Abstract
Cholesterol is vital to normal brain function including learning and memory but that involvement is as complex as the synthesis, metabolism and excretion of cholesterol itself. Dietary cholesterol influences learning tasks from water maze to fear conditioning even though cholesterol does not cross the blood brain barrier. Excess cholesterol has many consequences including peripheral pathology that can signal brain via cholesterol metabolites, proinflammatory mediators and antioxidant processes. Manipulations of cholesterol within the central nervous system through genetic, pharmacological, or metabolic means circumvent the blood brain barrier and affect learning and memory but often in animals already otherwise compromised. The human literature is no less complex. Cholesterol reduction using statins improves memory in some cases but not others. There is also controversy over statin use to alleviate memory problems in Alzheimer’s disease. Correlations of cholesterol and cognitive function are mixed and association studies find some genetic polymorphisms are related to cognitive function but others are not. In sum, the field is in flux with a number of seemingly contradictory results and many complexities. Nevertheless, understanding cholesterol effects on learning and memory is too important to ignore.
Keywords: Alzheimer’s disease, apolipoprotein E, ATP binding cassette transporters, cholesterol-fed rabbit, classical conditioning, eyeblink conditioning, fear conditioning, low-density lipoprotein receptors, statins, water maze, 24S-hydroxcholesterol
The Effects of Cholesterol on Learning and Memory
Cholesterol is ubiquitous in the central nervous system (CNS) and vital to normal brain function including signaling, synaptic plasticity, and learning and memory. Cholesterol is so important to brain function that it is generated independently of cholesterol metabolism in the rest of the body and is sequestered from the body by the blood brain barrier (BBB). A large number of studies pioneered by Dietschy and Turley among others have confirmed that systemic cholesterol levels do not influence cholesterol in the CNS (Dietschy, 2009; Dietschy and Turley, 2001; Dietschy and Turley, 2004). Given the importance of cholesterol to normal brain function, the current review will focus on the role of cholesterol in one of the most important functions of the brain – learning and memory.
Cholesterol Metabolism
Cholesterol metabolism in the CNS has a number of features in common with cholesterol metabolism in the rest of the body. In both locations, acetyl-CoA is converted to 3-hydrox-3-methylglutaryl-CoA (HMG-CoA) which is converted in a rate-limiting step to mevalonate by the enzyme HMG-CoA reductase. Mevalonate is then converted to squalene which is converted to lanosterol, which, through a series of 19 additional steps, is finally converted to cholesterol. In the adult CNS, cholesterol is synthesized almost exclusively in glial cells – astrocytes and, to a lesser extent, oligodendrocytes – with only a small amount of cholesterol synthesized in neurons. As a result of the critical role played by cholesterol in CNS function, cholesterol synthesis, metabolism, and excretion are all tightly regulated.
The major players in cholesterol synthesis, metabolism, and excretion and hence the major areas of interest in studying the effects of cholesterol on learning and memory include cholesterol itself, the enzyme HMG-CoA reductase, the cholesterol transport protein apolipoprotein E (ApoE), the adenosine triphosphate (ATP) binding cassette (ABC) transporter proteins A1 and G1 (ABCA1, ABCG1), the low-density lipoprotein receptor (LDLR) and LDLR-related protein (LRP), the oxysterols 24S-hydroxycholesterol and 27-hydroxycholesterol to which cholesterol is converted in the brain and body, respectively, and the liver X-activated receptors (LXRs) for which oxysterols are ligands and that induce expression of ApoE and ABCA1 genes (Benarroch, 2008).
CNS cholesterol represents almost 25% of the body’s total unesterified cholesterol and the majority (~70%) is found in the myelin sheath with the rest found in glial and neuronal membranes (Dietschy, 2009; Dietschy and Turley, 2001). Although a great deal of cholesterol is synthesized by neurons in the developing brain, synthesis is greatly reduced in the adult brain and takes place in glial cells (Goritz et al., 2006; Pfrieger, 2002). Cholesterol synthesis begins in the endoplasmic reticulum of astrocytes and involves HMG-CoA reductase which is regulated by cholesterol’s inhibition of sterol-regulated element binding protein (SREBP) that binds to the sterol-regulated element-1 of the HMG-CoA reductase gene in the nucleus to modify gene expression (Benarroch, 2008; Martins et al., 2009). Cholesterol in astrocytes is bound to ApoE and transported into the cerebrospinal fluid (CSF) via the ABCA1 transporter protein and taken up by neurons via the low-density lipoprotein receptor. The genes for both ApoE and ABCA1 are controlled by Liver × receptors and as such are important regulators of cholesterol synthesis. Liver × receptors, in turn, are activated by the cholesterol metabolite 24S-hydroxycholesterol and to a lesser extent by 27-hydroxycholesterol (Bjorkhem, 2009). Cholesterol in the CNS is converted to 24S-hydroxycholesterol by the enzyme 24S-hydroxylase (CYP24A1, a member of the cytochrome P450 family) and can cross the BBB to be excreted from the brain (Bjorkhem et al., 2009; Russell et al., 2009). Cholesterol in the rest of the body can be converted to 27-hydroxycholesterol by 27-hydroxylase (CYP27A1) and cross the BBB into the CNS (Heverin et al., 2005) to act as a ligand at specific receptors (e.g., LXR) and regulate enzymatic activity.
Learning and Memory Paradigms
Human tests
Assessment of learning and memory in humans, typically described more broadly as the measurement of cognitive function, consists of administering standardized tests designed to quantify intelligence, assess list learning and recall, determine comprehension, and probe different forms of procedural and episodic memory (Baldwin and Farias, 2009; Jacova et al., 2007; McDowell and Kristjansson, 1996). The number and type of test batteries designed to test cognitive function continues to grow with as many as 18 different batteries designed to be administered by computer (Wild et al., 2008). In some cases, learning and memory can be assessed with components of more global intelligence tests including the Working Memory Index of the Wechsler Adult Intelligence Scale (WAIS) or, in clinical settings, the cognitive subscale of the Alzheimer’s Disease Assessment Scale (ADAS-cog). In other cases, standardized tests have been specifically designed to measure cognitive impairment including the widely used Mini Mental Status Exam (MMSE). The MMSE is a brief instrument used to screen for dementia that is scored on a 30-point scale and tests orientation, registration, attention and calculation, recall, and language and practice. Scores have age and education norms with a score of less than 24 generally considered to be abnormal and indicative of mild cognitive impairment with decreasing scores considered to index increasing severity of impairment.
Animal tests
The vast majority of experiments that have examined the effects of cholesterol on animal learning and memory have used a relatively small number of behavioral paradigms including the water maze and radial-arm maze, cued and contextual fear conditioning, passive and active avoidance, and eyeblink conditioning.
Water maze
The water maze is a spatial navigation task also known as the Morris water maze (Morris, 1984) that assesses spatial learning and memory by allowing a mouse or rat to swim in a pool of opaque water in which an escape platform has been placed. In the cued or visible platform version, the platform is positioned above the water or its position just below the water is indicated by a flag or other proximal cue. In the hidden-platform version of the task, the platform is submerged and spatial cues are placed beyond the maze so that the subject must navigate to the platform using these spatial cues. In addition to assessing learning of and memory for a specific cue (e.g., the flag), the visible platform version of the task can assess whether sensory and motor abilities have been compromised by a treatment or condition. For example, a transgenic mouse may not be able to find the platform because it cannot swim as well as wild-type controls or cannot see the flag.
Learning the water maze task is usually assessed over a number of trials and measured as the time taken and the distance swum to the platform after the rodent is released from different positions around the edge of the pool. Memory for the task is assessed on days after training by allowing the rat or mouse to search for a platform that has been removed and measuring the time spent in the quadrant where the platform used to be, the number of times it crosses the position of the platform, and the time spent in the other quadrants of the pool.
Radial arm maze
The radial arm maze usually consists of a central start box from which radiate as many as twelve but usually eight identical runways (arms) with a goal box at the end of each arm where food or water can be left as a reward. The start box has a gate to each of the arms that can be closed limiting the number of arms to which the subject has access. In some paradigms the characteristics of the arms can modified so that they may be a different color, enclosed or provided with a different texture. Each arm contains photo beams that determine entry and may be used to determine speed. Runways entered and time to traverse the runway can also be assessed by an observer who should be “blind” to the experimental condition of the animals.
Fear conditioning
Cued fear conditioning involves the delivery of a brief foot shock through an electrified floor grid that is signaled by a cue such as a 30-second tone or light. Contextual fear conditioning involves the delivery of a brief foot shock that takes place in a different, usually novel environment. Cued and contextual fear conditioning may be combined within the same chamber. The rodent response to foot shock is freezing and increased heart rate. These two responses and particularly freezing have become standard indices of what is hypothesized to be fear of the cue or the context with which the foot shock has been paired.
Passive and active avoidance
Like fear conditioning, active and passive avoidance tasks involve a brief foot shock delivered through an electrified floor grid to a mouse or rat in a specific location. However, in avoidance tasks, the rodent must either move from the location where it has experienced the shock (active avoidance) or not enter that location (passive avoidance). An important aspect in both tasks is that rodents normally prefer a dark location over a brightly lit one and the foot shock usually occurs in the dark location. To avoid foot shock, a rat or mouse must either leave the dark side of the enclosure for the brightly lit side (active) or stay in the brightly lit side and not enter the dark side (passive). The day following active avoidance training, the latency with which a rodent leaves the previously shocked location is measured. The day following passive avoidance training the latency to enter the previously shocked dark location is measured. In some cases, a rat or mouse may never enter the dark location and an upper latency cut-off value is assigned (Crawley, 2000; Lu et al., 2009).
Eyeblink conditioning
Classical or Pavlovian conditioning of the eyeblink was originally developed in the early part of the 20th century and modeled after Pavlov’s studies with dogs pairing an innocuous or neutral stimulus such as a bell with a significant event such as food (Pavlov, 1927). In the case of eyeblink or eyelid conditioning, the neutral stimulus is typically a brief tone or flashing light and the significant event is a puff of air to the eye that elicits reflex closure of the eyelids. With repeated pairings of the tone and air puff, subjects soon begin to close their eyes before the puff reaches the cornea providing evidence that an association between the two has been formed. Eyeblink conditioning is most commonly conducted with rabbits (Gormezano et al., 1983) or rats (Schmajuk and Christiansen, 1990; Skelton, 1988).
Each of the foregoing behavioral paradigms and procedures has been used to assess the effects of dietary, genetic and pharmacological manipulations of cholesterol, cholesterol synthesis and cholesterol metabolism on animal learning and memory. In the case of the effects of cholesterol on human learning and memory, dietary and pharmacological manipulations as well as genetic differences have been assessed almost exclusively by psychological testing including intelligence tests, cognitive testing including tests of recall, cognitive impairment and dementia, and epidemiological analysis.
Cholesterol
Human Studies
There is a significant body of evidence that high cholesterol levels may be detrimental to human learning and memory. A significant number of studies show that elevated serum cholesterol is a risk factor for mild cognitive impairment (Foster, 2006; Kivipelto et al., 2001; Näslund et al., 2000; Solomon et al., 2007; Yaffe et al., 2002) and dementia (Solomon et al., 2009b; Whitmer et al., 2005) and that cholesterol levels are correlated with measures of intelligence (Atzmon et al., 2002; Muldoon et al., 1997; Reitan and Shipley, 1963; van Exel et al., 2002; Yaffe et al., 2002) except in the very elderly (Solomon et al., 2009a; West et al., 2008). Low HDL cholesterol has been correlated with deficits and declines in memory in midlife (Singh-Manoux et al., 2008). A study of cholesterol synthesis showed the level of the cholesterol precursors lanosterol and lathosterol are correlated with low memory performance as subjects age (Teunissen et al., 2003). Epidemiological evidence also suggests a strong relationship between cholesterol levels and Alzheimer’s disease - a disease noted for its severe decline in learning and memory (Canevari and Clark, 2007; Evans et al., 2000; Hartmann, 2001; Jarvik et al., 1995; Ledesma and Dotti, 2006; Lesser et al., 2009; Notkola et al., 1998; Simons et al., 2001; Sjogren et al., 2006; Stewart et al., 2001).
There are other human studies, however, showing that increased cholesterol improves learning and memory. For example, high cognitive functioning is correlated with high cholesterol (Elias et al., 2005; Panza et al., 2006) and cholesterol may protect against cognitive decline especially in the elderly (Mielke et al., 2005; Panza et al., 2006; van den Kommer et al., 2009; West et al., 2008). A factor that is in some dispute is the relationship between LDL and HDL and improved memory. West and colleagues have shown that better memory functioning is associated with higher total and LDL cholesterol levels in the very elderly whereas Atzmon and coworkers have suggested that only higher HDL levels are correlated with better cognitive function in the very elderly (Atzmon et al., 2002; West et al., 2008).
Taken together, the human data suggest that there is a relationship between cholesterol levels and adult learning and memory. This relationship appears to change as a function of age with cholesterol having its most detrimental effects in middle age and it’s most beneficial or protective effects in the very old. Interestingly, a recent study by Perry and colleagues found that there was no association between cognitive measures and serum cholesterol concentrations among the young (Perry et al., 2009).
Animal studies
Manipulations of cholesterol in animals have shown a number of different relationships between cholesterol and memory. For example, decreasing cholesterol in aged animals improves learning and memory for tasks such as the water maze (Kessler et al., 1986; Yehuda et al., 1998; Yehuda and Carasso, 1993). Feeding mice a 2% cholesterol diet for eight weeks may result in deficits in working memory in the water maze (Thirumangalakudi et al., 2008) but not always (Li et al., 2003). Feeding middle-aged rats a diet high in cholesterol andfat for eight weeks also resulted in deficits in working memory in the water maze (Granholm et al., 2008). Rats, mice and rabbits given calcium channel blockers that reduce the esterification of cholesterol and increase the hydrolysis of existing cholesterol esters (Nayler, 1999; Schachter, 1997) demonstrate improvements in a number of learning and memory paradigms including passive avoidance (Quartermain et al., 2001), water maze (Kane and Robinson, 1999; Quartermain et al., 2001), and eyeblink conditioning in rats and rabbits (Deyo et al., 1989; Kane and Robinson, 1999; Quartermain, 2000; Woodruff-Pak et al., 1997).
Elevating cholesterol in young DBA/2 mice improves performance in the water maze - a task normally impaired in this mutant (Miller and Wehner, 1994; Upchurch and Wehner, 1988). Dufour and colleagues showed feeding adult rats 2% cholesterol enhances water maze learning (Dufour et al., 2006). This effect was replicated by Micale and colleagues who also showed the enhancement in water maze learning could be reversed by blocking steroid synthesis (Micale et al., 2008). Animals that are either deficient in cholesterol or have cholesterol synthesis blocked have problems with learning and memory in the water maze (Endo et al., 1996; Voikar et al., 2002) and during classical conditioning of the rat eyeblink (Endo et al., 1996; O’Brien et al., 2002; Voikar et al., 2002; Xu et al., 1998). The learning deficits in eyeblink conditioning induced by impaired cholesterol synthesis were reversed by feeding rats cholesterol (Xu et al., 1998).
In a series of rabbit eyelid conditioning experiments we have documented the effects of feeding cholesterol on both learning and memory. Prompted by Sparks’ original observation that cholesterol-fed rabbits developed elevated levels of neuronal beta amyloid (Sparks et al., 1994), we were surprised to find that rabbits fed cholesterol for eight weeks showed improved trace classical conditioning and reflex facilitation of the NMR (Schreurs et al., 2003) and that these facilitating effects of cholesterol were a function of the concentration (Schreurs et al., 2007b) and duration of the cholesterol diet (Schreurs et al., 2007a). These facilitating effects were generalized beyond NMR conditioning because an eight-week, 2% cholesterol diet also facilitated rabbit heart rate conditioning – an index of conditioned fear (Schreurs et al., 2007c). It wasn’t until beta amyloid plaques were induced by adding 0.12 PPM copper to the drinking water given cholesterol-fed rabbits that learning suffered and rabbits performed more poorly than normal chow-fed controls (Sparks and Schreurs, 2003). Woodruff-Pak and her colleagues have shown that this cholesterol and copper-induced deficit in rabbit eyelid conditioning can be reversed by the administration of galantamine – an acetylcholinesterase inhibitor used to treat Alzheimer’s disease.
More recently, we have examined the effects of a 2% cholesterol diet on memory of NMR conditioning and found that an eight-week cholesterol diet following ten days of paired classical conditioning debilitated the rabbits’ ability to remember the association formed eight weeks earlier (Darwish et al., 2010). This effect occurred in the absence of detectable diet-induced changes in the cholesterol content of the brain which is consistent with the finds of Diestchy and Turley (2004). Surprisingly, there was a significantly higher level of cholesterol in the hippocampus and forebrain of rabbits given classical conditioning relative to unpaired controls regardless of their diet, suggesting that cholesterol levels in the brain can change as a function of experience (Dufour et al., 2006; Koudinov and Koudinova, 2001) as well as experience being able to change as a function of cholesterol.
Peripheral effects of cholesterol
The interesting question that is raised by the effects of cholesterol on learning and memory turns upon the inability of dietary cholesterol to cross the BBB into the CNS. The peripheral effects of feeding rabbits cholesterol are atherosclerosis, inflammation, and liver toxicity. As a result of elevated cholesterol, the liver produces increased lipoproteins rich in cholesterol esters that stay in the bloodstream and lead to atherosclerotic lesions. High levels of LDL trigger the endothelial cell expression of adherence molecules that mediate attachment of monocytes and lymphocytes to the rabbit artery wall that then migrate into the wall and result in fatty streaks (Jessup et al., 2004; Rader and Daugherty, 2008). Oxidized LDL in the artery wall accumulates in macrophages that have differentiated from monocytes and develop into foam cells. Other macrophages are activated by proinflammatory cytokines in the artery wall and release more proinflammatory mediators including reactive oxygen and nitrogen species, interleukin-1β, and tumor necrosis factor alpha (Hansson et al., 2008).
There is a body of literature documenting the effects of proinflammatory mediators including tumor necrosis factor alpha and interleukin-1β on synaptic plasticity (Di Filippo et al., 2008; Pickering and O’Connor, 2007) and membrane excitability (Schafers and Sorkin, 2008; Viviani et al., 2007). Moreover, systemic injection of the inflammatory cytokine interleukin-acquisition of classically conditioned eyeblink responses in rats (Servatius and Beck, 2003). Although there is evidence that these peripheral inflammatory mediators are transported across the BBB (Banks, 2005), breaches of the BBB including those produced by a high cholesterol diet (Chen et al., 2008; Sparks et al., 2000) may increase the influence of these mediators in the brain. Cholesterol in the rabbit induces hepatotoxicity by activating hepatic stellate cells, producing fibrosis, fat deposition and ballooning degeneration leading to focal necrosis, inflammatory reactions and lipogranuloma (Kainuma et al., 2006). Antioxidant activity of glutathione peroxidase and catalase decrease significantly in the cholesterol-fed rabbit liver giving rise to increased oxidation and formation of lipid peroxidation and oxysterols (Mahfouz and Kummerow, 2000).
Given the above, our ability to understand the mechanisms by which cholesterol influences learning and memory may turn upon its peripheral as well as central effects. For example, there is strong evidence that human cognitive impairment is correlated with the extent of cholesterol-induced atherosclerosis both in peripheral arterial disease (Rafnsson et al., 2009) and in carotid atherosclerosis (Romero et al., 2009). Regardless of the source of mediators thought to influence learning and memory, learning and memory are functions of the CNS and it is the CNS that must be reached by these mediators before they can have an effect.
Central effects of cholesterol
The central effects of feeding a cholesterol diet to rabbits have been explored by a number of investigators including Sparks and his colleagues who have examined microglia (Xue et al., 2007) and the role of copper in beta amyloid accumulation and clearance (Sparks et al., 2002b; Sparks, 2004; Sparks, 2007; Sparks et al., 2007) and Ghribi and his co-workers who have focused on the relationship between cholesterol and beta amyloid (Ghribi et al., 2006a; Ghribi et al., 2006b; Jaya Prasanthi et al., 2008; Sharma et al., 2008). The most consistent central effect of feeding rabbits cholesterol is the accumulation of intracellular beta amyloid – a finding first reported by Sparks (Sparks et al., 1994) and replicated in a number of different laboratories (Beach, 2008; Ghribi et al., 2006b; Ronald et al., 2009; Woodruff-Pak et al., 2007; Wu et al., 2003; Zatta et al., 2002).
There is a very large body of data implicating cholesterol in the deposition of beta amyloid, and thus, in Alzheimer’s disease (Canevari and Clark, 2007; Hirsch-Reinshagen and Wellington, 2007; Ledesma and Dotti, 2006; Reid et al., 2007; Sparks, 2007) but see (Elder et al., 2007). Although a detailed analysis of this literature is beyond the scope of the current review, it is important to note that the relationship between cholesterol and beta amyloid is complex (Bales, 2010; Grimm et al., 2007; Hartmann, 2001; Kirsch et al., 2003; Ledesma and Dotti, 2006; Lukiw et al., 2005; Martins et al., 2009; McLaurin et al., 2003; Panza et al., 2006; Raffai and Weisgraber, 2003; Roher et al., 1999; Sjogren et al., 2006; Wood et al., 2007; Yanagisawa, 2002) and may involve a number of factors including: (1) the dependence of beta and gamma secretase on cholesterol to cleave the amyloid precursor protein (APP) (Frears et al., 1999; Wahrle et al., 2002), (2) the connection between apoE and Alzheimer’s disease (Esler et al., 2002; Hartmann, 2001; Hoshino et al., 2002; Jarvik et al., 1995; Notkola et al., 1998), (3) a cholesterol-induced overproduction of beta amyloid which blocks cholesterol trafficking and leads to neurodegeneration (Liu et al., 1998; Yao and Papadopoulos, 2002), and (4) the role ApoE plays in calcium homeostasis (Hartmann et al., 1994; Veinbergs et al., 2002). There is also evidence that beta amyloid may affect cholesterol (Hartmann, 2006). It should be noted that there are many who argue that beta amyloid has important normal physiological functions (Grimm et al., 2007; Pearson and Peers, 2006; Wegiel et al., 2007) that take place throughout life (Wegiel et al., 2007) and it is only when there is an imbalance in beta amyloid production or clearance that it becomes toxic (Pearson and Peers, 2006; Sparks, 2007).
A growing body of evidence, particularly from cell culture systems and transgenic rodents, indicates it is the oligomeric form of beta amyloid that is important for its toxic effects and its effects on learning and memory (Billings et al., 2005; Billings et al., 2007; Dineley et al., 2002; Lesne et al., 2006; Ma et al., 2007; Morgan, 2003; Shankar et al., 2008). The initial debate about the relative effects of soluble versus insoluble beta amyloid (Despande et al., 2006; Gouras et al., 2005; Haass and Selkoe, 2007; Kayed et al., 2003; Zerbinatti et al., 2004) has given way to a more recent discussion of the oligomeric form of beta amyloid with higher soluble oligomers being more toxic and having greater effects on synaptic plasticity than monomers or dimers (Chafekar et al., 2008; Lacor et al., 2007; LaFerla et al., 2007; Shankar et al., 2007; Shankar et al., 2008; Walsh and Selkoe, 2007). However, Shankar et al. (2008) have shown that soluble beta amyloid dimers obtained from AD patients disrupted memory for passive avoidance in rats. Finally, it should be noted that despite the considerable body of evidence implicating beta amyloid in Alzheimer’s disease there is a significant minority who have argued that it is not all clear that beta amyloid is the cause of the disease (Joseph et al., 2001; Obrenovich et al., 2002; Pearson and Peers, 2006; Robakis, 2010; Robinson and Bishop, 2002; Savory et al., 2002).
Statins
One of the major strategies for treating high cholesterol and the cardiovascular disease that results is to interfere with the rate limiting step in cholesterol synthesis by inhibiting HMG-CoA reductase using statins – HMG-CoA reductase inhibitors. Statins have been very successful in lowering cholesterol and have been found to have a number of additional benefits including improved endothelial function, decreased oxidative stress, decreased inflammation, and improved immune responses (Jasinska et al., 2007; Liao and Laufs, 2005). As a result, it is difficult to determine whether statins’ effects on learning and memory are dependent on their effects on lowering cholesterol or on their non-cholesterol pleiotropic effects (Gotto Jr. and Farmer, 2001; Jasinska et al., 2007; Liao and Laufs, 2005).
Human studies
There is some evidence, although controversial, that lowering cholesterol levels with statins may reduce the rate of cognitive decline in Alzheimer’s disease patients (Arvanitakis et al., 2008; Haag et al., 2009; Hoglund et al., 2005; Hoyer and Riederer, 2007; Solomon and Kivipelto, 2009; Sparks et al., 2006; Zandi et al., 2005). These studies were prompted by earlier reports that statins lowered the risk of developing dementia (Jick et al., 2000; Wolozin et al., 2000). This area of research has been reviewed recently by McGuinness and colleagues for the Cochrane Database and they found there was no significant evidence that statins either helped or hindered cognition in Alzheimer’s patients. This is important because there is an older literature of case reports and clinical trials suggesting that statins may have a negative impact on cognition (Evans and Golomb, 2009; Muldoon et al., 2000). Given these mixed results of statin effects on cognition in Alzheimer’s disease patients, some have suggested it might be useful to parse the data in terms of whether or not statins in cross the BBB (Fassbender et al., 2002; Haag et al., 2009; Sparks et al., 2002a; Thelen et al., 2006). Interestingly, Haag et al. (2009) recently reported that statins reduced the risk of Alzheimer’s disease regardless of whether or not they crossed the BBB. Nevertheless, in the most recent international, multicenter, double-blind, randomized, parallel-group study with 640 patients, Feldman and associates reported no clinical benefit of atorvastatin as a treatment for mild or moderate Alzheimer’s disease (Feldman et al., 2010).
In contrast to studies with dementia patients, there is some evidence that statins may aid cognition in non-demented subjects. For example, a study by Parale and colleagues shows that statins improve cognitive function in non-demented patients over controls (Parale et al., 2006). In another study, Bernick and co-workers found that statin use slightly reduced the rate of cognitive decline in a group of normal subjects aged over 65 compared to matched non-treated controls (Bernick et al., 2005). Although the preponderance of demented-patient evidence suggests no strong positive or negative effects of statins on learning and memory, the data from non-demented subjects suggest that others may benefit. It could be argued that by the time dementia is advanced, there is so much pathology that statins can no longer be of benefit. To emphasize this point, Sparks and colleagues found that taking statins reduced the incidence of cognitive decline for those at risk for but not showing signs of Alzheimer’s disease (Sparks et al., 2008).
Animal studies
A number of animal studies have shown statins can facilitate learning and memory in rodents (Li et al., 2006; Lu et al., 2007). Li et al. (2006) showed that simvastatin was able to improve acquisition of the water maze task but had no effect on the visible platform version of the task suggesting statins affect learning without affecting sensory or motor function. Lu et al. (2007) demonstrated that rats suffering traumatic brain injury spent more time in the platform quadrant following water maze training if given simvastatin or atorvastatin compared to saline controls. Rats given simvastatin, a statin that crosses the BBB, did better than rats given atorvastatin, a statin that does not cross the BBB. There was no comparison of the effects of the statins on water maze performance in the absence of brain jury. In another water maze experiment, Koladiya and colleagues found that statins were able to reduce L-methione-induced vascular dementia that caused deficits in both acquisition and retention of the water maze task (Koladiya et al., 2008). However, both learning and memory in rats given the statins alone were no better than in controls.
Apolipoprotein E
ApoE is a glycoprotein secreted by glia that forms part of a lipoprotein particle that transports cholesterol through the CNS particularly to neurons. ApoE may also be involved in other transport functions particularly the clearance of beta amyloid. There is a significant body of literature showing that learning and memory in both human and non-human subjects is affected by the expression and specific allelic isoforms of ApoE (E2, E3, and E4). In Alzheimer’s disease patients, the ApoE4 allele is over-represented and the ApoE3 allele is under-represented (Martins et al., 2009) suggesting the former is a risk factor for the disease whereas the latter may be protective. The suggestion that there is an increased susceptibility for developing Alzheimer’s disease associated with the ApoE4 allele has been firmly established (Brouwers et al., 2008; Chen et al., 2002; Deary et al., 2002; Mayeux et al., 2001; Poirier, 2005; Sparks, 1997).
Human studies
Despite the strong association between ApoE genotype and Alzheimer’s disease, the evidence for the effects of ApoE on normal human learning and memory has been less clear. A number of early studies found that there was an association between ApoE genotype and cognition in middle-aged adults (Deary et al., 2002; Dik et al., 2001; Flory et al., 2000; Juva et al., 2000; Mayeux et al., 2001; Wilson et al., 2002) whereas other studies found no such relationship in the elderly (Small et al., 2000) or very elderly (Salo et al., 2002). More recent studies indicate that the effects of ApoE genotype on cognitive function may be affected by cholesterol levels (de Frias et al., 2007) but even when cognitive test results are adjusted for total cholesterol, the ApoE4 allele still has a significant negative effect (Liu et al., 2008). The interaction between ApoE allele and cholesterol levels points to the complexity of trying to examine a single aspect of cholesterol’s effects on learning and memory in isolation. This becomes particularly important for ApoE because it is involved not only in cholesterol transport but in beta amyloid aggregation and clearance, and may be involved in tau phosphorylation, synaptic plasticity and neuroinflammation (Kim et al., 2009).
Animal studies
Unlike human studies where ApoE cannot be manipulated, animal studies have examined the effects of ApoE insertion and deletion in transgenic mice as well as infusion of ApoE directly into rat brain to determine the effects of ApoE on learning and memory. Although some consistency has now emerged, it should be noted that, like the human studies, there are contradictory findings on the effects of ApoE on learning and memory in mice (Lominska et al., 2001) and that some of these differences may be due to strain and gender issues – a theme that appears to pervade research with transgenic mice (Arndt and Surjo, 2001; Brooks et al., 2004; van der Staay and Steckler, 2001). In a comprehensive study by Hartman and co-workers, ApoE knockout mice were engineered to have the human isoforms of ApoE3 or ApoE4 expressed in astrocytes and these mice were compared to ApoE knockout mice and wild type controls on a range of behavioral tasks (Hartman et al., 2001). Hartman and colleagues reported that all the ApoE mice were more reactive than the wild type mice on sensory/motor tasks including their startle response to a loud noise. However, ApoE mice showed no differences in water maze acquisition or retention. It was only when mice were tested in the radial arm maze that significant differences began to emerge in the acquisition of the task with ApoE4 mice needing more days to reach criterion and committing more errors than any other group. In contrast to the Hartman et al. (2001) water maze data, other groups have shown that ApoE deletions (Veinbergs et al., 2000) and human ApoE4 insertions (Bour et al., 2008) have deleterious effects on water maze performance. Importantly, the study by Bour et al. (2008) as well as an earlier study by the same group (Grootendorst et al., 2005) showed that there was some gender specificity to the effects of ApoE4 insertion on learning and memory with female mice showing poor water maze performance and retention whereas males where no worse than wild-type controls. Interestingly, there is one study that shows spatial maze learning – at least in the radial arm maze – is dependent on the age and background strain of the ApoE deficient mice (Lominska et al., 2001). Specifically, it wasn’t until six months of age that ApoE deficient mice on a C57Bl/6 background failed to show learning. One of the most recent ApoE experiments involved delivery of ApoE peptides directly into the hippocampus of rats rather than manipulation of gene expression in mice (Eddins et al., 2009). Eddins and colleagues found that infusion of segments of the ApoE protein into the ventral hippocampus produced impairment in the radial arm maze that persisted for several weeks after the infusion.
As in the case of statins, ApoE appears to be involved in a number of functions other than its direct effects on cholesterol. As mentioned above, ApoE has been implicated strongly in beta amyloid deposition and clearance with ApoE4 being less effective in clearing beta amyloid than ApoE2 or ApoE3 (Martins et al., 2009). Double transgenic mice that express both human beta amyloid and ApoE have provided evidence of the isoform-dependent effects of ApoE on beta amyloid accumulation (Bales, 2010). In a recent review, Kim et al. (2009) describe additional roles for ApoE including involvement in neurotoxicity, tau phosphorylation, neuroinflammation, cerebrovascular function and brain metabolism. The role of ApoE in learning and memory is further complicated because it is regulated by the ABCA1 and because it plays a role as a ligand for the low density lipoprotein receptor.
ATP Binding Cassette Transporters
Although 13 of 48 ATP binding cassette transporters are active in the CNS, only ABCA1 and ABCG1 have been examined for their role in learning and memory. The cholesterol transporter ABCA1 is a crucial regulator of ApoE and with the genetic loss of ABCA1 there is a significant increase in beta amyloid in the CNS (Kim et al., 2008).
Human studies
Reynolds and co-workers have conducted targeted genetic association analyses of ABCA1 and Alzheimer’s disease and found there to be a highly significant relationship between the two, implicating ABCA1 in dementia (Reynolds et al., 2009). Akram and colleagues recently found that there was a significant positive correlation between ABCA1 mRNA expression and dementia severity and that this differential expression was also reflected at the protein level (Akram et al., 2010).
The ATP binding cassette transporter ABCG1 is also important for cholesterol transport but there is considerable debate about whether or not it has a role in beta amyloid production with some suggesting it may even have a protective function (Kim et al., 2008). However, there is at least one study showing that ABCG1 is associated with Alzheimer’s disease (Wollmer et al., 2007).
Animal studies
In the only ABCA1 animal study to date, Lefterov et al. (2009) reported that the memory deficits shown by an Alzheimer’s disease transgenic mouse model (aged APP transgenic mice) in water maze acquisition and retention were exacerbated by a deficiency in ABCA1. Importantly, they found that mice deficient in ABCA1 without the APP mutation performed as well as wild-type controls (Lefterov et al., 2009).
There are two studies of learning and memory in transgenic mice over-expressing ABCG1 from different groups and both failed to find any effects of ABCG1 on water maze acquisition or retention (Burgess et al., 2008; Parkinson et al., 2009). Perhaps like ABCA1, ABCG1 may only have an effect when learning and memory are already compromised as in the human studies of dementia and the Lefterov et al. (2009) study of APP transgenic mice.
Low-Density Lipoprotein Receptors
The LDLR family is comprised of at least nine members, two of which – the low-density lipoprotein receptor and low-density lipoprotein receptor-related protein (LRP) – have been examined for a role in Alzheimer’s disease (Deane et al., 2009) and in mouse learning and memory (Qiu et al., 2006).
Human studies
There have been a number of studies of the association between members of the LDLR family and Alzheimer’s disease in humans and they have yielded conflicting results. Despite a number of studies finding no association between the LDLR gene polymorphism rs5925 and Alzheimer’s disease (Bertram et al., 2007; Rodriguez et al., 2006), one study has found that a different polymorphism (rs688) may be related to Alzheimer’s disease – at least in men (Zou et al., 2008). With the exception of one study where there was a genetic association between LRP and Alzheimer’s disease in a sample of Chinese patients (Zhou et al., 2008), other studies have found no significant association between LRP polymorphisms and Alzheimer’s disease (Bahia et al., 2008; Sagare et al., 2007) or learning and memory during aging in non-demented subjects (Reynolds et al., 2006). However, LRP has been shown to transport beta amyloid from the brain across the BBB and is lower in the serum of Alzheimer’s patients than controls (Sagare et al., 2007).
Animal studies
Studies of the effects of the LDLR family on murine learning and memory can be divided into those that employ genetic deletion of LDLR (Cao et al., 2006; Mulder et al., 2004; Thirumangalakudi et al., 2008) and, because deletion of LRP is a lethal mutation, those that manipulate LRP either by over-expression (Zerbinatti et al., 2004), administration of an antagonist (Harris-White et al., 2004), or reduced expression using antisense (Jaeger et al., 2010).
Cao and colleagues showed that cross-breeding LDLR-deficient mice with Tg2576 mice produced animals that at 10 months of age were hypercholesterolemic, had increased cerebral beta amyloid deposits and were impaired in water maze learning and retention compared to Tg2576 mice in which LDLRs were intact. Importantly, single transgenic mice deficient in LDLRs also became hypercholesterolemic but showed no deficits in water maze performance compared to control mice not deficient in LDLRs (Cao et al., 2006). These latter results are in contrast to an earlier study by Mulder et al. (2004) who found that compared to mice with LDLRs, LDLR-deficient mice had impaired memory for the location of a hidden platform in the water maze task. Differences between these two studies have been attributed to gender and background strain differences (Cao et al., 2006). However, it should be noted that, like any genetic manipulation, there is the potential for unwanted side effects. In this case, LDLR-deficient mice suffer from higher blood pressure and heart rate than controls (Mulder et al., 2004). Thirumangalakudi et al. (2008) also found memory impairment in LDLR-deficient mice compared to controls but in this case it was in retention of the radial-arm version of the water maze. In this task, hidden platforms are placed at the end of four of eight maze arms and the animal must locate a platform by swimming down the arms of the maze. Once a platform is found, it is removed and the animal must find another of the remaining platforms. Memory is assessed later in terms of the number of entries into arms that previously contained a platform (correct) and those that never contained a platform (error). The background strain for this latter study was the same as that in the Cao et al. (2006) study (C57BL/6) but the gender was not specified.
In a study of LRP, Zerbinatti et al. (2004) reported that over-expression of the LRP receptor in mice also over-expressing the amyloid precursor protein induced an increase in soluble beta amyloid as a function of age and resulted in poorer acquisition of the water maze task in both young and old mice. In a second LRP study, Harris-White and colleagues showed that receptor-associated protein (RAP), an LRP antagonist, attenuated the cellular targeting of beta amyloid induced by transforming growth factor and prevented deficits in water maze memory retention (Harris-White et al., 2004). Interestingly, as in a number of other cases where administration of a treatment to controls has no effect, administration of the RAP by itself did not significantly alter water maze memory scores compared to saline controls. In a third study, Sagare and colleagues chronically treated APP transgenic mice with LRP-IV – a major binding domain of LRP that binds beta amyloid with high affinity – and found treated mice performed better in a novel object recognition task than vehicle-treated APP mice (Sagare et al., 2007). Finally, Jaeger et al. (2009) used LRP mRNA antisense infusion into the brain to decrease the level of LRP and found impaired active avoidance and object recognition compared to animals infused with scrambled antisense. Unlike the Zerbianatti, Harris-White, and Sagare studies, the study by Jaeger and colleagues did not manipulate beta amyloid but did find it was increased in the brain and decreased crossing the BBB out of the CNS.
Liver × Receptors
LXRs are key transcription regulators of carbohydrate and lipid metabolism and are abundant throughout the body and brain. LXRs act as cholesterol and cholesterol metabolite sensors and regulate the expression of ABCA1 as well as ABCG1 and induce ApoE secretion. The importance of LXRs as system sterol sensors has led to the identification and testing of LXR agonists in a number of rodent models (Baranowski, 2008).
Human studies
There are no human studies of the effects of LXR on learning and memory.
Animal studies
Three different transgenic mouse studies have reported improvements in learning and memory following use of the LXR agonists TO901317 (Riddell et al., 2007; Vanmierlo et al., 2009) and GW3965 (Jiang et al., 2008). Riddell et al. (2007) showed that seven days of orally administered, brain penetrant TO0901317 lowered the level of beta amyloid in the hippocampus and improved memory of contextual fear conditioning in young APP transgenic mice (Tg2576) compared to wild-type controls. Similarly, Jiang et al. (2008) showed that six days of orally administered GW3965 to aged Tg2576 mice lowered the level of beta amyloid in the hippocampus and improved memory of contextual fear conditioning. The drug did not affect wild-type controls. Vanmierlo et al. (2009) found that extended administration of TO0901317 in the food of aged double transgenic APP-Presenilin (APP/PS) mice for six to nine weeks did not change beta amyloid plaque levels but did restore impaired object recognition memory. Object recognition involves exposing mice to two novel objects that can be explored and then, after a delay, presenting one of the now familiar objects with another novel object and allowing the mice to explore the two objects again. Mice that remember the familiar object will spend more time exploring the novel object. These improvements in learning and memory occurred against a backdrop of increased brain cholesterol precursor and decreased cholesterol metabolite levels (Vanmierlo et al., 2009) and elevated brain ApoE and ABCA1 levels (Jiang et al., 2008; Riddell et al., 2007). Clearly, LXR agonists have multiple CNS effects on key components of the cholesterol metabolic pathway. What remains to be determined is the causal connection between this multiplicity of effects and learning and memory. This is particularly important because as noted above, manipulation of ApoE or ABCA1 in wild-type controls has no effect on learning and memory.
Cholesterol Metabolites
Cholesterol is not degraded within the brain and cannot cross the BBB. However, cholesterol must be excreted and this is accomplished when cholesterol is converted into the brain-specific cholesterol metabolite 24S-hydroxcholesterol by the enzyme 24S-hydroxlase (Bjorkhem, 2002). In the periphery, systemic cholesterol is metabolized within the body to one of a number of products including the metabolite 27-hydroxycholesterol which can cross the BBB and enter the CNS (Heverin et al., 2005). The role of cholesterol metabolites in learning and memory in humans have been explored by measuring levels of 24S- and 27-hydoxycholesterol in serum and the cerebrospinal fluid (Kolsch et al., 2004; Papassotiropoulos et al., 2002; van den Kommer et al., 2009) and by identifying genetic polymorphisms related to cognitive impairment and Alzheimer’s disease (Fernandez del Pozo et al., 2006; Fu et al., 2009; Papassotiropoulos et al., 2005). Animal studies of the effects of cholesterol metabolites on learning and memory have included knockout mice lacking 24-hydroxylase (Kotti et al., 2006), infusion of 24-hydroxycholesterol into the brain (Zhao et al., 2009) and manipulation of the CYP46A1 gene that encodes the enzyme cholesterol 24-hydroxylase (Hudry et al., 2009).
Human studies
Lutjohann and colleagues have reported a series of studies in which they measured the cholesterol metabolites 24S- and 27-hydroxycholesterol in demented patients and controls. In one study they found that levels of 24S-hydroxycholesterol in the cerebrospinal fluid of Alzheimer’s patients and those with mild cognitive impairment were higher than in subjects without cognitive impairment (Papassotiropoulos et al., 2002). In a second study, Heverin et al. (2005) found increased post mortem levels of 27-hydroxcholesterol in the brains Alzheimer’s patients compared to controls. In a third study, cholesterol-corrected levels of both 24S- and 27-hydroxycholesterol were significantly reduced in the plasma of demented versus non-demented subjects and depressed patients (Kolsch et al., 2004). This relationship did not hold in a longitudinal study of cognitive decline in normal subjects between the ages of 55 and 85 where only the ratio between cholesterol and 27-hydroxycholesterol in carriers of the ApoE4 allele was a predictor of worsening cognitive function (van den Kommer et al., 2009). The variability in these data may be attributable, in part, to the very small quantities of these metabolites and the difficulty in measuring them. It is also interesting that what appear to be consistent findings in vitro have been less consistent in vivo suggesting that there are complex interactions among and between many of the steps in the synthesis and metabolism of cholesterol in the CNS (Bjorkhem, 2009; Russell et al., 2009).
A number of genetic association studies have found polymorphisms of the 24S-hydroxylase gene CYP46A1 in patients with mild cognitive impairment and Alzheimer’s disease. Papassotiropoulos et al. (2005) reported that a cluster of cholesterol-related genes including a single nucleotide polymorphism of CYP46A1 (rs754203) contributed to the risk for Alzheimer’s disease. Fernandez del Pozo and her colleagues examined the effect of the polymorphic site rs754203 on mild cognitive impairment and Alzheimer’s disease in patients with the ApoE3 or ApoE4 allele (Fernandez del Pozo et al., 2006). They found that the polymorphism increased the risk of Alzheimer’s disease in those with the ApoE3 form but not in those with the ApoE4 form. A number of polymorphisms of the CYP46A1 gene including rs754203 were examined in older Chinese subjects who were stratified into cognitively intact, mild, moderate, and severely demented using the Chinese versions of the MMSE and ADAS-cog (Fu et al., 2009). Fu et al. found at a 2-year follow up that subjects who deteriorated to dementia were more likely to carry the polymorphic site rs754203 or rs3742376 of the 24-hydroxylase gene than those that did not.
Animal studies
Kotti and her colleagues found that transgenic mice lacking 24-hydroxylase failed to learning the water maze task compared to wild-type controls (Kotti et al., 2006). These mice were also significantly worse than wild-type controls in both cued and contextual fear conditioning. Importantly, the 24-hydroxlase-deficient mice not only failed to synthesize or accumulate 24S-hydroxycholesterol, they show a 50% decrease in cholesterol synthesis in the brain. Zhao and co-workers showed that ten days after three infusions of 24S-hydroxycholesterol into the rat lateral ventricle, animals trained and tested in the water maze were found to have longer escape latencies, travel further and spend less time in the target quadrant than saline-injected and sham controls (Zhao et al., 2009). Importantly, the 24S-hydroxycholesterol infusions were found to be neurotoxic because they resulted in hippocampal lesions and significant numbers of apoptotic cells (Zhao et al., 2009). These neurotoxic effects were avoided by Hudry and her coworkers by using an adeno-associated virus encoding human CYP46A1 to increase 24S-hydroxycholesterol content in the cortex and hippocampus of APP/PS transgenic mice (Hudry et al., 2009). These investigators found that over-expression of CYP46A1 improved the cognitive deficits in APP/PS mice both in terms of the acquisition and retention of the water maze compared to APP/PS mice injected with a mutated version of the adeno-associated virus that did not over-express CYP46A1. They also found that mice over-expressing CYP46A1 had reduced levels of beta amyloid production and amyloid deposits compared to APP/PS control mice.
The differences between the various cholesterol metabolite studies that have been conducted in human and animal learning and memory attest to the complications that can arise when manipulating a complex metabolic pathway. For example, the 24-hydoxylase knock out study by Kotti et al. (2006) altered not only 24S-hydroxycholseterol but cholesterol synthesis itself. The reduction in cholesterol synthesis was compensated for by a decrease in HMG-CoA reductase activity as a result of negative feedback by cholesterol and, as a result, overall CNS cholesterol levels were unchanged (Kotti et al., 2006). Thus, 24S-hydroxycholesterol is not only a metabolite of cholesterol, it is also a involved in cholesterol synthesis because it regulates gene expression that maintains cholesterol homeostasis. Specifically, 24S-hydroxycholesterol can up-regulate ApoE via LXR (as well as ABCA1 and ABCG1) and increase ApoE-mediated cholesterol efflux from cells (Martins et al., 2009).
Conclusion
There is a large body of data showing that cholesterol is involved in learning and memory but the nature of that involvement appears to be as complex as the synthesis, metabolism and homeostasis of cholesterol itself. In some cases, there have been opposite behavioral results from the same transgenic manipulation only to find that the discrepancies may be attributable to strain or gender differences. We have examined data showing that dietary cholesterol can influence a diverse number of learning tasks from water maze to eyelid and fear conditioning even though cholesterol added to the diet does not cross the BBB. Manipulations of cholesterol within the CNS that circumvent the BBB through genetic, pharmacological, or metabolic measures have also produced a range of results but often in animals that have already been otherwise compromised. The same manipulations in normal controls have sometimes been found to be ineffective. The human cholesterol literature is no less complex. Reductions in cholesterol levels using statins having been found to be effective in improving learning and memory in some cases but not in others. Similarly, there is a great deal of controversy over whether statins can help alleviate the problems with learning and memory found in Alzheimer’s disease. Correlations of cholesterol levels with cognitive function have been found to be positive, negative, or to have no relationship at all. Association studies of cholesterol and cognition have found some genetic polymorphisms to be related to cognitive functions whereas others have not.
What is clear from all of these experiments is that cholesterol is critical to learning and memory and disturbances in cholesterol levels, synthesis or metabolism have significant consequences. These disturbances appear to have a range of direct and indirect effects. Although dietary cholesterol does not cross the BBB we have seen that there are a range of consequences of increasing cholesterol including significant peripheral pathology that may signal the brain along a number of different pathways including cholesterol metabolites, pro-inflammatory mediators and antioxidant processes. In cases where attempts have been made to model the complexity of these myriad effects, results have been difficult to interpret. Although a powerful tool in understanding the role of cholesterol, transgenic mouse models have significant short comings in terms of unlooked for side-effects and strain and gender issues. Finally, it seems clear that understanding the effects of cholesterol on learning and memory, although challenging, is too important to ignore.
Table 1.
Table 2.
Variable | Measure | Change | References |
---|---|---|---|
Elevated serum cholesterol | Water maze learning | Increase | (Dufour et al., 2006; Micale et al., 2008; Miller and Wehner, 1994; Upchurch and Wehner, 1988) |
Decrease | (Granholm et al., 2008; Thirumangalakudi et al., 2008) | ||
Eyeblink conditioning acquisition |
Increase | (Schreurs et al., 2003; Schreurs et al., 2007a; Schreurs et al., 2007b) |
|
Eyeblink conditioning memory |
Decrease | (Darwish et al., 2010) | |
Elevated cholesterol + copper | Decrease | (Sparks and Schreurs, 2003; Woodruff-Pak et al., 2007) | |
Decreased serum cholesterol | Water maze learning | Increase | (Kane and Robinson, 1999; Kessler et al., 1986; Quartermain et al., 2001; Yehuda et al., 1998; Yehuda and Carasso, 1993) |
Eyeblink conditioning | Increase | (Endo et al., 1996; O’Brien et al., 2002; Voikar et al., 2002; Xu et al., 1998) | |
Passive avoidance | Increase | (Quartermain et al., 2001) | |
Statin treatment | Water maze learning and zmemory |
Increase | (Li et al., 2006; Lu et al., 2007) |
No change | (Koladiya et al., 2008) | ||
Apolipoprotein E (ApoE4) | Water maze | Decrease | (Bour et al., 2008; Grootendorst et al., 2005; Veinbergs et al., 2000) |
Water maze | No change | (Hartman et al., 2001) | |
Radial arm maze | Decrease | (Eddins et al., 2009; Hartman et al., 2001; Lominska et al., 2001) | |
ATP Binding cassette – ABCA1 | Water maze | No change | (Lefterov et al., 2009) |
– ABCG1 | Water maze | No change | (Burgess et al., 2008; Parkinson et al., 2009) |
Low-density Lipoprotein Receptors | Water maze | Decrease | (Mulder et al., 2004) |
No change | (Cao et al., 2006) | ||
Water radial arm maze | Decrease | (Thirumangalakudi et al., 2008) | |
Active avoidance | Decrease | (Jaeger et al., 2010) | |
LRP + amyloid precursor protein | Water maze | Decrease | (Harris-White et al., 2004; Zerbinatti et al., 2004) |
Liver × receptor agonist | Fear conditioning | Increase | (Jiang et al., 2008; Riddell et al., 2007) |
Object recognition | Increase | (Vanmierlo et al., 2009) | |
Cholesterol metabolites (24S-hydroxycholesterol) |
Water maze | Decrease | (Hudry et al., 2009; Kotti et al., 2006; Zhao et al., 2009) |
Footnotes
Preparation of this manuscript and our experiments described herein were supported by NIH grant AG023211. The contents of this manuscript are solely the responsibility of the author and do not necessarily represent the official views of the NIA.
The author thanks Dr. D. Larry Sparks, Deya Darwish and Carrie Smith-Bell for critically reading the manuscript.
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.
Reference List
- Akram A, Schmeidler J, Kastel P, Hof PR, Haroutunian V. Increased expression of cholesterol transporter ABCA1 is highly correlated with severity of dementia in AD hippocampus. Brain Res. 2010;1318:167–177. doi: 10.1016/j.brainres.2010.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arndt SS, Surjo D. Methods for behavioural phenotyping of mouse mutants. How to keep the overview. Behav. Brain Res. 2001;125:39–42. doi: 10.1016/s0166-4328(01)00285-6. [DOI] [PubMed] [Google Scholar]
- Arvanitakis Z, Schneider JA, Wilson RS, Bienias JL, Kelly JF, Evans DA, Bennett DA. Statins, incident Alzheimer disease, change in cognitive function, and neuropathology. Neurology. 2008;70:1795–1802. doi: 10.1212/01.wnl.0000288181.00826.63. [DOI] [PubMed] [Google Scholar]
- Atzmon G, Gabriely I, Griener W, Davidson D, Schechter C, Brazilai N. Plasma HDL levels highly correlate with cognitive function in exceptional longevity. Journal of Gerontology Series A, Biological Sciences and Medical Sciences. 2002;57A:M712–M715. doi: 10.1093/gerona/57.11.m712. [DOI] [PubMed] [Google Scholar]
- Bahia VS, Kok F, Marie SN, Shinjo S, Caramelli P, Nitrini R. Polymorphisms of APOE and LRP genes in Brazilian individuals with Alzheimer disease. Alzheimer’s Disease and Associated Disorders. 2008;22:61–65. doi: 10.1097/WAD.0b013e31815a9da7. [DOI] [PubMed] [Google Scholar]
- Baldwin S, Farias ST. Neuropsychological assessment in the diagnosis of Alzheimer’s disease. Current Protocols in Neuroscience. 2009;49:10.3.1–10.3.8. doi: 10.1002/0471142301.ns1003s49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bales KR. Brain lipid metabolism, apolipoprotein E and the pathophysiology of Alzheimer’s disease. Neuropharmacology. 2010:xxx. doi: 10.1016/j.neuropharm.2010.01.005. In press. [DOI] [PubMed] [Google Scholar]
- Banks WA. Blood-brain barrier transport of cytokines: a mechanism for neuropathology. Curr. Pharm. Des. 2005;11:973–984. doi: 10.2174/1381612053381684. [DOI] [PubMed] [Google Scholar]
- Baranowski M. Biological role of liver × receptors. J. Physiol. Pharmacol. 2008;59(Suppl 7):31–55. [PubMed] [Google Scholar]
- Beach TG. Physiologic origins of age-related β-amyloid deposition. Neurodegenerative Diseases. 2008;5:143–145. doi: 10.1159/000113685. [DOI] [PubMed] [Google Scholar]
- Benarroch EE. Brain cholesterol metabolism and neurologic disease. Neurology. 2008;71:1368–1373. doi: 10.1212/01.wnl.0000333215.93440.36. [DOI] [PubMed] [Google Scholar]
- Bernick C, Katz R, Smith NL, Rapp S, Bhadelia R, Carlson M, Kuller LH. Statins and cognitive function in the elderly. Neurology. 2005;65:1388–1394. doi: 10.1212/01.wnl.0000182897.18229.ec. [DOI] [PubMed] [Google Scholar]
- Bertram L, Hsiao M, MCQueen MB, Parkinson M, Mullin K, Blacker D, Tanzi RE. The LDLR locus in Alzheimer’s disease: a family-based study and meta-analysis of case-control data. Neurobiol. Aging. 2007;18:e18–e21. doi: 10.1016/j.neurobiolaging.2005.11.005. [DOI] [PubMed] [Google Scholar]
- Billings LM, Green KN, McGaugh JL, LaFerla FM. Learning decreases Aβ*56 and tau pathology and ameliorates behavioral decline in 3xTg-AD mice. J. Neurosci. 2007;27:751–761. doi: 10.1523/JNEUROSCI.4800-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Billings LM, Oddo S, Green KN, McGaugh JL, LeFerla FM. Intraneuronal Aβcauses the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron. 2005;45:675–688. doi: 10.1016/j.neuron.2005.01.040. [DOI] [PubMed] [Google Scholar]
- Bjorkhem I. Do oxysterols control cholesterol homeostasis? The Journal of Clinical Investigation. 2002;110:725–730. doi: 10.1172/JCI16388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bjorkhem I. Are side-chain oxidized oxysterols regulators also in vivo? J. Lipid Res. 2009;50:S213–S218. doi: 10.1194/jlr.R800025-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bjorkhem I, Cedazo-Minguez A, Leoni V, Meaney S. Oxysterols and neurodegenerative disorders. Mol. Aspects Med. 2009;30:171–179. doi: 10.1016/j.mam.2009.02.001. [DOI] [PubMed] [Google Scholar]
- Bour A, Grootendorst J, Vogel E, Kelche C, Dodart J-C, Bales K, Moreau PH, Sullivan PM, Mathis C. Middle-aged human apoE4 targeted-replacement mice show retention deficits on a wide range of spatial memory tasks. Behav. Brain Res. 2008;193:174–182. doi: 10.1016/j.bbr.2008.05.008. [DOI] [PubMed] [Google Scholar]
- Brooks SP, Pask T, Jones L, Dunnett SB. Behavioural profiles of inbred mouse strains used as transgenic backgrounds. I: motor tests. Genes, Brain and Behavior. 2004;3:206–215. doi: 10.1111/j.1601-183X.2004.00072.x. [DOI] [PubMed] [Google Scholar]
- Brouwers N, Sleegers K, Van Broeckhoven C. Molecular genetics of Alzheimer’s disease: an update. Ann. Med. 2008;18:1–22. doi: 10.1080/07853890802186905. [DOI] [PubMed] [Google Scholar]
- Burgess BL, Parkinson PF, Racke MM, Hirsch-Reinshagen V, Fan J, Wong C, Stukas S, Theroux L, Chan JY, Donkin J, Wilkinson A, Balik D, Christie B, Poirier J, Lutjohann D, DeMattos RB, Wellington CL. ABCG1 influences the brain cholesterol biosynthetic pathway but does not affect amyloid precursor protein or apolipoprotein E metabolism in vivo. J. Lipid Res. 2008;49:1254–1267. doi: 10.1194/jlr.M700481-JLR200. [DOI] [PubMed] [Google Scholar]
- Canevari L, Clark JB. Alzheimer’s disease and cholesterol: the fat connection. Neurochem. Res. 2007;32:739–750. doi: 10.1007/s11064-006-9200-1. [DOI] [PubMed] [Google Scholar]
- Cao D, Fukuchi K, Wan H, Kim H, Li L. Lack of LDL receptor aggravates learning deficits and amyloid deposits in Alzheimer transgenic mice. Neurobiol. Aging. 2006;27:1632–1643. doi: 10.1016/j.neurobiolaging.2005.09.011. [DOI] [PubMed] [Google Scholar]
- Carlsson CM, Gleason CE, Hess TM, Moreland KA, Blazel HM, Koscik RL, Schreiber NTN, Johnson SC, Atwood CS, Puglielli L, Hermann BP, McBride PE, Stein JH, Sager MA, Asthana S. Effects of simvastatin on cerebrospinal fluid biomarkers and cognition in middle-aged adults at risk for Alzheimer’s disease. Journal of Alzheimer’s Disease. 2008;13:187–192. doi: 10.3233/jad-2008-13209. [DOI] [PubMed] [Google Scholar]
- Carlsson CM, Nondahl DM, Klein BE, McBride PE, Sager MA, Schubert CR, Klein R, Cruickshanks KJ. increased atherogenic lipoproteins are associated with cognitive impairment: effects of statins and subclinical atherosclerosis. Alzheimer’s Disease and Associated Disorders. 2009;23:11–17. doi: 10.1097/wad.0b013e3181850188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chafekar SM, Baas F, Scheper W. Oligomer-specific aβtoxicity in cell models is mediated by selective uptake. Biochim. Biophys. Acta. 2008;1782:523–531. doi: 10.1016/j.bbadis.2008.06.003. [DOI] [PubMed] [Google Scholar]
- Chen JC, Edwards CL, Vidyarthi S, Pitchumoni S, Trabizi S, Barboriak D, Charles HC, Doriaswamy PM. Learning and recall in subjects at genetic risk for Alzheimer’s disease. Journal of Neuropsychiatry and Clinical Neuroscience. 2002;14:58–63. doi: 10.1176/jnp.14.1.58. [DOI] [PubMed] [Google Scholar]
- Chen X, Gawryluk JW, Wagener JF, Ghribi O, Geiger JD. Caffeine blocks disruption of the blood brain barrier in a rabbit model of Alzheimer’s disease. Journal of Neuroinflammation. 2008;5:1–14. doi: 10.1186/1742-2094-5-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crawley JN. Behavioral phenotyping of transgenic and knockout mice. Wiley-Liss; New York: 2000. What’s wrong with my mouse? [Google Scholar]
- Darwish DS, Wang D, Konat GW, Schreurs BG. Dietary cholesterol impairs memory and memory increases brain cholesterol and sulfatide levels. Behav. Neurosci. 2010;124:115–123. doi: 10.1037/a0018253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Frias CM, Bunce D, Wahlin A, Adolfsson R, Sleegers K, Cruts M, Van Broeckhoven C, Nilsson L-G. Cholesterol and triglycerides moderate the effect of apolipoprotein E on memory functioning in older adults. Journal of Gerentology: Psychological Sciences. 2007;62B:P112–P118. doi: 10.1093/geronb/62.2.p112. [DOI] [PubMed] [Google Scholar]
- Deane R, Bell RD, Sagare A, Zlokovic BV. Clearance of amyloid-βacross the blood-brain barrier: implications for therapies in Alzheimer’s disease. CNS & Neurological Disorder - Drug Targets. 2009;8:16–30. doi: 10.2174/187152709787601867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deary IJ, Whiteman MC, Pattie A, Starr JM, Hayward C, Wright AF, Carothers A, Whalley LJ. Cognitive change and the APOE ε4 allele. Nature. 2002;418:932. doi: 10.1038/418932a. [DOI] [PubMed] [Google Scholar]
- Despande A, Mina E, Glabe C, Busciglio J. Different conformations of amyloid βinduce toxicity by distinct mechanisms in human cortical neurons. J. Neurosci. 2006;26:6011–6018. doi: 10.1523/JNEUROSCI.1189-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deyo RA, Straube KT, Disterhoft JF. Nimodipine facilitates associative learning in aging rabbits. Science. 1989;243:809–811. doi: 10.1126/science.2916127. [DOI] [PubMed] [Google Scholar]
- Di Filippo M, Sarchielli P, Picconi B, Calabresi P. Neuroinflammation and synaptic plasticity: theoretical basis for a movel, immune-centred, therapeutic approach to neurological disorders. Trends Pharmacol. Sci. 2008;29:402–412. doi: 10.1016/j.tips.2008.06.005. [DOI] [PubMed] [Google Scholar]
- Dietschy JM. Central nervous system: cholesterol turnover, brain development and neurodegeneration. Biol. Chem. 2009;390:287–293. doi: 10.1515/BC.2009.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dietschy JM, Turley SD. Cholesterol metabolism in the brain. Curr. Opin. Lipidol. 2001;12:105–112. doi: 10.1097/00041433-200104000-00003. [DOI] [PubMed] [Google Scholar]
- Dietschy JM, Turley SD. Cholesterol metabolism in the central nervous system during early development and in the mature animal. J. Lipid Res. 2004;45:1375–1397. doi: 10.1194/jlr.R400004-JLR200. [DOI] [PubMed] [Google Scholar]
- Dik MG, Jonker C, Comijs HC, Bouter LM, Twisk JWR, van Kamp GJ, Deeg DJH. Memory complaints and APOE-ε4 accelerate cognitive decline in cognitively normal elderly. Neurology. 2001;57:2217–2222. doi: 10.1212/wnl.57.12.2217. [DOI] [PubMed] [Google Scholar]
- Dineley KT, Xia X, Bui D, Sweatt JD, Zheng H. Accelerated plaque accumulation, associative learning deficits, and up-regulation of α7 nicotinic receptor protein in transgenic mice co-expressing mutant human preseniline 1 and amyloid precursor proteins. J. Biol. Chem. 2002;277:22768–22780. doi: 10.1074/jbc.M200164200. [DOI] [PubMed] [Google Scholar]
- Dufour F, Liu Q-Y, Gusev P, Alkon DL, Atzori M. Cholesterol-enriched diet affects spatial learning and synaptic function in hippocampal synapses. Brain Res. 2006;1103:88–98. doi: 10.1016/j.brainres.2006.05.086. [DOI] [PubMed] [Google Scholar]
- Eddins D, Klein RC, Yakel JL, Levin ED. Hippocampal infusion of apolipoprotein E peptides induce long-lasting cognitive impairment. Brain Res. Bull. 2009;79:111–115. doi: 10.1016/j.brainresbull.2009.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elder GA, Cho JY, English DF, Franciosi S, Schmeidler J, Sosa M.A. Gama, De Gasperi R, Fisher EA, Mathews PM, Haroutunian V, Buxbaum JD. Elevated plasma cholesterol does not affect brain Aβin mice lacking the low-density lipoprotein receptor. J. Neurochem. 2007;102:1220–1231. doi: 10.1111/j.1471-4159.2007.04614.x. [DOI] [PubMed] [Google Scholar]
- Elias PK, Elias MF, D’Agostino RB, Sullivan LM, Wolf PA. Serum cholesterol and cognitive performance in the Framingham heart study. Psychosom. Med. 2005;67:24–30. doi: 10.1097/01.psy.0000151745.67285.c2. [DOI] [PubMed] [Google Scholar]
- Endo Y, Nishimura J-I, Kimura F. Impairment of maze learning in rats following long-term glucocorticoid treatments. Neurosci. Lett. 1996;203:199–202. doi: 10.1016/0304-3940(95)12296-6. [DOI] [PubMed] [Google Scholar]
- Esler WP, Marshall JR, Stimson ER, Ghilardi JR, Vinters HV, Mantyh PW, Maggio JE. Apolipoprotein E affects amyloid formation but not amyloid growth in vitro: mechanistic implications for ApoE4 enhanced amyloid burden and risk for Alzheimer’s disease. Amyloid: Journal of Protein Folding Disorders. 2002;9:1–12. doi: 10.3109/13506120209072439. [DOI] [PubMed] [Google Scholar]
- Evans MA, Golomb BA. Statin-associated adverse cognitive effects: survey results from 171 patients. Pharmacotherapy. 2009;29:800–811. doi: 10.1592/phco.29.7.800. [DOI] [PubMed] [Google Scholar]
- Evans RM, Emsley CL, Gao S, Sahota A, Hall KS, Farlow MR, Hendrie H. Serum cholesterol, APOE genotype, and the risk of Alzheimer’s disease: a population-based study of African Americans. Neurology. 2000;54:240–242. doi: 10.1212/wnl.54.1.240. [DOI] [PubMed] [Google Scholar]
- Fassbender K, Stroick M, Bertsch T, Ragaschke A, Kuehl S, Walter S, Walter J, Brechtel K, Muehlhauser F, von Bergmann K, Lutjohann D. Effects of statins on human cerebral cholesterol metabolism and secretion of Alzheimer amyloid peptide. Neurology. 2002;59:1257–1258. doi: 10.1212/wnl.59.8.1257. [DOI] [PubMed] [Google Scholar]
- 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]
- Fernandez del Pozo V, Alvarez Alvarez M, Fernandez Martinez M, Galados Alcelay L, Gomez Busto F, Pena JA, Alfonso-Snachez MA, Zarranz Imirizaldu JJ, de Pancordo MM. Polymorphism in the cholesterol 24S-hydroxylase gene (CYP46A1) associated with the APOEpsilon3 allele increases the risk of Alzheimer’s disease and of mild cognitive impairment progressing to Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 2006;21:81–87. doi: 10.1159/000090215. [DOI] [PubMed] [Google Scholar]
- Flory JD, Manuck SB, Ferrell RE, Ryan CM, Muldoon MF. Memory performance and the apolipoprotein E polymorphism in a community sample of middle-aged adults. American Journal of Medical Genetics (Neuropsychiatric Genetics) 2000;96:707–711. doi: 10.1002/1096-8628(20001204)96:6<707::aid-ajmg1>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
- Foster TC. Biological markers of age-related memory deficits. Treatment of senescent physiology. CNS Drugs. 2006;20:153–166. doi: 10.2165/00023210-200620020-00006. [DOI] [PubMed] [Google Scholar]
- Frears ER, Stephens DJ, Walters CE, Davies H, Austen BM. The role of cholesterol in the biosynthesis of β-amyloid. NeuroReport. 1999;10:1699–1705. doi: 10.1097/00001756-199906030-00014. [DOI] [PubMed] [Google Scholar]
- Fu BY, Ma SL, Tang NL, Tam CW, Lui VW, Chiu HF, Lam LC. Cholesterol 24-hydroxylase (CYP46A1) polymorphisms are associated with faster cognitive deterioration in Chinese older persons: a two-year follow up study. Int. J. Geriatr. Psychiatry. 2009;24:921–926. doi: 10.1002/gps.2196. [DOI] [PubMed] [Google Scholar]
- Ghribi O, Golovko MY, Larsen B, Schrag M, Murphy EJ. Deposition of iron and β-amyloid plaques is associated with cortical cellular damage in rabbits fed with long-term cholesterol-enriched diets. J. Neurochem. 2006a;99:438–449. doi: 10.1111/j.1471-4159.2006.04079.x. [DOI] [PubMed] [Google Scholar]
- Ghribi O, Larsen B, Schrag M, Herman MM. High cholesterol content in neurons increases BACE, β-amyloid, and phosphorylated tau levels in rabbit hippocampus. Exp. Neurol. 2006b;200:460–467. doi: 10.1016/j.expneurol.2006.03.019. [DOI] [PubMed] [Google Scholar]
- Goritz C, Mauch DH, Pfrieger FW. Multiple mechanisms mediate cholesterol-induced synaptogenesis in a CNS neuron. Mol. Cell. Neurosci. 2006;29:190–201. doi: 10.1016/j.mcn.2005.02.006. [DOI] [PubMed] [Google Scholar]
- Gormezano I, Kehoe EJ, Marshall BS. Twenty years of classical conditioning research with the rabbit. In: Sprague JM, editor. Progress in psychobiology and physiological psychology. Academic Press; New York: 1983. pp. 197–275. [Google Scholar]
- Gotto AM, Jr., Farmer JA. Pleiotropic effects of statins: do they matter? Curr. Opin. Lipidol. 2001;12:391–394. doi: 10.1097/00041433-200108000-00004. [DOI] [PubMed] [Google Scholar]
- Gouras GK, Almeida CG, Takahashi RH. Intraneuronal Aβaccummulation and origin of plaques in Alzheimer’s disease. Neurobiol. Aging. 2005;26:1235–1244. doi: 10.1016/j.neurobiolaging.2005.05.022. [DOI] [PubMed] [Google Scholar]
- Granholm A-C, Bimonte-Nelson HA, Moore AB, Nelson ME, Freeman LR, Sambamurti K. Effects of a saturated fat and high cholesterol diet on memory and hippocampal morphology in middle-aged rats. Journal of Alzheimer’s Disease. 2008;14:133–145. doi: 10.3233/jad-2008-14202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grimm MOW, Grimm HS, Hartmann T. Amyloid beta as a regulator of lipid homeostasis. Trends in Molecular Medicine. 2007;13:337–344. doi: 10.1016/j.molmed.2007.06.004. [DOI] [PubMed] [Google Scholar]
- Grootendorst J, Bours A, Vogel E, Kelche C, Sullivan PM, Dodart J-C, Bales K, Mathis C. Human apoE targeted replacement mouse lines: h-apoE4 and h-apoE3 mice differ on spatial memory performance and avoidance behavior. Behav. Brain Res. 2005;159:1–14. doi: 10.1016/j.bbr.2004.09.019. [DOI] [PubMed] [Google Scholar]
- Haag MDH, Hofman A, Koudstaal PJ, Stricker BHC, Breteler MMB. Statins as 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]
- Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid β-peptide. Nature Reviews Molecular Cell Biology. 2007;8:101–112. doi: 10.1038/nrm2101. [DOI] [PubMed] [Google Scholar]
- Hansson GK, Robertson A-KL, Soderberg-Naucler C. Inflammation and atherosclerosis. Annual Review of Pathology: Mechanisms of Disease. 2008;1:297–329. doi: 10.1146/annurev.pathol.1.110304.100100. [DOI] [PubMed] [Google Scholar]
- Harris-White ME, Balverde Z, Lim GP, Kim P, Miller SA, Hammer H, Galasko D, Frautschy SA. Role of LRP in TGFbeta2-mediated neuronal uptake of Abeta and effects on memory. J. Neurosci. Res. 2004;77:217–228. doi: 10.1002/jnr.20149. [DOI] [PubMed] [Google Scholar]
- Hartman RE, Wozniak DF, Nardi A, Olney JW, Sartorius L, Holtzman DM. Behavioral phenotyping of GFAP-ApoE3 and -ApoE4 transgenic mice: ApoE4 mice show profound working memory impairments in the absence of Alzheimer’s-like neuropathology. Exp. Neurol. 2001;170:326–344. doi: 10.1006/exnr.2001.7715. [DOI] [PubMed] [Google Scholar]
- Hartmann H, Eckert A, Muller W. Apolipoprotein E and cholesterol affect neuronal calcium signalling: the possible relationship to β-neurotoxicity. Biochem. Biophys. Res. Commun. 1994;200:1185–1192. doi: 10.1006/bbrc.1994.1576. [DOI] [PubMed] [Google Scholar]
- Hartmann T. Cholesterol, Aβand Alzheimer’s disease. Trends Neurosci. 2001;11:S45–S48. doi: 10.1016/s0166-2236(00)01990-1. [DOI] [PubMed] [Google Scholar]
- Hartmann T. Role of amyloid precursor protein, amyloid-βand γ-secretase in cholesterol maintenance. Neurodegenerative Diseases. 2006;3:305–311. doi: 10.1159/000095271. [DOI] [PubMed] [Google Scholar]
- Heverin M, Meaney S, Lutjohann D, Diczfalusy U, Wahren J, Bjorkhem I. Crossing the barrier: net flux of 27-hydroxycholesterol into the human brain. J. Lipid Res. 2005;46:1047–1052. doi: 10.1194/jlr.M500024-JLR200. [DOI] [PubMed] [Google Scholar]
- Hirsch-Reinshagen V, Wellington CL. Cholesterol metabolism, apolipoprotein E, adenosine triphosphate-binding cassette transporters, and Alzheimer’s disease. Curr. Opin. Lipidol. 2007;18:325–332. doi: 10.1097/MOL.0b013e32813aeabf. [DOI] [PubMed] [Google Scholar]
- Hoglund K, Thelen KM, Syversen S, Sjogren M, von Bergmann K, Wallin A, Vanmechelen E, Vanderstichele H, Lutjohann D, Blennow K. The effect of simvastatin treatment on the amyloid precursor protein and brain cholesterol metabolism in patients with Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 2005;19:256–265. doi: 10.1159/000084550. [DOI] [PubMed] [Google Scholar]
- Hoshino T, Kamino K, Matsumoto M. Gene dose effect of the APOE-ε4 allele on plasma HDL cholesterol level in patients with Alzheimer’s disease. Neurobiol. Aging. 2002;23:41–45. doi: 10.1016/s0197-4580(01)00252-4. [DOI] [PubMed] [Google Scholar]
- Hoyer S, Riederer P. Alzheimer disease - no traget for statin treatment. A mini review. Neurochem. Res. 2007;32:695–706. doi: 10.1007/s11064-006-9168-x. [DOI] [PubMed] [Google Scholar]
- Hudry E, Van Dam D, Kulik W, De Deyn PP, Stet FS, Ahouansou O, Benraiss A, Delacourte A, Bougneres P, Aubourg P, Cartier N. Adeno-associated virus gene therapy with cholesterol 24-hydroxylase reduces the amyloid pathology before or after onset of amyloid plaques in mouse models of Alzheimer’s disease. Molecular Therapy. 2009;18:44–53. doi: 10.1038/mt.2009.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacova C, Kertesz A, Blair M, Fisk JD, Feldman HH. Neuropsychological testing and assessment of dementia. Alzheimer’s & Dementia. 2007;3:299–317. doi: 10.1016/j.jalz.2007.07.011. [DOI] [PubMed] [Google Scholar]
- Jaeger LB, Dohgu S, Hwang MC, Farr SA, Murphy MP, Fleegal-Demotta MA, Lynch JL, Robinson SM, Niehoff ML, Johnson SN, Kumar VB, Banks WA. Testing the neurovascular hypothesis of Alzheimer’s disease: LRP-1 antisense reduces blood-brain barrier clearance, increases brain levels of amyloid-beta protein, and impairs cognition. Journal of Alzheimer’s Disease. 2010;17:553–570. doi: 10.3233/JAD-2009-1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jarvik GP, Wijsman EM, Kukull WA, Schellenberg GD, Yu C, Larson EB. Interactions of apolipoprotein E genotype, total cholesterol level, age, and sex in prediction of Alzheimer’s disease. Neurology. 1995;45:1092–1096. doi: 10.1212/wnl.45.6.1092. [DOI] [PubMed] [Google Scholar]
- Jasinska M, Owczarek J, Orszulak-Michalak D. Statins: a new insight into their mechanisms of action and consequent pleiotropic effects. Pharmacological Reports. 2007;59:483–499. [PubMed] [Google Scholar]
- Jaya Prasanthi RP, Schommer E, Thomasson S, Thompson A, Feist G, Ghribi O. Regulation of β-amyloid levels in the brain of cholesterol-fed rabbit, a model system for sporadic Alzheimer’s disease. Mech. Ageing Dev. 2008;29:649–655. doi: 10.1016/j.mad.2008.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jessup W, Kritharides L, Stocker R. Lipid peroxidation in atherogenesis: an overview. Biochem. Soc. Trans. 2004;32:134–138. doi: 10.1042/bst0320134. [DOI] [PubMed] [Google Scholar]
- Jiang Q, Lee CYD, Mandrekar S, Wilkinson B, Cramer P, Zelcer N, Mann K, Lamb B, Wilson TM, Collins JL, Richardson JC, Smith JD, Comery TA, Riddell D, Holtzman DM, Tontonoz P, Landreth GE. ApoE promotes the proteolytic degradation of Aβ. Neuron. 2008;58:681–693. doi: 10.1016/j.neuron.2008.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jick H, Zornberg GL, Jick SS, Seshardi S, Drachman DA. Statins and the risk of dementia. Lancet. 2000;356:1627–1631. doi: 10.1016/s0140-6736(00)03155-x. [DOI] [PubMed] [Google Scholar]
- Joseph J, Shukitt-Hale B, Denisova NA, Martin A, Perry G, Smith MA. Copernicus revisited: amyloid beta in Alzheimer’s disease. Neurobiol. Aging. 2001;22:131–146. doi: 10.1016/s0197-4580(00)00211-6. [DOI] [PubMed] [Google Scholar]
- Juva K, Verkkoniemi A, Viramo P, Polvikoski T, Kainulainen K, Kontula K, Sulkava R. Apolipoprotein E, cognitive function, and dementia in a general population aged 85 years and over. Int. Psychogeriatr. 2000;12:379–387. doi: 10.1017/s1041610200006487. [DOI] [PubMed] [Google Scholar]
- Kainuma M, Fujimoto M, Sekiya N, Tsuneyama K, Cheng C, Takano Y, Tersawa K, Shimada Y. Cholesterol-fed rabbit as a unique model of nonalcoholic, nonobese, non-insulin-resistant fatty liver disease with characteristic fibrosis. J. Gastroenterol. 2006;41:971–980. doi: 10.1007/s00535-006-1883-1. [DOI] [PubMed] [Google Scholar]
- Kane KA, Robinson GB. Effect of chronic nimodipine on spatial learning and on long-term potentiation. Behav. Brain Res. 1999;98:95–101. doi: 10.1016/s0166-4328(98)00057-6. [DOI] [PubMed] [Google Scholar]
- Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, Glabe CG. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300:486–489. doi: 10.1126/science.1079469. [DOI] [PubMed] [Google Scholar]
- Kessler AR, Kessler B, Yehuda S. In vivo modulation of brain cholesterol level and learning performance by a novel plant lipid: indication of interactions between hippocampal-cortical cholesterol and learning. Life Sci. 1986;38:1185–1192. doi: 10.1016/0024-3205(86)90173-6. [DOI] [PubMed] [Google Scholar]
- Kim J-H, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer’s disease. Neuron. 2009;63:287–303. doi: 10.1016/j.neuron.2009.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim KW, Youn JC, Jhoo JH, Lee DY, Lee KU, Lee JH, Woo JI. Apolipoprotein E ε4 Allele is not associated with the cognitive impairment in community-dwelling normal elderly individuals. Int. J. Geriatr. Psychiatry. 2002;17:635–640. doi: 10.1002/gps.664. [DOI] [PubMed] [Google Scholar]
- Kim WS, Weickert CS, Garner B. Role of ATP-binding cassette transporters in brain lipid transport and neurological disease. J. Neurochem. 2008;104:1145–1166. doi: 10.1111/j.1471-4159.2007.05099.x. [DOI] [PubMed] [Google Scholar]
- Kirsch C, Eckert GP, Koudinov AR, Muller WE. Brain cholesterol, statins and Alzheimer’s disease. Pharmacopsychiatry. 2003;36:S113–S119. doi: 10.1055/s-2003-43058. [DOI] [PubMed] [Google Scholar]
- Kivipelto M, Helkala E-L, Hanninen T, Laakso MP, Hallikainen M, Alhainen K, Soininen H, Tuomilehto J, Nissinen A. Midlife vascular risk factors and late-life mild cognitive impairment: a population-based study. Neurology. 2001;56:1683–1689. doi: 10.1212/wnl.56.12.1683. [DOI] [PubMed] [Google Scholar]
- Koladiya RU, Jaggi AS, Singh N, Sharma BK. Ameliorative role of atorvastatin and pitavastatin in L-methionine induced vascular dementia in rats. BMC Pharmacolgy. 2008;9:8–14. doi: 10.1186/1471-2210-8-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolsch H, Heun R, Kerksiek A, Bergmann KV, Maier W, Lutjohann D. Altered levels of plasma 24S- and 27-hydroxycholesterol in demented patients. Neurosci. Lett. 2004;368:303–308. doi: 10.1016/j.neulet.2004.07.031. [DOI] [PubMed] [Google Scholar]
- Kotti TJ, Ramirez DMO, Pfeiffer BE, Huber KM, Russell DW. Brain cholesterol turnover required for geranylgeraniol production and learning in mice. Proceedings of the National Academy of Sciences, USA. 2006;103:3869–3874. doi: 10.1073/pnas.0600316103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koudinov AR, Koudinova NV. Essential role of cholesterol in synaptic plasticity and neuronal degeneration. FASEB J. 2001;15:1858–1860. doi: 10.1096/fj.00-0815fje. [DOI] [PubMed] [Google Scholar]
- Lacor PN, Buniel MC, Furlow PW, Clemente AS, Velasco PT, Wood M, Viola KL, Klein WL. Aβoligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J. Neurosci. 2007;27:796–807. doi: 10.1523/JNEUROSCI.3501-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LaFerla FM, Green KN, Oddo S. Intracellular amyloid-βin Alzheimer’s disease. Nature Reviews Neuroscience. 2007;8:499–509. doi: 10.1038/nrn2168. [DOI] [PubMed] [Google Scholar]
- Ledesma MD, Dotti CG. Amyloid excess in Alzheimer’s disease: what is cholesterol to be blamed for? FEBS Lett. 2006;580:5525–5532. doi: 10.1016/j.febslet.2006.06.038. [DOI] [PubMed] [Google Scholar]
- Lefterov I, Fitz NF, Cronican A, Lefterov P, Staufenbiel M, Koldamova R. Memory deficits in APP23/Abca1+/− mice correlate with the level of Abeta oligomers. ASN Neuro. 2009;1:xxx. doi: 10.1042/AN20090015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, Gallagher M, Ashe KH. A specific amyloid-βprotein assembly in the brain impairs memory. Nature. 2006;440:352–357. doi: 10.1038/nature04533. [DOI] [PubMed] [Google Scholar]
- Lesser GT, Haroutunian V, Purohit DP, Beeri MS, Schmeidler J, Honkanen L, Neufeld R, Libow L. Serum lipid levels are related to Alzheimer’s pathology in nursing home residents. Dement. Geriatr. Cogn. Disord. 2009;27:42–49. doi: 10.1159/000189268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li L, Cao D, Garber DW, Kim H, Fukuchi K. Association of aortic atherosclerosis with cerebral β-amyloidosis and learning deficits in a mouse model of Alzheimer’s disease. Am. J. Pathol. 2003;163:2155–2164. doi: 10.1016/s0002-9440(10)63572-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li L, Cao D, Kim H, Lester R, Fukuchi K. Simvastatin enhances learning and memory independent of amyloid load in mice. Ann. Neurol. 2006;60:729–739. doi: 10.1002/ana.21053. [DOI] [PubMed] [Google Scholar]
- 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]
- Liu F, Pardo LM, Schuur M, Sanchez-Juan P, Isaacs A, Sleegers K, de Koning I, Zorkoltseva IV, Axenovich TI, Wittman JCM, Janssens ACJW, van Sweiten JC, Aulchenko YS, Oostra BA, Van Duijn CM. The apolipoprotein E gene and its age-specific effects on cognitive function. Neurobiol. Aging. 2008;xx:xxx. doi: 10.1016/j.neurobiolaging.2008.09.015. [DOI] [PubMed] [Google Scholar]
- Liu Y, Peterson DA, Schubert D. Amyloid βpeptide alters intracellular vesicle trafficking and cholesterol homeostasis. Proceedings of the National Academy of Sciences, USA. 1998;95:13266–13271. doi: 10.1073/pnas.95.22.13266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lominska C, Levin JA, Wang J, Sikes J, Kao C, Smith JD. Apolipoprotein E deficiency effects on learning in mice are dependent upon the background strain. Behav. Brain Res. 2001;120:23–34. doi: 10.1016/s0166-4328(00)00365-x. [DOI] [PubMed] [Google Scholar]
- Lu D, Qu C, Goussev A, Jiang H, Lu C, Schallert T, Mahmood A, Chen J, Chopp M. Statins increase neurogenesis in the dentate gyrus, reduce delayed neuronal death in the hippocampal CA3 region, and improve spatial learning in rat after traumatic brain injury. J. Neurotrauma. 2007;24:1132–1146. doi: 10.1089/neu.2007.0288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu J, Wu D-M, Zheng Y-L, Sun D-X, Hu B, Shan Q, Zhang Z-F, Fan S-H. Trace amounts of copper exacerbate beta amyloid-induced neurotoxicity in the cholesterol-fed mice through TNF-mediated inflammatory pathway. Brain. Behav. Immun. 2009;23:193–203. doi: 10.1016/j.bbi.2008.09.003. [DOI] [PubMed] [Google Scholar]
- Lukiw WJ, Pappolla M, Pelaez RP, Bazan NG. Alzheimer’s disease - a dysfunction in cholesterol and lipid metabolism. Cell. Mol. Neurobiol. 2005;25:475–483. doi: 10.1007/s10571-005-4010-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma H, Lesne S, Kotilinek L, Steidl-Nicols JV, Sherman M, Younkin L, Younkin S, Forster C, Sergeant N, Delacourte A, Vassar R, Citron M, Kofuji P, Boland LM, Ashe KH. Involvment of β-site APP cleaving emzyme 1 (BACE1) in amyloid precusor protein-mediated enhancement of memory and activity-dependent synaptic plasticity. Proceedings of the National Academy of Sciences. 2007;104:8167–8172. doi: 10.1073/pnas.0609521104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahfouz MM, Kummerow FA. Cholesterol-rich diets have different effects on lipid peroxidation, cholesterol oxides, and antioxidant enzymes in rats and rabbits. Journal of Nutritional Biochemistry. 2000;11:293–302. doi: 10.1016/s0955-2863(00)00083-8. [DOI] [PubMed] [Google Scholar]
- Martins IJ, Berger T, Sharman MJ, Verdile G, Fuller SJ, Marins RN. Cholesterol metabolism and transport in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2009;111:1275–1308. doi: 10.1111/j.1471-4159.2009.06408.x. [DOI] [PubMed] [Google Scholar]
- Masse I, Bordet R, Deplanque D, Al Khedr A, Richard F, Libersa C, Pasquier F. Lipid lowering agents are associated with a slower cognitive decline in Alzheimer’s disease. Journal of Neurology Neurosurgery and Psychiatry. 2005;76:1624–1629. doi: 10.1136/jnnp.2005.063388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayeux R, Small SA, Tang M-X, Tycko B, Stern Y. Memory performance in healthy elderly without Alzheimer’s disease: effects of time and apolipoprotein-E. Neurobiol. Aging. 2001;22:683–689. doi: 10.1016/s0197-4580(01)00223-8. [DOI] [PubMed] [Google Scholar]
- McDowell I, Kristjansson E. Mental status testing. In: McDowell I, Newell C, editors. Measuring health: a guide to rating scales and questionnaires. Oxford University Press; New York: 1996. pp. 287–334. [Google Scholar]
- McLaurin J, Darabie AA, Morrison MR. Cholesterol, a modulator of membrane-associated aβ-fibrillogenesis. Pharmacopsychiatry. 2003;36:S130–S135. doi: 10.1055/s-2003-43054. [DOI] [PubMed] [Google Scholar]
- Micale V, Scapagnini G, Colombrita C, Mazzola C, Alkon DL, Drago F. Behavioral effects of dietary cholesterol in rats tested in experimental models of mild stress and cognition tasks. Eur. Neuropsychopharmacol. 2008;18:462–471. doi: 10.1016/j.euroneuro.2007.11.006. [DOI] [PubMed] [Google Scholar]
- Mielke MM, Zandi PP, Sjogren M, Gustafson D, Ostling S, Steen B, Skoog I. High total cholesterol levels in late life associated with a reduced risk of dementia. Neurology. 2005;64:1689–1695. doi: 10.1212/01.WNL.0000161870.78572.A5. [DOI] [PubMed] [Google Scholar]
- Miller S, Wehner JM. Cholesterol treatment facilitates spatial learning performance in DBA/2Ibg mice. Pharmacology Biochemistry and Behavior. 1994;49:257–261. doi: 10.1016/0091-3057(94)90487-1. [DOI] [PubMed] [Google Scholar]
- Morgan D. Learning and memory deficits in APP transgenic mouse models of amyloid deposition. Neurochem. Res. 2003;28:1029–1034. doi: 10.1023/a:1023255106106. [DOI] [PubMed] [Google Scholar]
- Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J. Neurosci. Methods. 1984;11:47–60. doi: 10.1016/0165-0270(84)90007-4. [DOI] [PubMed] [Google Scholar]
- Mulder M, Jansen PJ, Janssen BJ, van de Berg WD, van der Boom H, Havekes LM, de Kloet RE, Ramaekers FC, Blokland A. Low-density lipoprotein receptor-knockout mice display impaired spatial memory associated with a decreased synaptic density in the hippocampus. Neurobiol. Dis. 2004;16:212–219. doi: 10.1016/j.nbd.2004.01.015. [DOI] [PubMed] [Google Scholar]
- Muldoon MF, Barger SD, Ryan CM, Flory JD, Lehoczky JP, Matthews KA, Manuck SB. Effects of lovastatin on cognitive function and psychological well-being. Am. J. Med. 2000;108:538–547. doi: 10.1016/s0002-9343(00)00353-3. [DOI] [PubMed] [Google Scholar]
- Muldoon MF, Ryan CM, Matthews KA, Manuck SB. Serum cholesterol and intellectual performance. Psychosom. Med. 1997;59:382–387. doi: 10.1097/00006842-199707000-00008. [DOI] [PubMed] [Google Scholar]
- Näslund J, Haroutunian V, Mohs R, Davis KL, Davies P, Greengard P, Buxbaum JD. Correlation between elevated levels of amyloid β-peptide in the brain and cognitive decline. JAMA. 2000;283:1571–1577. doi: 10.1001/jama.283.12.1571. [DOI] [PubMed] [Google Scholar]
- Nayler WG. Review of preclinical data of calcium channel blockers and atherosclerosis. J. Cardiovasc. Pharmacol. 1999;33:S7–S-11. doi: 10.1097/00005344-199900002-00003. [DOI] [PubMed] [Google Scholar]
- Notkola I-L, Sulkava R, Pekkanen J, Erkinjunti T, Ehnholm C, Kivinen P, Tuomilehto J, Nissinen A. Serum total cholesterol, apolipoprotein Eε4 allele, and Alzheimer’s disease. Neuroepidemiology. 1998;17:14–20. doi: 10.1159/000026149. [DOI] [PubMed] [Google Scholar]
- O’Brien WT, Xu G, Batta A, Tint GS, Salen G, Dyer CA, Kendler A, Servatius RJ. Developmental sensitivity of associative learning to cholesterol synthesis inhibitors. Behav. Brain Res. 2002;129:141–152. doi: 10.1016/s0166-4328(01)00342-4. [DOI] [PubMed] [Google Scholar]
- Obrenovich ME, Joseph JA, Atwood CS, Perry G, Smith MA. Amyloid-β: a (life) preserver for the brain. Neurobiol. Aging. 2002;23:1097–1099. doi: 10.1016/s0197-4580(02)00038-6. [DOI] [PubMed] [Google Scholar]
- Panza F, D’Introno A, Colacicco AM, Capurso C, Pichichero G, Capurso SA, Capurso A, Solfrizzi V. Lipid metabolism in cognitive decline and dementia. Brain Research Reviews. 2006;51:275–292. doi: 10.1016/j.brainresrev.2005.11.007. [DOI] [PubMed] [Google Scholar]
- Papassotiropoulos A, Lutjohann D, Bagli M, Locatelli S, Jessen F, Buschfort R, Ptok U, Bjorkhem I, von Bergmann K, Heun R. 24S-hydroxycholesterol in cerebrospinal fluid is elevated in early stages of dementia. J. Psychiatr. Res. 2002;36:27–32. doi: 10.1016/s0022-3956(01)00050-4. [DOI] [PubMed] [Google Scholar]
- Papassotiropoulos A, Wollmer MA, Tsolaki M, Brunner F, Molyva D, Lutjohann D, Nitsch R, Hock C. A cluster of cholesterol-related genes confers susceptibility for Alzheimer’s disease. J. Clin. Psychiatry. 2005;66:940–947. [PubMed] [Google Scholar]
- Parale GP, Baheti NN, Kulkarni PM, Panchal NV. Effects of atorvastatin on higher functions. Eur. J. Clin. Pharmacol. 2006;62:259–265. doi: 10.1007/s00228-005-0073-z. [DOI] [PubMed] [Google Scholar]
- Parkinson PF, Kannangara TS, Eadie BD, Burgess BL, Wellington CL, Christie BR. Cognition, learning behaviour and hippocampal synaptic plasticity are not disrupted in mice over-expressing the cholesterol transporter ABCG1. Lipids in Health and Disease. 2009;8:xxx. doi: 10.1186/1476-511X-8-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pavlov IP. In: Conditioned reflexes: An investigation of the physiological activity of the cerebral cortex. Anrep GV, translator. Oxford University Press; London: 1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pearson HA, Peers C. Physiological roles for amyloid βpeptides. J. Physiol. (Lond) 2006;575:5–10. doi: 10.1113/jphysiol.2006.111203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perry LA, Stigger CB, Ainsworth BE, Zhang J. No association between cognitive acheivements, academic performance and serum cholesterol concentrations among school-aged children. Nutritional Neuroscience. 2009;12:160–166. doi: 10.1179/147683009X423328. [DOI] [PubMed] [Google Scholar]
- Pfrieger FW. Outsourcing in the brain: do neurons depend on cholesterol delivery by astrocytes? BioEssays. 2002;25:72–78. doi: 10.1002/bies.10195. [DOI] [PubMed] [Google Scholar]
- Pickering M, O’Connor JJ. Pro-inflammatory cytokines and their effects in the dentate gyrus. Prog. Brain Res. 2007;163:339–354. doi: 10.1016/S0079-6123(07)63020-9. [DOI] [PubMed] [Google Scholar]
- Poirier J. Apolipoprotein E, cholesterol transport and synthesis in sporadic Alzheimer’s disease. Neurobiol. Aging. 2005;26:355–361. doi: 10.1016/j.neurobiolaging.2004.09.003. [DOI] [PubMed] [Google Scholar]
- Qiu S, Korwek KM, Weeber EJ. A fresh look at an ancient family: emerging roles for low density lipoprotein receptors in synaptic plasticity and memory formation. Neurobiol. Learn. Mem. 2006;85:16–29. doi: 10.1016/j.nlm.2005.08.009. [DOI] [PubMed] [Google Scholar]
- Quartermain D. Chronic administration of the Ca2+ channel blocker amlodipine facilitates learning and memory in mice. Eur. J. Pharmacol. 2000;399:57–63. doi: 10.1016/s0014-2999(00)00368-x. [DOI] [PubMed] [Google Scholar]
- Quartermain D, deSoria VG, Kwan A. Calcium channel antagonists enhance retention of passive avoidance and maze learning in mice. Neurobiol. Learn. Mem. 2001;75:77–90. doi: 10.1006/nlme.1999.3958. [DOI] [PubMed] [Google Scholar]
- Rader DJ, Daugherty A. Translating molecular discoveries into new therapies for atherosclerosis. Nature. 2008;451:904–913. doi: 10.1038/nature06796. [DOI] [PubMed] [Google Scholar]
- Raffai RL, Weisgraber KH. Cholesterol: from heart attacks to Alzheimer’s disease. J. Lipid Res. 2003;44:1423–1430. doi: 10.1194/jlr.R300007-JLR200. [DOI] [PubMed] [Google Scholar]
- Rafnsson SB, Deary IJ, Fowkes FG. Peripheral arterial disease and cognitive function. Vasc. Med. 2009;14:51–61. doi: 10.1177/1358863X08095027. [DOI] [PubMed] [Google Scholar]
- Reid PC, Urano Y, Kodama T, Hamakubo T. Alzheimer’s disease: cholesterol, membrane rafts, isoprenoids and statins. Journal of Cellular and Molecular Medicine. 2007;11:383–392. doi: 10.1111/j.1582-4934.2007.00054.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reitan RM, Shipley RE. The relationship of serum cholesterol changes to psychological abilities. J. Gerontol. 1963;18:350–357. doi: 10.1093/geronj/18.4.350. [DOI] [PubMed] [Google Scholar]
- Reynolds CA, Hong M-G, Eriksson UK, Blennow K, Bennet AM, Johansson B, Malmburg B, Berg S, Wiklund F, Gatz M, Pedersen NL, Prince JA. A survey of ABCA1 sequence variation confirms association with dementia. Hum. Mutat. 2009;30:1–7. doi: 10.1002/humu.21076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reynolds CA, Prince JA, Feuk L, Brookes AJ, Gatz M, Pedersen NL. Longitudinal memory performance during normal aging: twin association models of APOE and other Alzheimer candidate genes. Behav. Genet. 2006;36:185–194. doi: 10.1007/s10519-005-9027-6. [DOI] [PubMed] [Google Scholar]
- Riddell DR, Zhou H, Comery TA, Kouranova E, Lo CF, Warwick HK, Ring RH, Kirksey Y, Aschmies S, Xu J, Kubek K, Hirst WD, Gonzales C, Chen Y, Murphy E, Leonard S, Vasylyev D, Oganesian A, Martone RL, Pangalos MN, Reinhart PH, Jacobsen JS. The LXR agonist TO901317 selectively lowers hippocampal Aβ42 and improves memory in the Tg2576 mouse model of Alzheimer’s disease. Mol. Cell. Neurosci. 2007;34:621–628. doi: 10.1016/j.mcn.2007.01.011. [DOI] [PubMed] [Google Scholar]
- Robakis NK. Are Aβand Its derivatives causative agents or innocent bystanders in AD? Neurodegenerative Diseases. 2010;7:32–37. doi: 10.1159/000266476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson SR, Bishop GM. Aβas a bioflocculant: implications for the amyloid hypothesis of Alzheimer’s disease. Neurobiol. Aging. 2002;23:1051–1072. doi: 10.1016/s0197-4580(01)00342-6. [DOI] [PubMed] [Google Scholar]
- Rodriguez E, Mateo I, Llorca J, Sanchez-Quintana C, Infante J, Berciano J, Combarros O. No association between low density lipoprotein receptor genetic variants and Alzheimer’s disease risk. American Journal of Medical Genetics (Neuropsychiatric Genetics) 2006;141B:541–543. doi: 10.1002/ajmg.b.30341. [DOI] [PubMed] [Google Scholar]
- Roher AE, Kuo Y-M, Kokjohn KM, Emmerling MR, Gracon S. Amyloid and lipids in the pathology of Alzheimer disease. Amyloid: Journal of Protein Folding Disorders. 1999;6:136–145. doi: 10.3109/13506129909007315. [DOI] [PubMed] [Google Scholar]
- Romero JR, Beiser A, Seshardi S, Benjamin J, Polak JF, Vasan RS, Au R, DeCarli C, Wolf PA. Carotid artery atherosclerosis, MRI indices of brain ischemia, aging, and cognitive impairment: the Framingham study. Stroke. 2009;40:1590–1596. doi: 10.1161/STROKEAHA.108.535245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ronald JA, Chen Y, Bernas L, Kizler HH, Rogers KA, Hegele RA, Rutt BK. Clinical field-strength MRI of amyloid plaques induced by low-level cholesterol feeding in rabbits. Brain. 2009;132:1346–1354. doi: 10.1093/brain/awp031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russell DW, Halford RW, Ramirez DMO, Shah R, Kotti T. Cholesterol 24-hydroxylase: an enzyme of cholesterol turnover in the brain. Annu. Rev. Biochem. 2009;78:1017–1040. doi: 10.1146/annurev.biochem.78.072407.103859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sagare A, Deane R, Bell RD, Johnson B, Hamm K, Pendu R, Marky A, Lenting PJ, Wu Z, Zarcone T, Goate A, Mayo K, Perlmutter D, Coma M, Zhong Z, Zlokovic BV. Clearance of amyloid-βby circulating lipoprotein receptors. Nat. Med. 2007;13:1029–1031. doi: 10.1038/nm1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salo A, Ylikoski R, Verkkoniemi A, Polvikoski T, Juva K, Rastas S, Kontula K, Kainulainen K, Niinisto L, Notkola I-L, Sulkava R. Does apolipoprotein E influence learning and memory in the nondemented oldest old? Int. Psychogeriatr. 2002;13:451–459. doi: 10.1017/s1041610201007864. [DOI] [PubMed] [Google Scholar]
- Savory J, Ghribi O, Herman MM. Is amyloid β-peptide neurotoxic or neuroprotective and what is its role in the binding of metal ions? Neurobiol. Aging. 2002;23:1089–1092. doi: 10.1016/s0197-4580(02)00037-4. [DOI] [PubMed] [Google Scholar]
- Schachter M. Calcium antagonists and atherosclerosis. Int. J. Cardiol. 1997;62:S9–S15. doi: 10.1016/s0167-5273(97)00236-2. [DOI] [PubMed] [Google Scholar]
- Schafers M, Sorkin L. Effects of cytokines on neuronal excitability. Neurosci. Lett. 2008;437:188–193. doi: 10.1016/j.neulet.2008.03.052. [DOI] [PubMed] [Google Scholar]
- Schmajuk NA, Christiansen BA. Eyeblink conditioning in rats. Physiology & Behavior. 1990;48:755–758. doi: 10.1016/0031-9384(90)90221-o. [DOI] [PubMed] [Google Scholar]
- Schreurs BG, Smith-Bell CA, Darwish DS, Stankovic G, Sparks DL. Classical conditioning of the rabbit’s nictitating membrane response is a function of the duration of dietary cholesterol. Nutritional Neuroscience. 2007a;10:159–168. doi: 10.1080/10284150701565540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schreurs BG, Smith-Bell CA, Darwish DS, Stankovic G, Sparks DL. High dietary cholesterol facilitates classical conditioning of the rabbit’s nictitating membrane response. Nutritional Neuroscience. 2007b;10:31–43. doi: 10.1080/10284150701232034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schreurs BG, Smith-Bell CA, Darwish DS, Wang D, Burhans L, Gonzales-Joekes J, Deci S, Stankovic G, Sparks DL. Cholesterol enhances classical conditioning of the rabbit heart rate response. Behav. Brain Res. 2007c;181:52–63. doi: 10.1016/j.bbr.2007.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schreurs BG, Smith-Bell CA, Lochhead J, Sparks DL. Cholesterol modifies classical conditioning of the rabbit (Oryctolagus cuniculus) nictitating membrane response. Behav. Neurosci. 2003;117:1220–1232. doi: 10.1037/0735-7044.117.6.1220. [DOI] [PubMed] [Google Scholar]
- Servatius RJ, Beck KD. Facilitated acquisition of the classically conditioned eyeblink response in male rats after systemic IL-1β. Integr. Physiol. Behav. Sci. 2003;38:169–178. doi: 10.1007/BF02688851. [DOI] [PubMed] [Google Scholar]
- Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL. Natural oligomers of the Alzheimer amyloid-βprotein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J. Neurosci. 2007;27:2866–2875. doi: 10.1523/JNEUROSCI.4970-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, Regan CM, Walsh DM, Sabatini BL, Selkoe DJ. Amyloid-βprotein dimers isolated directly from Alzheimer’s brains impairs synaptic plasticity and memory. Nat. Med. 2008;14:837–842. doi: 10.1038/nm1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma S, Prasanthi J, Schommer E, Feist G, Ghribi O. Hypercholesterolemia-induced Aβaccumulation in rabbit brain is associated with alteration in IGF-1 signaling. Neurobiol. Dis. 2008;32:426–432. doi: 10.1016/j.nbd.2008.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simons M, Keller P, Dichgans J, Schulz JB. Cholesterol and Alzheimer’s disease: Is there a link? Neurology. 2001;57:1089–1093. doi: 10.1212/wnl.57.6.1089. [DOI] [PubMed] [Google Scholar]
- Singh-Manoux A, Gimeno D, Kivimaki M, Brunner E, Marmot MG. Low HDL cholesterol is a risk factor for deficit and decline in memory in midlife: the Whitehall II study. Arteriosclerosis, Thrombosis and Vascular Biology. 2008;28:1556–1562. doi: 10.1161/ATVBAHA.108.163998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sjogren M, Mielke M, Gustafson D, Zandi P, Skoog I. Cholesterol and Alzheimer’s disease - is there a relation? Mech. Ageing Dev. 2006;127:138–147. doi: 10.1016/j.mad.2005.09.020. [DOI] [PubMed] [Google Scholar]
- Skelton RW. Bilateral cerebellar lesions disrupt conditioned eyelid responses in unrestrained rats. Behav. Neurosci. 1988;102:586–590. doi: 10.1037//0735-7044.102.4.586. [DOI] [PubMed] [Google Scholar]
- Small BJ, Graves AB, McEvoy CL, Crawford FC, Mullan M, Mortimer JA. Is ApoE-ε4 a risk factor for cognitive impairment in normal aging? Neurology. 2000;54:2082–2088. doi: 10.1212/wnl.54.11.2082. [DOI] [PubMed] [Google Scholar]
- Solomon A, Kareholt I, Ngandu T, Winblad B, Nissinen A, Tuomilehto J, Soininen H, Kivipelto M. Serum cholesterol changes after midlife and late-life cognition. Neurology. 2007;68:751–756. doi: 10.1212/01.wnl.0000256368.57375.b7. [DOI] [PubMed] [Google Scholar]
- Solomon A, Kareholt I, Ngandu T, Wolozin B, MacDonald SWS, Winblad B, Nissinen A, Tuomilehto J, Soininen H, Kivipelto M. Serum cholesterol, statins and cognition in non-demented elderly. Neurobiol. Aging. 2009a;30:1006–1009. doi: 10.1016/j.neurobiolaging.2007.09.012. [DOI] [PubMed] [Google Scholar]
- Solomon A, Kivipelto M. Cholesterol-modifying strategies for Alzheimer’s disease. Expert Reviews in Neurotherapeutics. 2009;9:695–709. doi: 10.1586/ern.09.25. [DOI] [PubMed] [Google Scholar]
- Solomon A, Kivipelto M, Wolozin B, Zhou J, Whitmer RA. Midlife serum cholesterol and increased risk of Alzheimer’s and vascular dementia three decades later. Dement. Geriatr. Cogn. Disord. 2009b;28:75–80. doi: 10.1159/000231980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sparks DL. Coronary artery disease, hypertension, ApoE and cholesterol: a link to Alzheimer’s disease. Annals New York Academy of Sciences. 1997;826:128–146. doi: 10.1111/j.1749-6632.1997.tb48466.x. [DOI] [PubMed] [Google Scholar]
- Sparks DL. Cholesterol, copper, and accumulation of thioflavine s-reactive Alzheimer’s-like amyloid βin rabbit brain. J. Mol. Neurosci. 2004;24:97–104. doi: 10.1385/jmn:24:1:097. [DOI] [PubMed] [Google Scholar]
- Sparks DL. Cholesterol metabolism and brain amyloidosis: evidence for a role of copper in the clearance of Aβthrough the liver. Current Alzheimer Research. 2007;4:165–169. doi: 10.2174/156720507780362119. [DOI] [PubMed] [Google Scholar]
- Sparks DL, Connor DJ, Browne PJ, Lopez JE, Sabbagh MN. HMG-CoA reductase inhibitors (statins) in the treatment of Alzheimer’s disease and why it would be ill-advised to use one that crosses the blood-brain barrier. Journal of Nutrition, Health and Aging. 2002a;6:324–331. [PubMed] [Google Scholar]
- 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. 2006;114:s3–s7. doi: 10.1111/j.1600-0404.2006.00690.x. [DOI] [PubMed] [Google Scholar]
- 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. Current Alzheimer Research. 2008;5:416–421. doi: 10.2174/156720508785132316. [DOI] [PubMed] [Google Scholar]
- Sparks DL, Kuo Y-M, Roher AE, Martin TA, Lukas RJ. Alterations of Alzheimer’s disease in the cholesterol-fed rabbit, including vascular inflammation. Preliminary observations. Annals New York Academy of Sciences. 2000;903:335–344. doi: 10.1111/j.1749-6632.2000.tb06384.x. [DOI] [PubMed] [Google Scholar]
- Sparks DL, Lochhead J, Horstman D, Wagoner T, Martin T. Water quality has a pronounced effect on cholesterol-induced accumulation of Alzheimer amyloid (aβ) in rabbit brain. Journal of Alzheimer’s Disease. 2002b;4:523–529. doi: 10.3233/jad-2002-4609. [DOI] [PubMed] [Google Scholar]
- Sparks DL, Martin T, Stankovic G, Waggoner T, Van Adel R. Influence of water quality on cholesterol induced systemic pathology. The Journal of Nutrition, Health & Aging. 2007;11:189–193. [PubMed] [Google Scholar]
- Sparks DL, Scheff SW, Hunsaker JC, III, Liu H, Landers T, Gross DR. Induction of Alzheimer-like β-amyloid immunoreactivity in the brains of rabbits with dietary cholesterol. Exp. Neurol. 1994;126:88–94. doi: 10.1006/exnr.1994.1044. [DOI] [PubMed] [Google Scholar]
- Sparks DL, Schreurs BG. Trace amounts of copper in water induce β-amyloid plaques and learning deficits in a rabbit model of Alzheimer’s disease. Proceedings of the National Academy of Sciences, USA. 2003;100:11065–11069. doi: 10.1073/pnas.1832769100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stewart R, Russ C, Richards M, Brayne C, Lovestone S, Mann A. Apolipoprotein E genotype, vascular risk and early cognitive impairment in an African Caribbean population. Dement. Geriatr. Cogn. Disord. 2001;12:251–256. doi: 10.1159/000051267. [DOI] [PubMed] [Google Scholar]
- Teunissen CE, de Vente J, von Bergmann K, Bosma H, Van Boxtel MPJ, De Bruijn C, Jolles J, Steinbusch HWM, Lutjohann D. Serum cholesterol, precursors and metabolites and cognitive performance in an aging population. Neurobiol. Aging. 2003;24:147–155. doi: 10.1016/s0197-4580(02)00061-1. [DOI] [PubMed] [Google Scholar]
- 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]
- Thirumangalakudi L, Prakasam A, Zhang R, Bimonte-Nelson HA, Sambamurti K, Kindy MS, Bhat NR. High cholesterol-induced neuroinflammation and amyloid precursor protein processing correlate with the loss of working memory in mice. J. Neurochem. 2008;106:475–485. doi: 10.1111/j.1471-4159.2008.05415.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Upchurch M, Wehner JM. DBA/2Ibg mice are incapable of cholinergically-based learning in the Morris water maze. Pharmacology Biochemistry & Behavior. 1988;29:325–329. doi: 10.1016/0091-3057(88)90164-5. [DOI] [PubMed] [Google Scholar]
- van den Kommer TN, Dik MG, Comijs HC, Fassbender K, Lutjohann D, Jonker C. Total cholesterol and oxysterols: early markers for cognitive decline in elderly? Neurobiol. Aging. 2009;30:534–545. doi: 10.1016/j.neurobiolaging.2007.08.005. [DOI] [PubMed] [Google Scholar]
- van der Staay FJ, Steckler T. Behavioral phenotyping of mouse mutants. Behav. Brain Res. 2001;125:3–12. doi: 10.1016/s0166-4328(01)00278-9. [DOI] [PubMed] [Google Scholar]
- van Exel E, de Craen AJM, Gussekloo J, Houx P, Bootsma-van der Weil A, Macfarlane PW, Blauw GJ, Westendorp RGJ. Association between high-density lipoprotein and cognitive impairment in the oldest old. Ann. Neurol. 2002;51:716–721. doi: 10.1002/ana.10220. [DOI] [PubMed] [Google Scholar]
- Vanmierlo T, Rutten K, Dederen J, Bloks VW, van Vark-van der Zee LC, Kuipers F, Kiliaan A, Blokland A, Sijbrands EJG, Steinbusch H, Prickaerts J, Lutjohann D, Mulder M. Liver × receptor activation restores memory in aged AD mice without reducing amyloid. Neurobiol. Aging. 2009 doi: 10.1016/j.neurobiolaging.2009.07.005. In press. [DOI] [PubMed] [Google Scholar]
- Veinbergs I, Everson A, Sagara Y, Masliah E. Neurotoxic effects of apolipoprotein E4 are mediated via dysregulation of calcium homeostasis. J. Neurosci. Res. 2002;67:379–387. doi: 10.1002/jnr.10138. [DOI] [PubMed] [Google Scholar]
- Veinbergs I, Mallory M, Sagara Y, Masliah E. Vitamin E supplementation prevents spatial learning deficits and dendritic alterations in aged apolipoprotein E-deficient mice. Eur. J. Neurosci. 2000;12:4541–4546. [PubMed] [Google Scholar]
- Viviani B, Gardoni F, Marinovich M. Cytokines and neuronal ion channels in health and disease. Int. Rev. Neurobiol. 2007;82:247–263. doi: 10.1016/S0074-7742(07)82013-7. [DOI] [PubMed] [Google Scholar]
- Voikar V, Rauvala H, Ikonen E. Cognitive deficit and development of motor impairment in a mouse model of Niemann-Pick type C disease. Behav. Brain Res. 2002;132:1–10. doi: 10.1016/s0166-4328(01)00380-1. [DOI] [PubMed] [Google Scholar]
- Wahrle S, Das P, Nyborg AC, McLendon C, Shoji M, Kawarabayashi T, Younkin LH, Younkin SG, Golde TE. Cholesterol-dependent γ-secretase activity in buoyant cholesterol-rich membrane microdomains. Neurobiol. Dis. 2002;9:11–23. doi: 10.1006/nbdi.2001.0470. [DOI] [PubMed] [Google Scholar]
- Walsh DM, Selkoe DJ. Aβoligomers - a decade of discovery. J. Neurochem. 2007;101:1172–1184. doi: 10.1111/j.1471-4159.2006.04426.x. [DOI] [PubMed] [Google Scholar]
- Wegiel J, Kuchna I, Nowicki K, Frackowiak J, Mazur-Kolecka B, Imaki H, Wegiel J, Mehta PD, Silverman WP, Reisberg B, deLeon M, Wisniewski T, Pirttilla T, Frey H, Lehtimaki T, Kivimaki T, Visser FE, Kamphorst W, Potempska A, Bolton D, Currie JR, Miller DL. Intraneuronal Aβimmunoreactivity is not a predictor of brain amyloidosis-βor neurofibrillary degeneration. Acta Neuropathol. (Berl) 2007;113:389–402. doi: 10.1007/s00401-006-0191-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West R, Beeri MS, Schmeidler J, Hannigan CM, Angelo G, Grossman HT, Rosendorff C, Silverman JM. Better memory functioning associated with higher total and low-density lipoprotein cholesterol levels in very elderly subjects without the apolipoprotein e4 allele. Am. J. Geriatr. Psychiatry. 2008;16:781–785. doi: 10.1097/JGP.0b013e3181812790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology. 2005;64:277–281. doi: 10.1212/01.WNL.0000149519.47454.F2. [DOI] [PubMed] [Google Scholar]
- Wild K, Howieson D, Webbe F, Seeyle A, Kaye J. Status of computerized cognitive testing in aging: a systematic review. Alzheimer’s & Dementia. 2008;4:428–437. doi: 10.1016/j.jalz.2008.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson RS, Schnieder JA, Barnes LL, Beckett LA, Aggarwal NT, Cochran EJ, Berry-Kravis E, Bach J, Fox JH, Evans DA, Bennett DA. The apolipoprotein E ε4 allele and decline in different cognitive systems during a 6-year period. Arch. Neurol. 2002;59:1154–1160. doi: 10.1001/archneur.59.7.1154. [DOI] [PubMed] [Google Scholar]
- Wollmer MA, Sleegers K, Ingelsson M, Zekanowski C, Brouwers N, Maruszak A, Brunner F, Huynh KD, Kilander L, Brudin RM, Hedlund M, Giedraitis V, Glaser A, Engelsborghs S, De Deyn PP, Kapaki E, Tsolaki M, Daniilidou M, Molyva D, Paraskevas GP, Thal DR, Barcikowska M, Kuznicki J, Lannfelt L, Van Broeckhoven C, Nitsch RM, Hock C, Papassotiropoulos A. Association study of cholesterol-related genes in Alzheimer’s disease. Neurogenetics. 2007;8:179–188. doi: 10.1007/s10048-007-0087-z. [DOI] [PubMed] [Google Scholar]
- Wolozin B, Kellman W, Celesia GC, Siegel G. Decreased prevalence of Alzheimer disease associated with 3-hydroxy-3-methyglutaryl coenzyme A reductase inhibitors. Arch. Neurol. 2000;57:1439–1443. doi: 10.1001/archneur.57.10.1439. [DOI] [PubMed] [Google Scholar]
- Wood WG, Igbavboa U, Eckert GP, Muller WE. Cholesterol - a Janus-faced molecule in the central nervous system. In: Reith NEA, editor. Handbook of Neurochemistry and Molecular Neurobiology: Neural Membrane and Transport. Springer; New York: 2007. pp. 152–171. [Google Scholar]
- Woodruff-Pak DS, Agelan A, Del Valle L. A rabbit model of Alzheimer’s disease: valid at neuropathological, cognitive, and therapeutic levels. Journal of Alzheimer’s Disease. 2007;11:371–383. doi: 10.3233/jad-2007-11313. [DOI] [PubMed] [Google Scholar]
- Woodruff-Pak DS, Chi J, Li Y-T, Pak MH, Fanelli RJ. Nimodipine ameliorates impaired eyeblink classical conditioning in order rabbits in the long-delay paradigm. Neurobiol. Aging. 1997;18:641–649. doi: 10.1016/s0197-4580(97)00159-0. [DOI] [PubMed] [Google Scholar]
- Wu C-W, Liao P-C, Lin C, Kuo C-J, Chen S-T, Cehn H-I, Kuo Y-M. Brain region-dependent increases in β-amyloid and apolipoprotein E levels in hypercholesterolemic rabbits. J. Neural Transm. 2003;110:641–649. doi: 10.1007/s00702-002-0809-1. [DOI] [PubMed] [Google Scholar]
- Xu G, Servatius RJ, Shefer S, Tint GS, O’Brien WT, Batta AK, Salen G. Relationship between abnormal cholesterol synthesis and retarded learning in rats. Metabolism. 1998;47:878–882. doi: 10.1016/s0026-0495(98)90130-5. [DOI] [PubMed] [Google Scholar]
- Xue Q-S, Sparks DL, Streit WJ. Microglial activation in the hippocampus of hypercholesterolemic rabbits occurs independent of increased amyloid production. Journal of Neuroinflammation. 2007;4:20–30. doi: 10.1186/1742-2094-4-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yaffe K, Barret-Connor E, Lin F, Grady D. Serum lipoprotein levels, statin use, and cognitive function in older women. Arch. Neurol. 2002;59:378–384. doi: 10.1001/archneur.59.3.378. [DOI] [PubMed] [Google Scholar]
- Yanagisawa K. Cholesterol and pathological processes in Alzheimer’s disease. J. Neurosci. Res. 2002;70:361–366. doi: 10.1002/jnr.10348. [DOI] [PubMed] [Google Scholar]
- Yao Z-X, Papadopoulos V. Function of β-amyloid in cholesterol transport: a lead to neurotoxicity. FASEB J. 2002;16:1677–1679. doi: 10.1096/fj.02-0285fje. [DOI] [PubMed] [Google Scholar]
- Yehuda S, Carasso RL. Modulation of learning, pain thresholds, and thermoregulation in the rat by preparations of free purified α-linolenic and linoleic acids: determination of the optimal ω3-to-ω6 ratio. Proceedings of the National Academy of Sciences, USA. 1993;90:10345–10349. doi: 10.1073/pnas.90.21.10345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yehuda S, Rabinovitz S, Motofsky DI. Modulation of learning and neuronal membrane composition in the rat by essential fatty acid preparation: time-course analysis. Neurochem. Res. 1998;23:627–634. doi: 10.1023/a:1022430620205. [DOI] [PubMed] [Google Scholar]
- Zandi PP, Sparks DL, Khachaturian AS, Tschanz JT, Norton M, Steinberg M, Wesl-Bohmer KA, Breitner JCS, Cache County Investigators Do statins reduce risk of incident dementia and Alzheimer’s disease? Arch. Gen. Psychiatry. 2005;62:217–224. doi: 10.1001/archpsyc.62.2.217. [DOI] [PubMed] [Google Scholar]
- Zatta P, Zambenedetti P, Stella MP, Licastro F. Astrocytosis, microgliosis, metallothionein-I-II and amyloid expression in high cholesterol-fed rabbits. Journal of Alzheimer’s Disease. 2002;4:1–9. doi: 10.3233/jad-2002-4101. [DOI] [PubMed] [Google Scholar]
- Zerbinatti CV, Wozniak DF, Cirrito J, Cam JA, Osaka H, Bales KR, Zhuo M, Paul SM, Holtzman DM, Bu G. Increased soluble amyloid-βpeptide and memory deficits in amyloid model mice overexpressing the low-density lipoprotein receptor-related protein. Proceedings of the National Academy of Sciences, USA. 2004;101:1075–1080. doi: 10.1073/pnas.0305803101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao S, Liao W, Xu N, Xu H, Yu C, Liu X, Li C. Polar metabolite of cholesterol induces rat cognitive dysfunctions. Neuroscience. 2009;164:398–403. doi: 10.1016/j.neuroscience.2009.08.027. [DOI] [PubMed] [Google Scholar]
- Zhou Y-T, Zhang Z-X, Chan P, He X-M, Tang M-N, Wu C-B, Hong Z. Genetic association between low-density lipoprotein receptor-related protein gene polymorphisms and Alzheimer’s disease in Chinese Han population. Neurosci. Lett. 2008;444:109–111. doi: 10.1016/j.neulet.2008.07.093. [DOI] [PubMed] [Google Scholar]
- Zou F, Gopalraj RK, Lok J, Zhu H, Ling IF, Simpson JF, Tucker HM, Kelly JF, Younkin SG, Dickson DW, Petersen RC, Graff-Radford NR, Bennet DA, Crook JE, Younkin SG, Estus S. Sex-dependent association of a common low-density lipoprotein receptor polymorphism with RNA splicing efficiency in the brain and Alzheimer’s disease. Hum. Mol. Genet. 2008;17:929–935. doi: 10.1093/hmg/ddm365. [DOI] [PMC free article] [PubMed] [Google Scholar]