. These processes include isoprenylation and myelination, modulation of immune response, and effects on oxidative and nitrosative stress levels.
Isoprenylation
The isoprenylation process is significantly and directly affected by HMG-CoA reductase activity. Through the inhibition of HMG-CoA reductase activity, statins reduce the formation of L-mevalonic acid and subsequent prevention of isoprenoid synthesis [4,7,8]. The seventh step of the cholesterol synthesis yields farnesyl pyrophosphate (FPP), which can be converted into squalene and hence to cholesterol, but is also used for the production of geranylgeranyl pyrophosphate (GGPP). Both FPP and GGPP are required to enable proper subcellular localization and trafficking of intracellular proteins [4,9]. FPP is also the substrate for production of coenzyme Q10 (CoQ10) and dolichol [10]. Coenzyme Q10 is an antioxidant, and dolichol may function as a radical scavenger. Of these two, only CoQ10 has been seriously studied in relation to statin treatment: statins decrease CoQ10 levels in plasma and tissue, which may be responsible for several of statins’ side effects [11,12]. Farnesyl pyrophosphate and GGPP on the other hand have received much attention as possible mediators of the non-cholesterol-dependent effects of statins [5,13]. In particular, small GTP-binding proteins, including members of the Ras and Rho GTPase family, require prenylation post-translational modifications to function as modulators of the actin cytoskeleton and to participate in intracellular signaling [9]. However, depending on statin type, length of treatment, and cellular type the final outcome could be different. First of all, statins exhibit inhibitory effects on small G proteins, by altering the isoprenylation process [4,14]. Neuroprotective effects of simvastatin for example, include the improvement of behavioral function associated with an inhibition of Rho-associated kinase (ROCK) in a rabbit model of ischemic stroke [15], and the prevention of dopaminergic neuronal loss trough the inhibition of p21(ras)-induced NF-kappaB increase in a mouse model of Parkinson’s disease [16] (Figure 1). Conversely, prolonged treatment of human mature oligodentrocyte with simvastatin was associated with cell death [17] (Figure 1). Moreover, evidence for decreased learning and memory following long-term simvastatin treatment exists [18]. Lovastatin, was effective to ameliorate experimental autoimmune encephalomyelitis by reducing the activities of Rho/Ras family GTPase in glial cells [19]. In contrast, the same drugs efficiently inhibited Ras-induced ERK1/2 phospshorylation in rat brain neuroblasts [20], as well as geranylgeranylation of Rho family GTPase in neurons [21], leading to an increase in apotosis and tau phosphorylation, respectively [20,21] (Figure 1). Noting that inflammatory processes occur in several neurodegenerative disorders, such as Alzheimer disease, statin-mediated inhibition of Rho GTPases seems to attenuate beta-amyloid (Aβ) peptide-stimulated inflammation in microglia [8]. Moreover, mevastatin and lovastatin were reported to, respectively, decrease and increase neurite outgrowth in different strains of PC12 cells through different mechanisms [22,23].
Figure 1.
Cholesterol-independent neuroprotective versus neurotoxic effects of statins. Black arrows: stimulation; dotted lines: inhibition.
Downstream consequences of reduced isoprenoid synthesis may include changes in vascular function, modulation of the insulin/phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) pathway [24] and possibly a reduction in reactive oxygen species production [25]. Thus, statin-mediated reduction of protein isoprenylation could have widespread effects on protein transport, trafficking, mRNA stability and gene transcription [4]. However, the clinical significance of the results discussed above is debatable. The question whether inhibition of isoprenylation process occurs in the brain therefore remains open.
Myelination
Although initially statins were thought to be beneficial for myelination and proposed as a treatment for multiple sclerosis (MS), a demyelinating condition [26,27], these benefits are being re-evaluated, since contradictory results have been described. As for isoprenylation processes, both in vitro and in vivo results suggest that statin- and cell type play pivotal roles in the final outcome. Simvastatin treatment had a detrimental effect on oligodentrocyte outgrowth, a key step in the re(myelination) process [28], particularly in the early myelination stage [29] (Figure 1). Interestingly, simvastatin and lovastatin had an opposite effect on the myelin basic protein (MBP), since simvastatin treatment greatly increased the densities of MBP in oligodentrocytes of neonatal rats after hypoxia-ischemia damage [30], whereas lovastatin reduced MBP expression in primary oligodentrocytes, probably by impairment of the isoprenylation process [31] (Figure 1).
Immunomodulatory effects
The immune response plays a role in neurodegeneration, not only in MS, but also in Alzheimer and Parkinson diseases [32]. Statins may have immunomodulatory effects, which also could be mediated through reduced protein prenylation, although, so far, no singular mechanism has been proposed [13]. In vitro, lovastatin prevents expression of TNF and IL-1β [33], and prubocol reduces glial activation [34]. In addition, microglial cultures exposed to atorvastatin and simvastatin showed reduced levels of the pro-inflammatory cytokine, IL-6 [35] (Figure 1). However, the effects of statins on the immune cells of the CNS, the microglia, have received little attention and need to be further explored.
Oxidative and nitrosative stress
Another intriguing aspect related to the pleiotropic effects induced by statin treatment regards the modulation of oxidative stress-related modifications that occur in neurodegenerative disorders [36]. Statins can inhibit endothelial O2−· formation by preventing the isoprenylation of p21 Rac, which is critical for the assembly of NADPH oxidase after activation of PKC [37] (Figure 1). In addition, SOD3 activity was more than doubled by simvastatin, and simvastatin treatment also increased the number of functionally active endothelial progenitor cells [38] (Figure 1). Moreover, statins increase the expression of endothelial nitric oxide synthase (eNOS) by inhibition of Rho isoprenylation [39], and statins can also directly activate eNOS via post-translational mechanisms involving activation of the PI3K/Akt pathway [40] (Figure 1). Statins showed positive effects against Alzheimer-relevant Aβ-induced oxidative stress in mice models of AD [41,42] as well as a reduction in CSF tau protein phosphorylation in humans [43]. However, although statin treatment appears to provide greater benefits, it is difficult to tease out whether the benefits are really due to lower cholesterol levels or to statin pleiotropy [6]. Atorvastatin treatment was neuroprotective against cell degeneration induced by Aβ(1–40), reducing inflammatory and oxidative responses and increasing the expression of glutamatergic transporters [44]. Murphy et al. (2010) showed that long-term atorvastatin did not affect Aβ levels, despite a significant reduction in β-secretase 1 (BACE1) protein levels and activity in the brain of aged beagles [45]. Later, we found that although no change in Aβ levels occur, long-term atorvastatin significantly reduced lipoperoxidation, protein oxidation and nitration, and increased GSH levels in parietal cortex of the same animals [46] (Figure 1). This effect was cholesterol- and Aβ-independent and specific for brain [46]. Conversely, side effects of long-term statin treatment include a decrease in CoQ10 levels resulting in energy metabolism impairment in heart, skeletal muscle, and liver [47] (Figure 1). Supplementation of the diet with CoQ10 was reported to reverse many of these alterations [48]. At the same time, the effect of lipophilic statins can result in elevated tissue oxidative stress through NO reacting with metabolically derived O20˙− to form peroxynitrite and other reactive oxidants, which can have negative effects on endothelial cells [49] (Figure 1).
Cholesterol oxidation products
Statin effects on oxidative stress should take into account cholesterol reduction, considering that cholesterol itself, can be oxidized with likely loss of its functions. Cholesterol can undergo oxidative modifications at least by two mechanisms: a direct radical attack involving ROS or RNS (non-enzymatic mechanism), or by the activity of a specific enzymes (enzymatic mechanism). This leads to the formation of cholesterol oxidation products, i.e., oxysterols. These latter moieties are major regulators of cholesterol homeostasis in the central nervous system [50]. Among oxysterols, 7-ketocholesterol (7-K) and 25-hydroxycholesterol (25-OH) have been shown to cause apoptotic neuronal death by inducing mitochondrial dysfunction [51], Ca2+ influx and perturbation of intracellular ionic homeostasis [52,53].
Although some evidence suggests the importance of cholesterol oxidation products both as in vivo markers of oxidative stress [54–56], as well as for their pro-oxidant features [51–53,57], few studies exist regarding the effect of statins on cholesterol oxidation products in vivo [54,58,59]. Our group showed that atorvastatin can have two independent effects on cholesterol and cholesterol oxidation products, since a reduction of cholesterol was not associated with a reduction of 7-K or 25-OH and vice versa. In fact, the levels of both 7-K and 25-OH were reduced in brain, while 7-K levels were significantly increased in serum in dogs receiving atorvastatin [46] (Figure 1).
These results, together with those showing a marked peripheral reduction of CoQ10 after long-term treatment with statins, suggest that statins can exert antioxidant/pro-oxidant effects depending on the site of action and on the mechanisms modulated. Due to duration of statin treatment, it would be interesting to carry out in vivo studies to analyze in the brain changes that occur to cholesterol oxidation products and CoQ10. Can these changes to be correlated? Do statins decrease CoQ10 in the brain? Is reduction/increase of CoQ10 associated with different levels of oxysterols?
We believe that more detailed research into the pharmacology of statins, particularly the concentrations achieved in the CNS and the level at which they block the production of cholesterol and they modulate all the above pathways, may prove beneficial to better understating of the potential use of statins in neurodegenerative disorders.
3. Statins and dementia: suggestion to use evidence-based medicine as a basis of future studies
Although the several lines of preclinical evidence showed neuroptrotective effects of statins in ameliorating cognitive dysfunction, clinical data largely have not supported such a conclusion. In addition, clinical studies show opposite results depending whether or not they were observational studies or randomized clinical trials (RCT).
Early clinical data based on cohort and case-control studies, demonstrated that statins reduced the risk to develop dementia, including Alzheimer disease (AD), and this protective effect was maintained over a 6 year follow-up period [60–67]. These findings were recently contradicted by Benito-Leon et al., who demonstrated that statins did not improve cognition in elderly subjects with a median age of 72 years [68]. Similar results, were obtained in a large cohort study, which involved more than 2 million subjects aged 30–84 years of whom 10.7% received statins [69]. Only a weak improvement in cognitive performance (evaluated by the Hopkins word list and Rivermead paragraph) was found in individuals with mild cognitive impairment, arguably the earliest form of AD, treated with statins [70].
In order to confirm these epidemiological studies, some RCT were performed with comparable outcomes. The PROSPER study, which involved about 6,000 people aged 70–82 years, demonstrated that pravastatin (40 mg/day) did not improve cognitive function over a follow-up of 3 years [71,72]. The LEADe study tested the hypothesis that atorvastatin (80 mg/day) over 72 weeks delayed cognitive decline in subjects with mild-moderate AD. The results of this study, did not support any significant positive effect of atorvastatin on cognitive or global functions in patients receiving the statin compared to those with placebo [73]. In 2008 the CLASP study was designed to evaluate the neuroprotective role of simvastatin (20–40 mg/day) for 18 months in mild-moderate AD patients [74]. The results of this trial, not yet published, failed to demonstrate a therapeutic role for simvastatin [74]. On the other hand, the ADCLT trial demonstrated that atorvastatin (80 mg/day) for 1 year exhibited a significant positive effect on cognitive performance after 6 months of therapy compared with placebo. However, this beneficial effect was narrowed to individuals who matched restricted criteria, such as a higher MMSE score at the baseline, total cholesterol levels higher than 200 mg/dl and the presence of an apolipoprotein-E-4 allele [75,76]. With the purpose to put together and analyze the results from all the RCT trials about statins and dementia, McGuinness et al. performed a meta-analysis and concluded that there is not evidence strong enough to recommend statins for the treatment of dementia and AD [77]. This statement agrees with the guidelines of the British Association for Psychopharmacology who do not recommend statins for the prevention or treatment of AD [78].
Pitfalls
The conflicting results described above prompt the question, “what are the reasons why statins had such inconsistent beneficial effects in aged or demented individuals?” Even if it is not possible to single out the main drawback, some criticisms need to be addressed and carefully evaluated.
Ability to cross membranes
Due to different chemical structures, different statins exhibit a variable degree of lipophilicity. The prodrugs simvastatin and lovastatin have the the highest logD (index of lipophylicity), atorvastatin, fluvastatin and pitavastatin an intermediate logD while pravastatin has the lowest [1]. Such considerations might suggest use of lipophilic statins in demented patients to increase the fraction that reaches the brain. However, both the LEADe and CLASP trial failed to demonstrate a beneficial effects of atorvastatin and lipophilic simvastatin on cognitive function in AD subjects, consistent with those obtained in the PROSPER study performed with lipophobic pravastatin. Therefore, the different degree of liposolubility likely is not the key determinant that limited effectiveness of statins in these clinical trials.
Age
Epidemiological data demonstrate the incidence of AD increases with age and doubles every 5 years after 65 years of age with 1275 new cases/100000 persons/year [79]. In the Western hemisphere, the prevalence of AD was calculated as about 1% in subjects aged 60–64 but increased to between 33% and 50% in people aged 85 or older [80]. Keeping this is mind, both cohort studies and RCT that examined the role of statins in AD were based on populations aged 65–84 years. Although the clinical studies and the meta-analysis discussed above do not support an overall beneficial effect for statins in dementia and AD, it is noteworthy that those studies in which statins had a major effect on cognitive functions recruited individuals aged 68–74 years [61,63,81]. Consequently, it is possible that 68–74 years of age should not be considered as a “threshold” or upper limit for statin efficacy in preventing dementia, since both the LEADe and PROSPER studies enrolled individuals within the same range of age, and these studies failed to demonstrate any beneficial effect of statins in people with AD. Subjects aged 80 or older also did not have any beneficial effects from statins [61].
Cholesterol blood levels at baseline
An important issue to consider when giving a statin is the degree of reduction of cholesterol levels in serum. Recalling that cholesterol is a main component of cell membranes, in particular myelin [82], if cholesterol blood levels fall due to uncontrolled therapy with lipid lowering agents, nervous function would also decrease. An increase in total blood cholesterol levels at midlife age was associated with an increased risk to develop AD [83,84]. As summarized by McGuinnes et al. in two recent meta-analyses, AD subjects recruited for large clinical trials had serum LDL cholesterol around 131–147 mg/dL [85]. The same authors reported that after the administration of atorvastatin (80 mg) for 52 weeks or simvastatin (40 mg) for 26 weeks a reduction of LDL-cholesterol of 50–54% in AD patients was observed [77,85]. These values of LDL-cholesterol, before and after statin treatment, are still acceptable and do not imply any possible adverse effects. However, although there was beneficial effect of statins on LDL-cholesterol plasma levels, no beneficial effect on cognitive function was observed [77,85]. Even in hypercholesterolemic patients, statins did not reduce Aβ in both plasma and cerebrospinal fluid (CSF), suggesting the lack of any statin-mediated on Aβ deposition or clearance [43,86]. Considering that an excessive reduction in cholesterol plasma levels is not advisable, statins could not be administered to AD patients with low cholesterol plasma levels such as those affected by liver failure. Evans et al. showed that in AD patients heterozygous for APOE4 allele or carriers of PS1 mutations, the administration of simvastatin or atorvastatin slightly reduced the concentration of CSF-cholesterol at 6–7 months followed by a peak at 2 years and a return to baseline levels after three years [87]. This finding lends support to the idea that, despite the changes in plasma cholesterol levels, only minimal changes in brain cholesterol occur after statin therapy and, therefore, the effect on cognitive functions are independent of the “local” cholesterol metabolism.
Interaction with xenobiotics
Patients with AD, as well as other types of dementia, usually take other drugs for other age-related disorders or co-morbitites associated with AD. As mentioned above, all statins, with the exception of pravastatin, are metabolized by CYP3A4 or CYP2C9, and their plasma levels could be reduced or increased in the case of concomitant administration of drugs that induce or inhibit these CYP isoforms. Drugs used in AD, such as donepezil and galantamine, also are metabolized by CYP3A4 [88], and, therefore, could compete with statins. As a consequence of this interaction, statin plasma levels could increase as well as the risk of side effects. Also, AD patients can often be supplemented with dietary products including curcumin, grapefruit juice and green tea as these natural substances are widely considered as free radical scavenger and therefore neuroprotective. Unfortunately, these natural substances are inhibitors of CYP3A4 and, therefore, increase plasma concentrations of statins [89–91]. Interestingly, it was reported that the concomitant assumption of simvastatin and grapefruit or green tea originated rhabdomyolisis [92,93].
Taken together, the results from evidence-based medicine suggest that the ideal AD subject with some possibility to have an increased cognitive performance upon treatment with statins is aged 65–74 years, harbors an ApoE-4 allele [76], is normocholesterolemic and sparingly uses drugs that inhibit CYP3A4.
Conclusions
Although the evidence provided with preclinical data substantiated the potential beneficial effects of statins in dementia and AD, the results provided by epidemiological studies and RCT are quite contradictory. Several criticisms related to the rate of inhibition of cholesterol synthesis in normocholesterolemic patients, age and concomitant administration of drug or dietary supplements also should be considered. Increased selectively in RCT involving AD subjects aged 65–75 years, the age which seems to receive more benefit from statin therapy, in which the primary end-point is the careful analysis of cognitive function through a battery of neurological tests, should be carried out. Another recommendation to consider is to study in-depth the effect of statins in individuals with mild cognitive impairment in order to understand whether or not the antioxidant effect of these drugs could block or slow the transition phase to AD. This recommendation could be especially important since preclinical studies with atorvastatin decreased brain-resident oxidative stress in beagle dogs, and this benefit was correlated with decreased levels of Aβ(1–42), which has the same sequence as that of humans [46]. Of course, oxidative stress is strongly associated with amnestic MCI and AD [94–96], and Aβ is hypothesized to contribute to this oxidative stress [97].
In conclusion, our opinion is that current clinical evidence is not strong enough to support the widespread use of statins to treat dementia and AD. However, strong consideration for researchers and clinicians in the near future to investigate whether or not statin therapy should be restricted to selected populations of demented individuals with the best chance of efficacy derived from evidence-based medicine is recommended.
ACKNOWLEDGEMENTS
This work was supported in part by a NIH grant to D.A.B. [AG-05119].
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Shitara Y, Sugiyama Y. Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme a (hmg-coa) reductase inhibitors: Drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions. Pharmacol Ther. 2006;112:71–105. doi: 10.1016/j.pharmthera.2006.03.003. [DOI] [PubMed] [Google Scholar]
- 2.Bersot TP. Drug therapy for hypercholesterolemia and dyslipidemia. In: Laurence LB, editor. Goodman and gilman's, the pharmacological basis of therapeutics. McGraw-Hill; 2011. pp. 877–908. [Google Scholar]
- 3.Law M, Rudnicka AR. Statin safety: A systematic review. Am J Cardiol. 2006;97:52C–60C. doi: 10.1016/j.amjcard.2005.12.010. [DOI] [PubMed] [Google Scholar]
- 4.Wang CY, Liu PY, Liao JK. Pleiotropic effects of statin therapy: Molecular mechanisms and clinical results. Trends Mol Med. 2008;14:37–44. doi: 10.1016/j.molmed.2007.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Liao JK, Laufs U. Pleiotropic effects of statins. Annu Rev Pharmacol Toxicol. 2005;45:89–118. doi: 10.1146/annurev.pharmtox.45.120403.095748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Butterfield DA. Atorvastatin and Abeta(1–40): Not as simple as cholesterol reduction in brain and relevance to alzheimer disease. Exp Neurol. 2011;228:15–18. doi: 10.1016/j.expneurol.2010.12.014. [DOI] [PubMed] [Google Scholar]
- 7.Cole SL, Vassar R. Isoprenoids and Alzheimer's disease: A complex relationship. Neurobiol Dis. 2006;22:209–222. doi: 10.1016/j.nbd.2005.11.007. [DOI] [PubMed] [Google Scholar]
- 8.Cordle A, Koenigsknecht-Talboo J, Wilkinson B, Limpert A, Landreth G. Mechanisms of statin-mediated inhibition of small g-protein function. J Biol Chem. 2005;280:34202–34209. doi: 10.1074/jbc.M505268200. [DOI] [PubMed] [Google Scholar]
- 9.McTaggart SJ. Isoprenylated proteins. Cell Mol Life Sci. 2006;63:255–267. doi: 10.1007/s00018-005-5298-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Corsini A, Bellosta S, Baetta R, Fumagalli R, Paoletti R, Bernini F. New insights into the pharmacodynamic and pharmacokinetic properties of statins. Pharmacol Ther. 1999;84:413–428. doi: 10.1016/s0163-7258(99)00045-5. [DOI] [PubMed] [Google Scholar]
- 11.Littarru GP, Langsjoen P. Coenzyme q10 and statins: Biochemical and clinical implications. Mitochondrion. 2007;7 Suppl:S168–S174. doi: 10.1016/j.mito.2007.03.002. [DOI] [PubMed] [Google Scholar]
- 12.Marcoff L, Thompson PD. The role of coenzyme q10 in statin-associated myopathy: A systematic review. J Am Coll Cardiol. 2007;49:2231–2237. doi: 10.1016/j.jacc.2007.02.049. [DOI] [PubMed] [Google Scholar]
- 13.Greenwood J, Steinman L, Zamvil SS. Statin therapy and autoimmune disease: From protein prenylation to immunomodulation. Nat Rev Immunol. 2006;6:358–370. doi: 10.1038/nri1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.van der Most PJ, Dolga AM, Nijholt IM, Luiten PG, Eisel UL. Statins: Mechanisms of neuroprotection. Prog Neurobiol. 2009;88:64–75. doi: 10.1016/j.pneurobio.2009.02.002. [DOI] [PubMed] [Google Scholar]
- 15.Lapchak PA, Han MK. Simvastatin improves clinical scores in a rabbit multiple infarct ischemic stroke model: Synergism with a rock inhibitor but not the thrombolytic tissue plasminogen activator. Brain Res. 2010;1344:217–225. doi: 10.1016/j.brainres.2010.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ghosh A, Roy A, Matras J, Brahmachari S, Gendelman HE, Pahan K. Simvastatin inhibits the activation of p21ras and prevents the loss of dopaminergic neurons in a mouse model of parkinson's disease. J Neurosci. 2009;29:13543–13556. doi: 10.1523/JNEUROSCI.4144-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Miron VE, Rajasekharan S, Jarjour AA, Zamvil SS, Kennedy TE, Antel JP. Simvastatin regulates oligodendroglial process dynamics and survival. Glia. 2007;55:130–143. doi: 10.1002/glia.20441. [DOI] [PubMed] [Google Scholar]
- 18.Wagstaff LR, Mitton MW, Arvik BM, Doraiswamy PM. Statin-associated memory loss: Analysis of 60 case reports and review of the literature. Pharmacotherapy. 2003;23:871–880. doi: 10.1592/phco.23.7.871.32720. [DOI] [PubMed] [Google Scholar]
- 19.Paintlia AS, Paintlia MK, Singh AK, Singh I. Inhibition of rho family functions by lovastatin promotes myelin repair in ameliorating experimental autoimmune encephalomyelitis. Mol Pharmacol. 2008;73:1381–1393. doi: 10.1124/mol.107.044230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cerezo-Guisado MI, Garcia-Roman N, Garcia-Marin LJ, Alvarez-Barrientos A, Bragado MJ, Lorenzo MJ. Lovastatin inhibits the extracellular-signal-regulated kinase pathway in immortalized rat brain neuroblasts. Biochem J. 2007;401:175–183. doi: 10.1042/BJ20060731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Meske V, Albert F, Richter D, Schwarze J, Ohm TG. Blockade of hmg-coa reductase activity causes changes in microtubule-stabilizing protein tau via suppression of geranylgeranylpyrophosphate formation: Implications for Alzheimer's disease. Eur J Neurosci. 2003;17:93–102. doi: 10.1046/j.1460-9568.2003.02433.x. [DOI] [PubMed] [Google Scholar]
- 22.Nusse O, Neer EJ. Localization of g alpha 0 to growth cones in pc12 cells: Role of g alpha 0 association with receptors and g beta gamma. J Cell Sci. 1996;109(Pt 1):221–228. doi: 10.1242/jcs.109.1.221. [DOI] [PubMed] [Google Scholar]
- 23.Qiu MS, Pitts AF, Winters TR, Green SH. Ras isoprenylation is required for ras-induced but not for ngf-induced neuronal differentiation of pc12 cells. J Cell Biol. 1991;115:795–808. doi: 10.1083/jcb.115.3.795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Farah S, Agazie Y, Ohan N, Ngsee JK, Liu XJ. A rho-associated protein kinase, rokalpha, binds insulin receptor substrate-1 and modulates insulin signaling. J Biol Chem. 1998;273:4740–4746. doi: 10.1074/jbc.273.8.4740. [DOI] [PubMed] [Google Scholar]
- 25.Sundaresan M, Yu ZX, Ferrans VJ, Sulciner DJ, Gutkind JS, Irani K, Goldschmidt-Clermont PJ, Finkel T. Regulation of reactive-oxygen-species generation in fibroblasts by rac1. Biochem J. 1996;318(Pt 2):379–382. doi: 10.1042/bj3180379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Paintlia AS, Paintlia MK, Singh I, Skoff RB, Singh AK. Combination therapy of lovastatin and rolipram provides neuroprotection and promotes neurorepair in inflammatory demyelination model of multiple sclerosis. Glia. 2009;57:182–193. doi: 10.1002/glia.20745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Weber MS, Youssef S, Dunn SE, Prod'homme T, Neuhaus O, Stuve O, Greenwood J, Steinman L, Zamvil SS. Statins in the treatment of central nervous system autoimmune disease. J Neuroimmunol. 2006;178:140–148. doi: 10.1016/j.jneuroim.2006.06.006. [DOI] [PubMed] [Google Scholar]
- 28.Smolders I, Smets I, Maier O, vandeVen M, Steels P, Ameloot M. Simvastatin interferes with process outgrowth and branching of oligodendrocytes. J Neurosci Res. 2010;88:3361–3375. doi: 10.1002/jnr.22490. [DOI] [PubMed] [Google Scholar]
- 29.Xiang Z, Reeves SA. Simvastatin induces cell death in a mouse cerebellar slice culture (csc) model of developmental myelination. Exp Neurol. 2009;215:41–47. doi: 10.1016/j.expneurol.2008.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Li A, Lv S, Yu Z, Zhang Y, Ma H, Zhao H, Piao H, Li S, Zhang N, Sun C. Simvastatin attenuates hypomyelination induced by hypoxia-ischemia in neonatal rats. Neurol Res. 2010;32:945–952. doi: 10.1179/016164110X12670144737774. [DOI] [PubMed] [Google Scholar]
- 31.Maier O, De Jonge J, Nomden A, Hoekstra D, Baron W. Lovastatin induces the formation of abnormal myelin-like membrane sheets in primary oligodendrocytes. Glia. 2009;57:402–413. doi: 10.1002/glia.20769. [DOI] [PubMed] [Google Scholar]
- 32.Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T. Inflammation and Alzheimer's disease. Neurobiol Aging. 2000;21:383–421. doi: 10.1016/s0197-4580(00)00124-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pahan K, Sheikh FG, Namboodiri AM, Singh I. Lovastatin and phenylacetate inhibit the induction of nitric oxide synthase and cytokines in rat primary astrocytes, microglia, and macrophages. J Clin Invest. 1997;100:2671–2679. doi: 10.1172/JCI119812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Champagne D, Pearson D, Dea D, Rochford J, Poirier J. The cholesterol-lowering drug probucol increases apolipoprotein e production in the hippocampus of aged rats: Implications for Alzheimer's disease. Neuroscience. 2003;121:99–110. doi: 10.1016/s0306-4522(03)00361-0. [DOI] [PubMed] [Google Scholar]
- 35.Lindberg C, Crisby M, Winblad B, Schultzberg M. Effects of statins on microglia. J Neurosci Res. 2005;82:10–19. doi: 10.1002/jnr.20615. [DOI] [PubMed] [Google Scholar]
- 36.Orr JD. Statins in the spectrum of neurologic disease. Curr Atheroscler Rep. 2008;10:11–18. doi: 10.1007/s11883-008-0003-5. [DOI] [PubMed] [Google Scholar]
- 37.Wallerath T, Poleo D, Li H, Forstermann U. Red wine increases the expression of human endothelial nitric oxide synthase: A mechanism that may contribute to its beneficial cardiovascular effects. J Am Coll Cardiol. 2003;41:471–478. doi: 10.1016/s0735-1097(02)02826-7. [DOI] [PubMed] [Google Scholar]
- 38.Landmesser U, Bahlmann F, Mueller M, Spiekermann S, Kirchhoff N, Schulz S, Manes C, Fischer D, de Groot K, Fliser D, Fauler G, Marz W, Drexler H. Simvastatin versus ezetimibe: Pleiotropic and lipid-lowering effects on endothelial function in humans. Circulation. 2005;111:2356–2363. doi: 10.1161/01.CIR.0000164260.82417.3F. [DOI] [PubMed] [Google Scholar]
- 39.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. doi: 10.1161/01.cir.97.12.1129. [DOI] [PubMed] [Google Scholar]
- 40.Kureishi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer DJ, Sessa WC, Walsh K. The hmgcoa reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nat Med. 2000;6:1004–1010. doi: 10.1038/79510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kurinami H, Sato N, Shinohara M, Takeuchi D, Takeda S, Shimamura M, Ogihara T, Morishita R. Prevention of amyloid beta-induced memory impairment by fluvastatin, associated with the decrease in amyloid beta accumulation and oxidative stress in amyloid beta injection mouse model. Int J Mol Med. 2008;21:531–537. [PubMed] [Google Scholar]
- 42.Tong XK, Nicolakakis N, Fernandes P, Ongali B, Brouillette J, Quirion R, Hamel E. Simvastatin improves cerebrovascular function and counters soluble amyloid-beta, inflammation and oxidative stress in aged APP mice. Neurobiology of Disease. 2009;35:406–414. doi: 10.1016/j.nbd.2009.06.003. [DOI] [PubMed] [Google Scholar]
- 43.Riekse RG, Li G, Petrie EC, Leverenz JB, Vavrek D, Vuletic S, Albers JJ, Montine TJ, Lee VM, Lee M, Seubert P, Galasko D, Schellenberg GD, Hazzard WR, Peskind ER. Effect of statins on Alzheimer' disease biomarkers in cerebrospinal fluid. J Alzheimers Dis. 2006;10:399–406. doi: 10.3233/jad-2006-10408. [DOI] [PubMed] [Google Scholar]
- 44.Piermartiri TC, Figueiredo CP, Rial D, Duarte FS, Bezerra SC, Mancini G, de Bem AF, Prediger RD, Tasca CI. Atorvastatin prevents hippocampal cell death, neuroinflammation and oxidative stress following amyloid-beta(1–40) administration in mice: Evidence for dissociation between cognitive deficits and neuronal damage. Exp Neurol. 2010;226:274–284. doi: 10.1016/j.expneurol.2010.08.030. [DOI] [PubMed] [Google Scholar]
- 45.Murphy MP, Morales J, Beckett TL, Astarita G, Piomelli D, Weidner A, Studzinski CM, Dowling AL, Wang X, Levine H, 3rd, Kryscio RJ, Lin Y, Barrett E, Head E. Changes in cognition and amyloid-beta processing with long term cholesterol reduction using atorvastatin in aged dogs. J Alzheimers Dis. 2010;22:135–150. doi: 10.3233/JAD-2010-100639. [DOI] [PubMed] [Google Scholar]
- 46.Barone E, Cenini G, Di Domenico F, Martin S, Sultana R, Mancuso C, Murphy MP, Head E, Butterfield DA. Long-term high-dose atorvastatin decreases brain oxidative and nitrosative stress in a preclinical model of alzheimer disease: A novel mechanism of action. Pharmacol Res. 2011;63:172–180. doi: 10.1016/j.phrs.2010.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bliznakov EG, Wilkins DJ. Biochemical and clinical consequences of inhibiting coenzyme q(10) biosynthesis by lipid-lowering hmg-coa reductase inhibitors (statins): A critical overview. Adv Ther. 1998;15:218–228. [Google Scholar]
- 48.Langsjoen PH, Langsjoen JO, Langsjoen AM, Lucas LA. Treatment of statin adverse effects with supplemental coenzyme Q10 and statin drug discontinuation. Biofactors. 2005;25:147–152. doi: 10.1002/biof.5520250116. [DOI] [PubMed] [Google Scholar]
- 49.Parker RA, Huang Q, Tesfamariam B. Influence of 3-hydroxy-3-methylglutaryl-coa (hmgcoa) reductase inhibitors on endothelial nitric oxide synthase and the formation of oxidants in the vasculature. Atherosclerosis. 2003;169:19–29. doi: 10.1016/s0021-9150(03)00100-x. [DOI] [PubMed] [Google Scholar]
- 50.Vaya J, Schipper HM. Oxysterols, cholesterol homeostasis, and alzheimer disease. J Neurochem. 2007;102:1727–1737. doi: 10.1111/j.1471-4159.2007.04689.x. [DOI] [PubMed] [Google Scholar]
- 51.Han JH, Kim YJ, Han ES, Lee CS. Prevention of 7-ketocholesterol-induced mitochondrial damage and cell death by calmodulin inhibition. Brain Res. 2007;1137:11–19. doi: 10.1016/j.brainres.2006.12.041. [DOI] [PubMed] [Google Scholar]
- 52.Panini SR, Yang L, Rusinol AE, Sinensky MS, Bonventre JV, Leslie CC. Arachidonate metabolism and the signaling pathway of induction of apoptosis by oxidized ldl/oxysterol. J Lipid Res. 2001;42:1678–1686. [PubMed] [Google Scholar]
- 53.Trousson A, Bernard S, Petit PX, Liere P, Pianos A, El Hadri K, Lobaccaro JM, Ghandour MS, Raymondjean M, Schumacher M, Massaad C. 25-hydroxycholesterol provokes oligodendrocyte cell line apoptosis and stimulates the secreted phospholipase a2 type iia via lxr beta and pxr. J Neurochem. 2009;109:945–958. doi: 10.1111/j.1471-4159.2009.06009.x. [DOI] [PubMed] [Google Scholar]
- 54.Arca M, Natoli S, Micheletta F, Riggi S, Di Angelantonio E, Montali A, Antonini TM, Antonini R, Diczfalusy U, Iuliano L. Increased plasma levels of oxysterols, in vivo markers of oxidative stress, in patients with familial combined hyperlipidemia: Reduction during atorvastatin and fenofibrate therapy. Free Radic Biol Med. 2007;42:698–705. doi: 10.1016/j.freeradbiomed.2006.12.013. [DOI] [PubMed] [Google Scholar]
- 55.Kathir K, Dennis JM, Croft KD, Mori TA, Lau AK, Adams MR, Stocker R. Equivalent lipid oxidation profiles in advanced atherosclerotic lesions of carotid endarterectomy plaques obtained from symptomatic type 2 diabetic and nondiabetic subjects. Free Radic Biol Med. 2010;49:481–486. doi: 10.1016/j.freeradbiomed.2010.05.006. [DOI] [PubMed] [Google Scholar]
- 56.Larsson H, Bottiger Y, Iuliano L, Diczfalusy U. In vivo interconversion of 7beta-hydroxycholesterol and 7-ketocholesterol, potential surrogate markers for oxidative stress. Free Radic Biol Med. 2007;43:695–701. doi: 10.1016/j.freeradbiomed.2007.04.033. [DOI] [PubMed] [Google Scholar]
- 57.Berthier A, Lemaire-Ewing S, Prunet C, Monier S, Athias A, Bessede G, Pais de Barros JP, Laubriet A, Gambert P, Lizard G, Neel D. Involvement of a calcium-dependent dephosphorylation 17 of bad associated with the localization of trpc-1 within lipid rafts in 7-ketocholesterol-induced Thp-1 cell apoptosis. Cell Death Differ. 2004;11:897–905. doi: 10.1038/sj.cdd.4401434. [DOI] [PubMed] [Google Scholar]
- 58.Kumar BS, Chung BC, Lee YJ, Yi HJ, Lee BH, Jung BH. A GC/MS-based simultaneous quantitative analytical method for urinary oxysterols and bile acids in rats. Anal Biochem. 2010 doi: 10.1016/j.ab.2010.09.031. [DOI] [PubMed] [Google Scholar]
- 59.Thelen KM, Laaksonen R, Paiva H, Lehtimaki T, Lutjohann D. High-dose statin treatment does not alter plasma marker for brain cholesterol metabolism in patients with moderately elevated plasma cholesterol levels. J Clin Pharmacol. 2006;46:812–816. doi: 10.1177/0091270006289851. [DOI] [PubMed] [Google Scholar]
- 60.Beydoun MA, Beason-Held LL, Kitner-Triolo MH, Beydoun HA, Ferrucci L, Resnick SM, Zonderman AB. Statins and serum cholesterol's associations with incident dementia and mild cognitive impairment. J Epidemiol Community Health. 2010 doi: 10.1136/jech.2009.100826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Li G, Shofer JB, Rhew IC, Kukull WA, Peskind ER, McCormick W, Bowen JD, Schellenberg GD, Crane PK, Breitner JC, Larson EB. Age-varying association between statin use and incident Alzheimer's disease. J Am Geriatr Soc. 2010;58:1311–1317. doi: 10.1111/j.1532-5415.2010.02906.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Smeeth L, Douglas I, Hall AJ, Hubbard R, Evans S. Effect of statins on a wide range of health outcomes: A cohort study validated by comparison with randomized trials. Br J Clin Pharmacol. 2009;67:99–109. doi: 10.1111/j.1365-2125.2008.03308.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Haag MD, Hofman A, Koudstaal PJ, Stricker BH, Breteler MM. Statins are associated with a reduced risk of Alzheimer disease regardless of lipophilicity. The Rotterdam study. J Neurol Neurosurg Psychiatry. 2009;80:13–17. doi: 10.1136/jnnp.2008.150433. [DOI] [PubMed] [Google Scholar]
- 64.Horsdal HT, Olesen AV, Gasse C, Sorensen HT, Green RC, Johnsen SP. Use of statins and risk of hospitalization with dementia: A Danish population-based case-control study. Alzheimer Dis Assoc Disord. 2009;23:18–22. doi: 10.1097/WAD.0b013e318180f55b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Rosenberg H, Allard D. Women and statin use: A women's health advocacy perspective. Scand Cardiovasc J. 2008;42:268–273. doi: 10.1080/14017430801993180. [DOI] [PubMed] [Google Scholar]
- 66.Sparks DL, Kryscio RJ, Sabbagh MN, Connor DJ, Sparks LM, Liebsack C. Reduced risk of incident AD with elective statin use in a clinical trial cohort. Curr Alzheimer Res. 2008;5:416–421. doi: 10.2174/156720508785132316. [DOI] [PubMed] [Google Scholar]
- 67.Cramer C, Haan MN, Galea S, Langa KM, Kalbfleisch JD. Use of statins and incidence of dementia and cognitive impairment without dementia in a cohort study. Neurology. 2008;71:344–350. doi: 10.1212/01.wnl.0000319647.15752.7b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Benito-Leon J, Louis ED, Vega S, Bermejo-Pareja F. Statins and cognitive functioning in the elderly: A population-based study. J Alzheimers Dis. 2010;21:95–102. doi: 10.3233/JAD-2010-100180. [DOI] [PubMed] [Google Scholar]
- 69.Hippisley-Cox J, Coupland C. Unintended effects of statins in men and women in england and wales: Population based cohort study using the qresearch database. BMJ. 2010;340:c2197. doi: 10.1136/bmj.c2197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sparks DL, Kryscio RJ, Connor DJ, Sabbagh MN, Sparks LM, Lin Y, Liebsack C. Cholesterol and cognitive performance in normal controls and the influence of elective statin use after conversion to mild cognitive impairment: Results in a clinical trial cohort. Neurodegener Dis. 2010;7:183–186. doi: 10.1159/000295660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Trompet S, van Vliet P, de Craen AJ, Jolles J, Buckley BM, Murphy MB, Ford I, Macfarlane PW, Sattar N, Packard CJ, Stott DJ, Shepherd J, Bollen EL, Blauw GJ, Jukema JW, Westendorp RG. Pravastatin and cognitive function in the elderly. Results of the prosper study. J Neurol. 2010;257:85–90. doi: 10.1007/s00415-009-5271-7. [DOI] [PubMed] [Google Scholar]
- 72.Shepherd J, Blauw GJ, Murphy MB, Cobbe SM, Bollen EL, Buckley BM, Ford I, Jukema JW, Hyland M, Gaw A, Lagaay AM, Perry IJ, Macfarlane PW, Meinders AE, Sweeney BJ, Packard CJ, Westendorp RG, Twomey C, Stott DJ. The design of a prospective study of pravastatin in the elderly at risk (prosper). Prosper study group. Prospective study of pravastatin in the elderly at risk. Am J Cardiol. 1999;84:1192–1197. doi: 10.1016/s0002-9149(99)00533-0. [DOI] [PubMed] [Google Scholar]
- 73.Feldman HH, Doody RS, Kivipelto M, Sparks DL, Waters DD, Jones RW, Schwam E, Schindler R, Hey-Hadavi J, DeMicco DA, Breazna A. Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease: Leade. Neurology. 2010;74:956–964. doi: 10.1212/WNL.0b013e3181d6476a. [DOI] [PubMed] [Google Scholar]
- 74.Waters DD. Exploring new indications for statins beyond atherosclerosis: Successes and setbacks. J Cardiol. 2010;55:155–162. doi: 10.1016/j.jjcc.2009.12.003. [DOI] [PubMed] [Google Scholar]
- 75.Sparks DL, Sabbagh M, Connor D, Soares H, Lopez J, Stankovic G, Johnson-Traver S, Ziolkowski C, Browne P. Statin therapy in Alzheimer's disease. Acta Neurol Scand Suppl. 2006;185:78–86. doi: 10.1111/j.1600-0404.2006.00689.x. [DOI] [PubMed] [Google Scholar]
- 76.Sparks DL, Connor DJ, Sabbagh MN, Petersen RB, Lopez J, Browne P. Circulating cholesterol levels, apolipoprotein E genotype and dementia severity influence the benefit of atorvastatin treatment in Alzheimer's disease: Results of the alzheimer's disease cholesterol-lowering treatment (adclt) trial. Acta Neurol Scand Suppl. 2006;185:3–7. doi: 10.1111/j.1600-0404.2006.00690.x. [DOI] [PubMed] [Google Scholar]
- 77.McGuinness B, O'Hare J, Craig D, Bullock R, Malouf R, Passmore P. Statins for the treatment of dementia. Cochrane Database Syst Rev. 2010:CD007514. doi: 10.1002/14651858.CD007514.pub2. [DOI] [PubMed] [Google Scholar]
- 78.O'Brien JT, Burns A. Clinical practice with anti-dementia drugs: A revised (second) consensus statement from the british association for psychopharmacology. J Psychopharmacol. 2010 doi: 10.1177/0269881110387547. [DOI] [PubMed] [Google Scholar]
- 79.Querfurth HW, LaFerla FM. Alzheimer's disease. N Engl J Med. 2010;362:329–344. doi: 10.1056/NEJMra0909142. [DOI] [PubMed] [Google Scholar]
- 80.Mayeux R. Clinical practice. Early Alzheimer's disease. N Engl J Med. 2010;362:2194–2201. doi: 10.1056/NEJMcp0910236. [DOI] [PubMed] [Google Scholar]
- 81.Simons M, Schwarzler F, Lutjohann D, von Bergmann K, Beyreuther K, Dichgans J, Wormstall H, Hartmann T, Schulz JB. Treatment with simvastatin in normocholesterolemic patients with Alzheimer's disease: A 26-week randomized, placebo-controlled, double-blind trial. Ann Neurol. 2002;52:346–350. doi: 10.1002/ana.10292. [DOI] [PubMed] [Google Scholar]
- 82.Saher G, Brugger B, Lappe-Siefke C, Mobius W, Tozawa R, Wehr MC, Wieland F, Ishibashi S, Nave KA. High cholesterol level is essential for myelin membrane growth. Nat Neurosci. 2005;8:468–475. doi: 10.1038/nn1426. [DOI] [PubMed] [Google Scholar]
- 83.Reiman EM, Chen K, Langbaum JB, Lee W, Reschke C, Bandy D, Alexander GE, Caselli RJ. Higher serum total cholesterol levels in late middle age are associated with glucose hypometabolism in brain regions affected by Alzheimer's disease and normal aging. Neuroimage. 2010;49:169–176. doi: 10.1016/j.neuroimage.2009.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Fonseca AC, Resende R, Oliveira CR, Pereira CM. Cholesterol and statins in Alzheimer's disease: Current controversies. Exp Neurol. 2010;223:282–293. doi: 10.1016/j.expneurol.2009.09.013. [DOI] [PubMed] [Google Scholar]
- 85.McGuinness B, Craig D, Bullock R, Passmore P. Statins for the prevention of dementia. Cochrane Database Syst Rev. 2009:CD003160. doi: 10.1002/14651858.CD003160.pub2. [DOI] [PubMed] [Google Scholar]
- 86.Hoglund K, Wiklund O, Vanderstichele H, Eikenberg O, Vanmechelen E, Blennow K. Plasma levels of beta-amyloid(1–40), beta-amyloid(1–42), and total beta-amyloid remain unaffected in adult patients with hypercholesterolemia after treatment with statins. Arch Neurol. 2004;61:333–337. doi: 10.1001/archneur.61.3.333. [DOI] [PubMed] [Google Scholar]
- 87.Evans BA, Evans JE, Baker SP, Kane K, Swearer J, Hinerfeld D, Caselli R, Rogaeva E, St George-Hyslop P, Moonis M, Pollen DA. Long-term statin therapy and CSF cholesterol levels: Implications for Alzheimer's disease. Dement Geriatr Cogn Disord. 2009;27:519–524. doi: 10.1159/000221835. [DOI] [PubMed] [Google Scholar]
- 88.Jann MW, Shirley KL, Small GW. Clinical pharmacokinetics and pharmacodynamics of cholinesterase inhibitors. Clin Pharmacokinet. 2002;41:719–739. doi: 10.2165/00003088-200241100-00003. [DOI] [PubMed] [Google Scholar]
- 89.Kiani J, Imam SZ. Medicinal importance of grapefruit juice and its interaction with various drugs. Nutr J. 2007;6:33. doi: 10.1186/1475-2891-6-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Stump AL, Mayo T, Blum A. Management of grapefruit-drug interactions. Am Fam Physician. 2006;74:605–608. [PubMed] [Google Scholar]
- 91.Hare JT, Elliott DP. Grapefruit juice and potential drug interactions. Consult Pharm. 2003;18:466–472. [PubMed] [Google Scholar]
- 92.Dreier JP, Endres M. Statin-associated rhabdomyolysis triggered by grapefruit consumption. Neurology. 2004;62:670. doi: 10.1212/wnl.62.4.670. [DOI] [PubMed] [Google Scholar]
- 93.Werba JP, Giroli M, Cavalca V, Nava MC, Tremoli E, Dal Bo L. The effect of green tea on simvastatin tolerability. Ann Intern Med. 2008;149:286–287. doi: 10.7326/0003-4819-149-4-200808190-00019. [DOI] [PubMed] [Google Scholar]
- 94.Perluigi M, Sultana R, Cenini G, Di Domenico F, Memo M, Pierce WM, Coccia R, Butterfield DA. Redox proteomics identification of 4-hydroxynonenal-modified brain proteins in alzheimer's disease: Role of lipid peroxidation in Alzheimer's disease pathogenesis. Proteomics Clin Appl. 2009;3:682–693. doi: 10.1002/prca.200800161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Sultana R, Perluigi M, Newman SF, Pierce WM, Cini C, Coccia R, Butterfield DA. Redox proteomic analysis of carbonylated brain proteins in mild cognitive impairment and early Alzheimer's disease. Antioxid Redox Signal. 2010;12:327–336. doi: 10.1089/ars.2009.2810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Sultana R, Perluigi M, Butterfield DA. Oxidatively modified proteins in Alzheimer's disease (AD), mild cognitive impairment and animal models of ad: Role of Abeta in pathogenesis. Acta Neuropathol. 2009;118:131–150. doi: 10.1007/s00401-009-0517-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Butterfield DA, Drake J, Pocernich C, Castegna A. Evidence of oxidative damage in Alzheimer's disease brain: Central role for amyloid beta-peptide. Trends Mol Med. 2001;7:548–554. doi: 10.1016/s1471-4914(01)02173-6. [DOI] [PubMed] [Google Scholar]
- 98.Bellosta S, Paoletti R, Corsini A. Safety of statins: Focus on clinical pharmacokinetics and drug interactions. Circulation. 2004;109:III50–III57. doi: 10.1161/01.CIR.0000131519.15067.1f. [DOI] [PubMed] [Google Scholar]
- 99.Mukhtar RY, Reid J, Reckless JP. Pitavastatin. Int J Clin Pract. 2005;59:239–252. doi: 10.1111/j.1742-1241.2005.00461.x. [DOI] [PubMed] [Google Scholar]








