Did you arrive here by via search engine?
Click here to view the original version of this article

Click to Print This Page
(This section will not print)

Lipoproteins and Apolipoproteins In The Brain

Course Authors

Simona Vuletic, M.D.

Dr. Vuletic is Acting Assistant Professor of Medicine, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA.

Within the past 12 months Dr. Vuletic reports no commercial conflicts of interest.

Albert Einstein College of Medicine, CCME staff and interMDnet staff have nothing to disclose.

Estimated course time: 1 hour(s).

Albert Einstein College of Medicine – Montefiore Medical Center designates this enduring material activity for a maximum of 1.0 AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

In support of improving patient care, this activity has been planned and implemented by Albert Einstein College of Medicine-Montefiore Medical Center and InterMDnet. Albert Einstein College of Medicine – Montefiore Medical Center is jointly accredited by the Accreditation Council for Continuing Medical Education (ACCME), the Accreditation Council for Pharmacy Education (ACPE), and the American Nurses Credentialing Center (ANCC), to provide continuing education for the healthcare team.

 
Learning Objectives

Upon completion of this Cyberounds®, you should be able to:

  • List basic differences between systemic and brain lipoprotein systems

  • Describe the origins of apolipoproteins and lipoproteins in the brain

  • Discuss physiological relevance of lipoprotein transfer and recycling in the brain

  • Apply their overview of the roles of specific apolipoproteins in the brain to physiological and pathophysiological processes.

 

Lipoproteins are extracellular macromolecular complexes composed of lipids, lipid-soluble molecules and proteins. Apolipoproteins are specialized lipoprotein-associated proteins that stabilize lipoprotein particles and enable transfer of hydrophobic molecules through an aqueous environment in intravascular and extracellular space.(1) Binding of apolipoproteins to specialized lipoprotein receptors on the cell surface facilitates bi-directional transfer of lipids and lipid-soluble molecules, such as vitamins E, D, K and A, between lipoprotein particles and cells. In this capacity, apolipoproteins are essential for regulation of cellular lipid composition, lipid recycling, removal of excess lipids from the cells, maintenance of cellular anti-oxidative potential and delivery of critical structural and functional components.

Presence of some apolipoproteins on lipoprotein particles is essential for activation of enzymes involved in lipid and lipoprotein metabolism; for example, apoC-II interacts with lipoprotein lipase (LPL).(2) Furthermore, binding of apolipoproteins to the lipoprotein receptors initiates signal transduction pathways, thus regulating numerous intracellular signaling events involved in inflammation, apoptosis and other physiological and pathophysiological cellular responses.

Recent studies have shown that some apolipoproteins such as apolipoprotein E (apoE) and apoD translocate to the nucleus following cellular reuptake,(3) but mechanisms responsible for the apolipoprotein translocation to the nucleus and their functions in the nuclear compartment are largely unknown. The expansion of apolipoprotein function from their role in lipid transfer to numerous other processes suggests a much more complex role for these proteins at the cellular level.

Lipoproteins in the brain significantly differ from the systemic lipoproteins.

Barriers

The brain is isolated from the systemic circulation by the blood-brain barrier (BBB) and blood-cerebrospinal fluid (CSF) barrier. The barriers physically separate the brain from the fluctuations of various blood components, and transfer across these barriers requires predominantly active transfer of the molecules that enter the brain tissue.(5) Blood represents a complex system, containing large number of circulating cells, has rich protein and lipid composition and is subjected to enormous fluctuations of various components. The composition of cerebrospinal fluid (CSF) is by comparison significantly simpler, more stable and distinct from blood. Therefore, the type of apolipoproteins and composition of lipoproteins present in the brain also significantly differ from the systemic lipoproteins.

Systemic Circulation

In the systemic circulation, apoB and apoA-I are two major apolipoproteins associated with lipoprotein particles, and define two principal categories of systemic lipoproteins, apoB-containing lipoproteins (LpB) and apoAI-containing lipoproteins (LpA-I). LpB [chylomicrons; very low density lipoproteins, VLDL; intermediate density lipoproteins, IDL; low density lipoproteins, LDL; and lipoprotein(a), Lp(a)] are typically more lipid-rich than high density lipoproteins, HDL, which contain apoA-I as the major apolipoprotein, and represent a highly diverse subpopulation of systemic lipoproteins that are relatively lipid-poor compared to LpB, and contain much more varied protein cargo.(6)(7)(8) Put simply, chylomicrons transport lipids absorbed from the intestinal tract, the VLDL-LDL cascade lipoproteins transport lipids to the cells, while HDL particles are involved in bi-directional lipid transfer between lipoproteins and between lipoproteins and cells.(9)

The HDL-dependent reverse cholesterol transfer, a process that transports cholesterol from the cells to liver for recycling or excretion through bile, is of particular importance for the maintenance of cellular lipid composition.(10) Although lipid transfer has long been thought to be the main function of HDL particles, recent proteomics studies showing a very heterogeneous protein composition of HDL particles suggest a much more complex metabolic role for this lipoprotein family.(6)(7)(8)

The Brain

In contrast, under normal circumstances, the brain does not contain LpB since apoB is not expressed in the brain, and does not cross the uncompromised BBB.(11) The main HDL apolipoprotein, apoA-I, is also not expressed in the brain but it crosses the BBB and is present on the brain lipoproteins.(12) Unlike apoA-I, apoE is expressed and secreted by the brain glial cells and represents the main apolipoprotein in the brain.(13)(14)(15) Besides apoE, other major apolipoproteins expressed in the brain are apoD and apoJ.(16)(17) Other apolipoproteins are also present, but in lower concentrations (Table 1), and recent studies implicate some of these minor apolipoprotein components in pathophysiological processes and brain diseases including schizophrenia, Alzheimer’s disease (AD) and Parkinson’s disease.(18)(19)(20)(21) Expression of the brain apolipoproteins is significantly different in various brain structures and regions.

Table 1. Apolipoproteins In The Brain.

Apolipoprotein CNS/CSF Source
ApoA-I Present Blood
ApoA-II Present Blood
ApoA-IV Present Brain
ApoB Absent N/A
ApoC-I Present Brain
ApoC-II Present Brain?
ApoC-III Present Brain?
ApoD Present Brain
ApoE Present Brain
ApoF Present Brain
ApoJ (clusterin) Present Brain
ApoL Present Brain

Brain Lipoprotein System

Differences in apolipoprotein composition of the brain lipoprotein particles are accompanied by a marked difference in their lipid composition. Under normal conditions, brain lipoproteins have significantly lower levels of esterified cholesterol than similar particles in plasma, and their predominant phospholipid component is phosphatidylethanolamine. The functional significance of these differences is not well understood, and our understanding of the roles of the brain apolipoprotein and lipoprotein metabolism in health and disease is currently limited.

The brain is an exceptionally lipid-rich organ.

The brain is an exceptionally lipid-rich organ. Lipids represent approximately 50% of the brain’s dry-weight, and the brain contains a quarter of the total body cholesterol.(22)(23) Unlike the rest of the body, essentially all (>99.5%) of the brain cholesterol is unesterified.(24) In humans, virtually all of the brain cholesterol is locally synthesized.(22)(25) Most cholesterol forms do not cross the BBB into the brain with the minor exception of certain oxidized forms of cholesterol.(26)(27) Although the net-flux of oxidized forms of cholesterol into the brain may be minimal, long-term effects of this process on development of neurodegenerative diseases, such as AD, cannot be neglected and may at least partially explain the increased risk of neurodegenerative diseases, including AD, in people with hypercholesterolemia.(28)(29)

In adults, the brain synthesizes on average 30 µg of cholesterol per day, (22)(30) has an average turnover rate of 0.02% and long half-life (4-6 months to 5 years).(24) Removal of cholesterol from the brain is partially performed through cholesterol oxidation by the cholesterol 24(S)-hydroxylase, an enzyme expressed by selected neurons.(31) 24(S)-hydroxycholesterol is easily transferred through the BBB into the peripheral circulation and excreted through liver into bile.(32) The blood levels of 24(S)-hydroxycholesterol depend almost entirely on the cholesterol elimination from the brain and are used as an approximate measure of cholesterol turnover in the brain.(31) This mechanism is responsible for approximately 40% of the cholesterol removal from the brain; the remaining 60% is transferred through the BBB by an unknown mechanism.(33)

Role of Brain Lipids and Lipoproteins

Lipids are critical structural and functional components of the brain tissue, and transfer of lipids within the brain tissue is essential for the maintenance of its structural and functional integrity. Normal functioning of the brain requires constant recycling of lipids, which is dependent on lipoprotein metabolism. Neurons are particularly sensitive to alterations in lipoprotein metabolism because they synthesize relatively low levels of cholesterol and other lipids, and depend largely on lipid transfer from glial cells, which are the main source of lipid and protein components of the brain lipoproteins. (34)(35)(36)(37) Furthermore, maintenance of myelin sheath, which is exceptionally lipid-rich, is dependent on normal lipoprotein metabolism.(24) It is therefore not surprising that practically all brain diseases and disorders are accompanied by alterations in lipid and lipoprotein metabolism (for examples see (38)(39)).

By size and density, brain lipoproteins secreted by astrocytes resemble HDL particles and are, therefore, typically described as HDL-like. An in vitro study suggested that microglia secrete lipoprotein particles in size similar to LDL, with size, composition and shape distinct from those secreted by astrocytes.(40) Lipoproteins are secreted in a nascent form, mature in the extracellular space by acquisition of lipid and protein components, exchange or deliver lipids to neurons, finally reaching the CSF as mature lipoprotein particles. Just as the plasma lipoproteins represent a window into millions of lipoprotein-cell interactions with the liver and peripheral tissues, so too the CSF lipoproteins represent a window into the interactions between lipoproteins and cells in the brain.

Several published studies reported the composition of different lipoprotein particles secreted by glial cells in vitro or isolated from CSF.(40)(41)(42)(43)(44)(45)(46)(47)(48)(49) Our current knowledge of the brain lipoprotein components suggests that these particles have a very complex protein composition, and that there are numerous types of the brain lipoproteins based on the apolipoproteins associated with them. However, the functional significance of these different sub-classes of brain lipoproteins is for the most part currently unknown.

Lipoproteins play a critical role in lipid recycling, which is relevant not only for lipid delivery, but also for other functions, including formation and function of synapses and neuronal repair.(37)(50)(51)(52) Published studies have shown that brain lipoproteins bind β-amyloid and that lipoprotein-bound Aβ does not form fibrils, a process that is considered a pre-requisite for formation of amyloid plaques.(53)(54)(55) Furthermore, Aβ clearance from the brain appears to be lipoprotein-dependent.(56)(57)(58) Lipoproteins also represent a transport mechanism for lipid-soluble vitamins, thus playing a role in regulation of the brain anti-oxidative potential (see, for example, (59)). In systemic circulation, lipoproteins are also involved in neutralization of lipopolysaccharide(60) and likely have a similar role in the brain during Gram-negative infections.

Much of our knowledge regarding brain lipid and lipoprotein metabolism comes from studies in various animal models. It should be noted that the lipid and lipoprotein metabolism in the human brain exhibits some significant differences compared to animal models. For example, mice do not have naturally occurring apoE isoforms, which play important differential roles in lipoprotein metabolism in human brain (see, for example, (61)). Therefore, results of in vivo studies using mouse, rat or other animal models should be interpreted with caution, as they may not have direct relevance to the human brain lipid and lipoprotein metabolism.

In summary, the brain lipoprotein system - comprised of proteins, lipids and lipoprotein receptors - is markedly different from the systemic lipoproteins and functions in a significantly different environment. It is, therefore, likely that the brain apolipoproteins have unique functions that are specific for the nervous system, as well as functions similar to those in the peripheral tissues.

Apolipoproteins In The Brain

ApoE
ApoE is the main apolipoprotein in human brain. The elegant studies by Linton and colleagues have shown that all of the brain apoE is brain-derived.(15) ApoE in the brain is expressed and secreted by glial cells, predominantly astrocytes, (12)(14)(15)(33)(62)(63) and its interaction with ABCA1 is critical for normal lipidation and function of apoE-containing lipoproteins (LpE).(64)

ApoE plays a critical role in maintenance of the brain structural integrity.

The expression of apoE by neurons has been controversial. ApoE is not expressed by neurons under normal physiological conditions.(65)(66) Recently published studies have shown that apoE is expressed by certain neurons in response to injury.(67)(68) It should be noted that the majority of studies regarding neuronal apoE involved transgenic mice over-expressing human apoE (for example (69)). Mouse neurons may not contain the same regulatory mechanisms for suppression of human apoE expression, supported by the fact that mouse apoE mRNA was undetectable in neurons in these transgenic mice. Only a small number of studies evaluated constitutive apoE expressed by human neurons, in which apoE expression had to be induced by toxins or severe serum starvation.(67)(70)(71)herefore, although the claims regarding neuronal apoE expression are perpetuated in the published literature, it is unlikely that neurons express apoE under normal conditions. However, apoE can enter neurons through a receptor-mediated endocytosis(72)(73) and has been shown to translocate to the nucleus of neurons when in lipidated form.(4)

ApoE plays a critical role in maintenance of the brain structural integrity and neuronal repair.(74)(75) LpE in the brain is involved in both delivery and uptake of lipids. The uptake is considered important for lipid recycling, which is a process essential for normal synaptic structure and function.(37)(51) Furthermore, lipid uptake by LpE is also involved in removing lipids from the sites of damage, which is a pre-requisite for efficient neuronal repair.(76) The ability of apoE to interact with its receptors on neuronal membrane is responsible for the uptake of lipoprotein particles and is considered to be the main mechanism for delivery of lipids to neurons.(76)(77)(78) A study by Wang and colleagues suggested that apoE-mimetics can be used in therapy of the brain injury,(79) which is in line with the known apoE functions in the brain.

Human apoE is present in three isoforms – ε2, ε3 and ε4 – with genotype combinations [APOE2/2; 2/3; 2/4; 3/3; 3/4 and 4/4) that are functionally significant both in the brain and in the rest of the organism.(80)(81)(82)(83) ApoE2 contains cysteine residues at positions 112 and 158; apoE3 contains cysteine at position 112 and arginine at position 158; apoE4 contains arginine at both positions.(84)

These sequence differences are associated with significant three-dimensional and functional consequences.(85)(86) ApoE isoforms differ in their affinity for lipids, and the composition of LpE is isoform-specific (84)(86)(87)(88). Furthermore, apoE isoforms have different affinity for apoE receptors, which are involved in lipid delivery, with apoE2 binding to the receptors with much lower affinity than apoE3 or apoE4, which have similar affinity for apoE receptor binding (idem).

In contrast, all three apoE isoforms have significantly different ability to elicit lipid efflux from neurons, with apoE4 having the weakest lipid efflux ability and apoE2 the strongest.(89) These findings suggest that apoE interactions with the ATP-binding cassette transporters ABCA1 and ABCG1, which are responsible for lipid efflux in astrocytes and neurons, are strongly apoE isoform-dependent, while apoE interactions with the receptors involved in lipid delivery show virtually no difference between the apoE3 and apoE4 isoforms. Furthermore, apoE isoform-specific interactions with other receptors, such as NMDA (N-methyl-D-aspartate) receptor, have been reported.(90)(91)

ApoJ functions as a multi-talented molecular chaperone.

These isoform-dependent functional differences are likely relevant for the observed association of the apoE4 isoform with AD, Parkinson’s disease, multiple sclerosis, stroke, brain hemorrhage, anxiety; numerous neuromuscular diseases and other neuronal diseases and processes,(52)(92)(93)(94)(95)(96)(97)(98)(99) since reduced efflux ability may lead to a slow, yet significant, increase in the intracellular lipid levels and reduced efficiency of lipid recycling in neurons. Furthermore, apoE binding to the extracellular matrix components is isoform-specific,(100) suggesting a possibility that apoE isoform function in the extracellular space, as well as at the cellular level, may be significantly different.

Additionally, an intriguing study suggested that apoE3 and apoE4 also have different stability and/or retention in the brain tissue, since people with apoE3/4 phenotype had significantly higher levels of apoE4 than apoE3 in CSF despite similar expression levels of these two isoforms.(101) In contrast, Riddell and colleagues reported that levels of apoE4 were lower than those of apoE3 in mice expressing human apoE3/4, and that apoE4 had shorter half-life compared to apoE3 in vitro.(102)

ApoE2 has been associated with a lowered risk of AD, since the presence of at least one APOE2 allele appears to be protective against AD.(99)(103) In contrast, the presence of APOE4 is associated with an increased risk and earlier onset of AD and is still the strongest confirmed genetic risk factor in late-onset AD.(92)(104)(105)(106)ApoE plays a role of a molecular chaperone in the brain, transfers α-tocopherol, binds beta-amyloid, modulates cleavage of amyloid precursor protein, promotes proteolysis of beta-amyloid peptides, plays a role in removal of beta-amyloid from the brain, affects growth of neurons, regulates morphology of neuronal processes, affects synaptic function, and regulates numerous aspects of microtubule-associated protein tau, including its phosphorylation in neurons, and many of these functions are apoE isoform-sensitive.(107)(108)(109)(110)(111)(112)(113)(114)(115)(116)(117)(118)(119)(120)(121)(122)(123)(124)(125)(126)(127)(128)(129)(130).

Furthermore, presence of apoE4 has been associated with reduced blood flow, worse outcome of traumatic brain injury and stroke due to lower efficiency at neuronal repair, reduced brain volume, impaired memory in older subjects, altered transfer of electric potentials along axons (LTP), and greater susceptibility to neurodegeneration regardless of putative causative factors, suggesting that apoE isoforms have differential effects on many brain processes.(52)(91)(131)(132)(133)(134)(135)(136)(137)(138)

Among other confirmed or suspected roles of apoE, its function in repair of neurons appears to be of particular importance. Studies have shown that levels of apoE levels significantly increase following neuronal injury in vivo. (139)(140)(141)(142) ApoE-/- mice have insufficient recovery from neuronal injury(143) and an infusion of apoE ameliorated neuronal damage following cerebral ischemia, neurodegeneration and cognitive impairment in apoE-/- mice.(144)(145)

Despite the observed differences in apoE3/3 vs. apoE4-positive subjects in various physiological and pathophysiological processes, a study by Levi and colleagues suggests that the impact of an unfavorable APOE genotype can be overcome or attenuated by an enriched, challenging environment.(146)These findings are supported by observations that early and prolonged education, as well as regular physical, mental and social activity, significantly reduce the impact of the APOEε4 genetic risk factor on neurological diseases and disorders (for example (147)(148)(149)). In contrast, lack of education, sedentary lifestyle, unchallenging mental and impoverished social environments increase risks of neurological disorders in all people (idem), and likely even more so in those with apoE4.

ApoA-I
ApoA-I in the brain is derived from the systemic circulation(12)(62) and represents a significant apolipoprotein component of the brain lipoproteins. Some studies have shown that apoA-I is expressed and secreted by the endothelial cells comprising the BBB. ApoA-I mRNA has been reported in the spinal cord in animal models but it is unclear whether the same is true in humans.(150)(151)

Compared to plasma, apoA-I levels in the brain are very low (approximate values of 0.2-0.4 mg/dl in CSF vs. 120-160 mg/dl in plasma; 152, 153). Despite the low levels of apoA-I in the brain, its ability to interact with the ATP-binding cassette transporters, ABCA1 and ABCG1, in the brain is likely significant for normal functioning of brain cells.

ApoJ also plays an important role in neurological disorders.

These ATP transporters are involved in apolipoprotein lipidation and recycling of lipids within the brain tissue. Specific knockdown of the ABCA1 expression in the brain was associated with a compensatory increase in apoA-I in the brain in vivo, and the mice had significantly reduced synapse number, as well as disturbed synapse structure and function.(154) Mauch and colleagues suggested that recycling of cholesterol in the brain is critical for the synapse development and function.(51) In vivo studies confirmed the functional relevance of the brain ABCA1-dependent lipid efflux for normal brain function.(64)(154)

Given the fact that apoE interacts with the ABC transporters in the brain,(155)(156) the role of apoA-I in the brain is likely more complex than providing an additional ligand to these receptors. A new study by Wang and colleagues suggested that CSF levels of apoA-I are decreased in patients with Parkinson’s disease,(157) and a study by Huang and colleagues has shown decreased apoA-I levels in the brain, as well as in the periphery, in patients with schizophrenia.(158) A recent in vivo study suggested that an apoA-I mimetic peptide improved cognitive function and reduced amyloid burden in an AD mouse model, as combination treatment with the peptide and statins significantly reduced inflammation in the brain of these mice, while statin alone did not have such effect.(159) These findings suggest that apoA-I has specific physiological functions in the brain. Although mice lacking apoA-I have been engineered,(160) the CNS phenotypic characteristics have not been reported.

ApoD
ApoD is a member of the lipocalin family and is considered to be an atypical apolipoprotein due to a relative lack of α-helices.(161) It binds multiple hydrophobic molecules including arachidonic acid and steroid hormones.(161)(162) In humans, expression of apoD is low in neonates, and increases during development and under oxidative stress.(163) Although the highest levels of apoD expression are in glial cells (astrocytes, oligodendrocytes and microglia), neurons also express apoD.(163)(164)(165)(166) Neuronal injury significantly increases apoD expression in both central and peripheral neurons.(167)(168)(169) ApoD functions are likely tissue-specific, since its association with different ligands is tissue-dependent.

Physiological functions of apoD are still poorly understood but published studies have shown that it has transfer and proteolytic activity, regulates signal transduction and protects against oxidative stress.(170)(171) ApoD is considered neuroprotective and is most likely involved in tissue repair.(167)(172)(173) Interestingly, in vivo studies in Drosophila have shown that apoD homolog knock-out in glial cells is associated with reduced lifespan and decreased stress resistance, with the opposite results in the overexpression model.(174)(175) These findings have been confirmed in a mouse model, showing that apoD deficiency is associated with an increased level of the brain lipid peroxidation, reduced learning and locomotor abilities.(171)

Increased ApoD has been reported with stroke,(173) traumatic brain injury,(168) neurodegenerative diseases such as AD,(176) demyelinating diseases such as multiple sclerosis and chronic inflammatory demyelinating polyneuropathy,(177) viral encephalopathy(172) and in psychiatric diseases including schizophrenia and bipolar disorder.(178) Published studies reported increased levels of apoD mRNA and protein in AD,(176) and an association of apoD levels with the extent of the disease (Braak stage) and apoE genotype.(179) In AD, apoD levels were reportedly positively associated with neurofibrillary tangles (idem). Its relationship with amyloid plaques is unclear, since some studies reported no correlation,(179) while others reported that they correlate with compact, but not with diffuse, amyloid plaques.(180) These findings, combined with the known apoD functions, suggest that the observed increased levels of apoD in various brain disorders may be associated with an attempt to repair damaged neurons.

ApoJ (clusterin)
ApoJ is nearly ubiquitously expressed and is one of the more abundant apolipoproteins in the brain. ApoJ functions as a multi-talented molecular chaperone, and has many different functions in the periphery including lipid transfer, cell survival, tissue remodeling, cell-cell and cell-matrix interactions, and inhibition of complement activation.(181)(182) Although clusterin is considered a secreted protein, alternative splicing creates specific apoJ isoforms found in cytoplasmic and nuclear compartments.(183)(184)

ApoJ is expressed in the brain by both glial cells and a subset of neurons.(165)(185)(186) Through its role in cell survival (see section below on apolipoprotein-receptor interactions) apoJ plays a significant role in neuroprotection, and different apoJ forms can either promote or protect against apoptosis, depending on their intra- or extracellular localization.(182)(184)(187)(188) Retrotranslocation of apoJ has been reportedly induced by stress.(189) The intracellular forms have been found to play a role in DNA repair, transcription and organization of microtubules.(184)(190)(191)(192)(193) Neurons expressing apoJ have higher resistance to apoptosis.(194)

Probably the best researched correlation between apoJ and brain disorders is its relationship with AD. ApoJ mRNA is elevated in AD.(183) ApoJ binds Aβ peptides, preventing formation of fibrils, and is involved in its clearance from the brain.(55)(122)(129)(195)(196) Given these known apoJ functions, it is not surprising it plays an important role in processes protecting the brain against development of AD.(197) ApoJ genetic variants have been associated with an increased risk of AD, suggesting that structural alterations in those variants significantly affect its function in the brain and reduce its ability to protect the brain from pathophysiological processes involved in development of AD.(198)

ApoJ also plays an important role in other neurological disorders. For example, brain injury increases apoJ expression in vivo,(142)(199) and increased apoJ expression by astrocytes is associated with better survival following an ischemic injury in vivo.(200) An interesting study by Dati and colleagues has shown that recombinant human apoJ significantly accelerated remyelination and motor-evoked potentials of damaged nerves in peripheral neuropathy, suggesting strong neuronal regeneration effects in vivo.(201) ApoJ has also been reported to associate with α-synuclein, therefore playing a role in Parkinson’s disease, dementia with Lewy bodies and multiple system atrophy.(202)

ApoL
ApoL proteins are involved in regulation of apoptosis and immune response(203). ApoL proteins are encoded by six genes and multiple splice-isoforms.(204) All six genes are expressed in the brain; apoL1 and apoL2 are expressed in the frontal cortex neurons.(204)(205) It should be noted that of the six apoL proteins, only apoL1 contains signal peptide and is secreted outside of the cell, while the other apoL proteins are intracellular. (206) ApoL1 is associated with HDL in plasma and therefore fulfils the requirements for classification as an apolipoprotein.(207)(208) ApoL1 gene polymorphisms affect concentration of triglycerides in plasma(209) and apoL1 has specific role in protection against trypanosome infection.(210)

Elevated apoL levels and differential expression of APOL have been found in several neuropsychiatric diseases.(205)(211) For example, significant increases in apoL1, apoL2 and apoL4 expression have been shown in schizophrenia, while apoL2 is significantly increased in bipolar affective disorder.(205) Studies suggested that specific APOL single nucleotide polymorphisms are associated with an increased risk for development of schizophrenia.(18)(212) ApoL2 mRNA levels were reportedly also increased in prefrontal cortex of persons abusing drugs.(213) The mechanisms responsible for the observed correlations between apoL and psychiatric diseases are currently unknown. Interestingly, patients with schizophrenia have significantly different lipid composition of the cell membrane, not only in the brain but in peripheral cells as well, suggesting a systemic dysregulation of lipid and lipoprotein metabolism, and a possible role of apoL1 in pathophysiology of schizophrenia.(214)(215)

Apolipoprotein-receptor Interactions

Apolipoprotein interactions with receptors play a role not only in lipid transfer between lipoprotein particles and cells, but also initiate signal transduction cascades. For example, binding of apoE to its receptors, APOE2 or VLDLR, on neuronal membrane regulates phosphorylation of Disabled-1 (Dab1), activates the phosphatidylinositol (PI3K)-Akt pathway, reduces activity of glycogen synthase kinase 3β (GSK3β) and reduces phosphorylation of microtubule-associated protein tau.(126)(216)(217)(218) ApoA-I binding to ABCA1 transporters recruits Janus kinase 2 (JAK2), leading to binding of a transcription factor STAT3 (signal transducer and transcription activator 3), phosphorylation and translocation of activated STAT3 to the nucleus where it regulates transcription of numerous genes.(219)(220) A recent study has shown that apolipoprotein binding to ABCA1 activates the RhoA signaling.(221)

PLTP regulates apoE expression in astrocytes.

The ability of apolipoproteins to activate signal transduction in cells containing relevant receptors is functionally significant, although not necessarily completely separated from their lipid transfer functions since binding of apolipoproteins to specific receptors is often dependent on their lipidation status. Binding of LpE to the LDL receptor-related protein (LRP) protects neurons from apoptosis through a signal transduction pathway involving protein kinase Cδ (PKCδ) and GSK3β, while lipid-free apoE does not exert the same protection.(222) Lipid-poor apoA-I binds to ABCA1, while lipidated apoA-I binds with higher affinity to ABCG1.(223)(224) Binding of apoJ to megalin is also dependent on apoJ lipidation status, since binding affinity for megalin significantly increased after apoJ lipidation.(225)

Therefore, lipid components of apoJ-containing particles play a significant role in apoJ’s ability to induce cell survival through the PI3K-Akt-Bad-cytochrome c pathway (225)(226). These known interactions between apolipoproteins and receptors most likely represent only a subset of their physiological roles in the brain, and require further investigation.

PLTP – The “Other” Extracellular Lipid Transfer Protein in the Brain

Phospholipid transfer protein (PLTP) is not considered to be an apolipoprotein despite its association with lipoprotein particles and its role in lipid transfer. It instead belongs to a family of proteins involved in transfer of lipids and lipopolysaccharides that includes cholesteryl ester transfer protein (CETP), bactericidal permeability-increasing protein (BPI) and lipopolysaccharide-binding protein (LBP).(227) Its α-helix/β-sheet structure somewhat resembles apoD. PLTP is likely the only extracellular lipid transfer protein in the brain besides apolipoproteins, since CETP is most probably not expressed in the brain under normal circumstances. The vast majority of the brain cholesterol is in the unesterified form, and accumulation of the esterified form is associated with brain disorders,(224)(228) suggesting that CETP has no functional role in the healthy brain.

Known PLTP functions include transfer of different classes of phospholipids, unesterified cholesterol and α-tocopherol between different lipoprotein particles, and between lipoproteins and cells.(229)(230)(231)(232)(233) PLTP is critical for maintenance of the tissue levels of α-tocopherol including the brain.(232)(234) An interesting case study reported an association between a specific mutation in PLTP gene and ataxia due to low levels of α-tocopherol,(235) suggesting that the role of PLTP in regulation of the brain anti-oxidative potential needs to be further explored. PLTP plays a significant role in lipid efflux through its interaction with ABCA1 (231) and its binding to ABCA1 activates JAK2 in a manner similar to apoA-I.(219)(220)

PLTP is nearly ubiquitously expressed and is highly expressed in the brain. (236) Brain PLTP is differentially expressed by various brain regions and it is secreted by all types of the brain cells (idem). PLTP levels are altered in neurodegenerative, demyelinating and inflammatory diseases, and it is down-regulated in Down syndrome.(236)(237)(238) PLTP activity in CSF may be a good marker of neuronal cell death. It likely has predictive value in multiple sclerosis and probably in other inflammatory diseases of the brain, since PLTP activity levels are significantly decreased in active inflammatory brain diseases and recover in the remission phase.(238)

A large proportion of the brain PLTP is associated with the apoE-containing lipoproteins but a significant proportion is apoE-free (idem). PLTP regulates apoE expression in astrocytes and there is a high correlation between these two proteins in CSF (idem). PLTP also reduces phosphorylation of tau through binding to insulin receptor and insulin-like growth factor 1 receptor on neuronal membrane, activating the PI3K-Akt pathway and increasing levels of the inactive form of GSK3β.(239) Although it is currently considered a secreted protein, we have shown that it re-enters the cell and translocates to the nucleus.(240)

Conclusions

Although our knowledge of lipid transfer mechanisms within the brain and the roles of lipid transfer proteins in the brain in health and disease significantly increased in the last two decades, it is still limited. Lipoprotein and apolipoprotein functions in the brain encompass more than lipid transfer, and include regulation of cell signaling pathways and other intracellular functions.

Published studies show a strong relationship between the brain lipid and lipoprotein metabolism and neurodegenerative, demyelinating, inflammatory and psychiatric diseases. These findings indicate that the brain lipid and lipoprotein metabolism plays a major role in the development of brain diseases. Given the exceptional importance of lipids in the brain structure and function, more research is needed to understand physiological and pathophysiological relevance of the apolipoprotein- and lipoprotein-dependent functions in human brain.

In summary:
Lipoprotein metabolism is important for the brain structure and function; Alterations in the brain lipid and lipoprotein metabolism could cause and likely contribute to development of numerous brain disorders; Better understanding of lipoprotein metabolism in the brain is critical for future preventive and therapeutic approaches to brain diseases.

Acknowledgments

The author thanks Dr. John J. Albers for critical reading of the manuscript.

This article is dedicated to the memory of Dr. John “Jack” F. Oram, a brilliant scientist, an inspiring colleague and a wonderful friend, who recently lost a long and courageous battle with lung cancer. Jack dedicated his life to science, and – among many noteworthy achievements – his work and his stubborn belief that cellular lipid efflux is a receptor-mediated process led to the discovery of ATP-binding cassette A1 (ABCA1) and its role in lipid efflux and cell signaling.


Footnotes

1Li WH, Chan LC. Structure and evolution of apolipoprotein genes, in Lipoproteins in health and disease (Betteridge DJ, Illingworth DR, Shepherd J editors); Oxford University Press, New York 1999; Chapter 9, p.163.
2Olivecrona T, Olivecrona G. Lipoprotein and hepatic lipase in lipoprotein metabolism, in Lipoproteins in health and disease (Betteridge DJ, Illingworth DR, Shepherd J editors); Oxford University Press, New York 1999; Chapter 12, pp.227-228.
3Do Carmo S, Levros LC, Rassart E. Modulation of apolipoprotein D expression and translocation under specific stress conditions, Biochim. Biophys Acta Mol Cell Res 2007; 1773:954–969.
4Kim WS, Elliot DA, Kockx M, Kritharides L, Rye KA, Jans DA, Garner B. Analysis of apolipoprotein E nuclear localization using green fluorescent protein and biotinylation approaches, Biochem J 2008; 409:701–709.
5Ueno M, Nakagawa T, Wu B, Onodera M, Huang CL, Kusaka T, Araki N, Sakamoto H. Transporters in the brain endothelial barrier. Curr Med Chem 2010 In Press.
6Vaisar T, Pennathur S, Green PS, Gharib SA, Hoofnagle AN, Cheung MC, Byun J, Vuletic S, Kassim S, Singh P, Chea H, Knopp RH, Brunzell J, Geary R, Chait A, Zhao XQ, Elkon K, Marcovina S, Ridker P, Oram JF, Heinecke JW. Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of HDL. J Clin Invest 2007; 117:746-756.
7Davidson WS, Silva RA, Chantepie S, Lagor WR, Chapman MJ, Kontush A. Proteomic analysis of defined HDL subpopulations reveals particle-specific protein clusters: relevance to antioxidative function. Arerioscler Thromb Vasc Biol 2009; 29:870-876.
8Davidsson P, Huithe J, Fagerberg B, Carnejo G. Proteomics of apolipoproteins and associated proteins from plasma high-density lipoproteins. Arterioscler Thromb Vasc Biol 2010; 30:156-163.
9Packard CJ, Shepherd J. Physiology of the lipoprotein transport system: an overview of lipoprotein metabolism, in Lipoproteins in health and disease (Betteridge DJ, Illingworth DR, Shepherd J editors); Oxford University Press, New York 1999; Chapter 2, pp.17-31.
10Lund-Katz S, Phillips MC. High density lipoprotein structure-function and role in reverse cholesterol transport. Subcell Biochem 2010; 51:183-227.
11Roheim PS, Carey M, Forte T, Vega GL. Apolipoproteins in human cerebrospinal fluid. Proc Natl Acad Sci USA 1979; 76:4646-4649.
12Pitas RE, Boyles JK, Lee SH, Hui D, Weisgraber KH. Lipoproteins and their receptors in the central nervous system. Characterization of the lipoproteins in cerebrospinal fluid and identification of apolipoprotein B, E(LDL) receptors in the brain. J Biol Chem 1987b; 262:14352-14360.
13Boyles JK, Pitas RE, Wilson E, Mahley RW, Taylor JM. Apolipoprotein E associated with astrocytic glia of the central neurvous system and with nonmyelinating glia of the peripheral nervous system. J Clin Invest 1985; 76:1501-1513.
14Pitas RE, Boyles JK, Lee SH, Foss D, Mahley RW. Astrocytes synthesize apolipoprotein E and metabolize apolipoprotein E-containing lipoproteins. Biochim Biophys Acta 1987a; 917:148-161.
15Linton MF, Gish R, Hubi ST, Butler E, Esquivel C, Bry WI, Boyles JK, Wardell MR, Young SG. Phenotypes of apolipoprotein B and apolipoprotein E after liver transplantation. J Clin Invest 1991; 88:270-281.
16Drayna D, Fielding C, McLean J, Baer B, Castro G, Chen E, Comstock L, Henzel W, Kohr W, Rhee L, Wion K, Lawn R. Cloning and expression of human apolipoprotein D cDNA. J Biol Chem 1986; 261:16535-16539.
17De Silva HV, Harmony JA, Stuart WD, Gil CM, Robbins J. Apolipoprotein J: structure and tissue distribution. Biochemistry 1990; 29:5380-5389.
18McGhee KA, Morris DW, Schwaiger S, Nangle JM, Donohoe G, Clarke S, Meagher D, Quinn J, Scully P, Waddington JL, Gill M, Corvin A. Investigation of the apolipoprotein-L (APOL) gene family and schizophrenia using a novel DNA pooling strategy for public database SNPs. Schizophr Res 2005; 76:231-238.
19Petit-Turcotte C, Stohl SM, Beffert U, Cohn JS, Aumont N, Tremblay M, Dea D, Yang L, Poirier J, Shachter NS. Apolipoprotein C-I expression in the brain in Alzheimer’s disease. Neurobiol Dis 2001; 8:953-963.
20Abildayeva K, Berbée JF, Blokland A, Jansen PJ, Hoek FJ, Meijer O, Lütjohann D, Gautier T, Pillot T, De Vente J, Havekes LM, Ramaekers FC, Kuipers F, Rensen PC, Mulder M. Human apolipoprotein C-I expression in mice impairs learning and memory functions. J Lipid Res 2008; 49:856-869.
21Zhang J, Sokal I, Peskind ER, Quinn JF, Jankovic J, Kenney C, Chung KA, Millard SP, Nutt JG, Montine TJ. CSF multianalyte profile distinguishes Alzheimer and Parkinson diseases. Am J Clin Pathol 2008: 129:526-529.
22Dietschy JM, Turley SD. Cholesterol metabolism in the brain. Curr Opin Lipidol 2001; 12:105-112.
23Dietschy JM. Central nervous system: cholesterol turnover, brain development and neurodegeneration. Biol Chem 2009; 390:287-293.
24Björkhem I, Meaney S. Brain cholesterol: long secret life behind a barrier. Arterioscler Thromb Vasc Biol 2004; 24:806-815.
25Dietschy 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.
26Heverin M, Meaney S, Lütjohann D, Diczfalusy U, Wahren J, Björkhem I. Crossing the barrier: net flux of 27-hydroxycholesterol into the human brain. J Lipid Res 2005; 46:1047-1052.
27Björkhem I. Crossing the barrier: oxysterols as cholesterol transporters and metabolic modulators in the brain. J Intern Med 2006; 260:493-508.
28Björkhem I, Heverin M, Leoni V, Meany S, Diczfalusy U. Oxysterols and Alzheimer’s disease. Acta Neurol Scand Suppl 2006; 185:43-49.
29Björkhem I, Cedazo-Minguez A, Leoni V, Meaney S. Oxysterols and neurodegenerative diseases. Mol Aspects Med 2009; 30:171-179.
30Kennedy Keller R, Small M, Fliesler SJ. Enzyme blockade: a nonradioactive method to determine the absolute rate of cholesterol synthesis in the brain. J Lipid Res 2004; 45:1952-1957.
31Björkhem I, Lütjohann D, Diczfalusy U, Stahle L, Ahlburg G, Wahren J. Cholesterol homeostasis in human brain: turnover of 24S-hydroxycholesterol and evidence for a cerebral origin of most of this oxysterol in the circulation. J Lipid Res 1998; 44:1780-1789.
32Björkhem I, Andersson U, Ellis E, Alvellius G, Ellegard L, Diczfalusy U, Sjovall J, Einarsson C. From brain to bile: evidence that conjugation and omega-hydroxylation are important for elimination of 24S-hydroxycholesterol (cerebrosterol) in humans. J Biol Chem 2001; 276:37004-37010.
33Vance JE, Hayashi H. Formation and function of apolipoprotein E-containing lipoproteins in the nervous system. Biochim Biophys Acta 2010; In Press; doi: 10.1016/jbbalip.2010.02.007.
34Koudinov AR, Koudinova NV. Essential role for cholesterol in synaptic plasticity and neuronal degeneration. FASEB J 2001; 15:1858-1860.
35Koudinov AR, Koudinova NV. Cholesterol, synaptic function and Alzheimer’s disease. Pharmacopsychiatry 2003; 36:S107-S112.
36Pfrieger FW. Outsourcing in the brain: do neurons depend on cholesterol delivery by astrocytes? Bioassays 2003; 25:72-78.
37Pfrieger FW. Role of cholesterol in synapse formation and function. Biochim Biophys Acta 2003; 1610:271-280.
38Brooksbank BW, Martinez M. Lipid abnormalities in the brain in adult Down’s syndrome and Alzheimer’s disease. Mol Chem Neuropathol 1989; 11:157-185.
39Adibhatla RM, Hatcher JF. Altered lipid metabolism in brain injury and disorders. Subcell Biochem 2008; 48:nihpa41041.
40Xu Q, Li Y, Cyras C, Sanan DA, Cordell B. Isolation and characterization of apolipoproteins from murine microglia. Identification of a low density lipoprotein-like apolipoprotein J-rich but E-poor spherical particle. J Biol Chem 2000; 275:31770-31777.
41Borghini I, Barja F, Pometta D, James RW. Characterization of subpopulations of lipoprotein particles isolated from human cerebrospinal fluid. Biochim Biophys Acta 1995; 1255:192-200.
42Rebeck GW, Alonzo NC, Berezovska O, Harr SD, Knowles RB, Growdon JH, Hyman BT, Mendez AJ. Structure and functions of human cerebrospinal fluid lipoproteins from individuals of different APOE genotypes. Exp Neurol 1998; 149:175-182.
43Fagan AM, Holtzman DM, Munson G, Mathur T, Schneider D, Chang LK, Getz GS, Reardon CA, Lunkens J, Shah JA, LaDu MJ. Unique lipoproteins secreted by primary astrocytes from wild type, apoE(-/-) and human apoE transgenic mice. J Biol Chem 1999; 274:30001-30007.
44Yamauchi K, Tozuka M, Hidaka H, Hidaka E, Kondo Y, Katsuyama T. Characterization of apolipoprotein E-containing lipoproteins in cerebrospinal fluid: effect of phenotype on the distribution of apolipoprotein E. Clin Chem 1999; 45:1431-1438.
45Demeester N, Castro G, Desrumaux C, De Geitere C, Fruchart JC, Santens P, Mulleners E, Engelborghs S, De Deyn PP, Vandekerckhove J, Rosseneu M, Labeur C. Characterization and functional studies of lipoproteins, lipid transfer proteins, and lecithin-cholesterol acyltransferase in CSF of normal individuals and patients with Alzheimer’s disease. J Lipid Res 2000; 41:963-974.
46DeMattos RB, Brendza RP, Heuser JE, Kierson M, Cirrito JR. Fryer J, Sullivan PM,, Fagan AM,,, Han X, Holtzman DM. Purification and characterization of astrocyte-secreted apolipoprotein E and J-containing lipoproteins from wild-type and human apoE transgenic mice. Neurochem Int 2001; 39:415-425.
47Koch S, Donarski N, Goetze K, Kreckel M, Stuerenburg HJ, Buhmann C, Beisiegel U. Characterization of four lipoprotein classes in human cerebrospinal fluid. J Lipid Res 2001; 42:1143-1151.
48Peng D, Song C, Reardon CA, Liao S, Getz GS. Lipoproteins produced by apoE-/- astrocytes infected with adenovirus expressing human apoE. J Neurochem 2003; 86:1391-1402.
49Morikawa M, Fryer JD,,, Sullivan PM,,, Christofer EA, Wahrle SE, DeMattos RB, O’Dell MA, Fagan AM, Lashuel HA, Walz T, Asai K, Holtzman DM. Production and characterization of astrocyte-derived human apolipoprotein isoforms from immortalized astrocytes and their interactions with amyloid beta. Neurobiol Dis 2005; 19:66-76.
50Poirier J, A Baccichet, D Dea, S Gauthier. Cholesterol synthesis and lipoprotein reuptake during synaptic remodeling in hippocampus in adult rats. Neuroscience 1993; 55:81-90.
51Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto A, Pfrieger FW. CNS synaptogenesis promoted by glia-derived cholesterol Science 2001; 294:1354-1357.
52Bales KR. Brain lipid metabolism, apolipoprotein E and the pathophysiology of Alzheimer’s disease. Neuropharmacology 2010 In Press.
53Wood SJ, Chan W, Wetzel R. Seeding of A beta fibril formation is inhibited by all three isotypes of apolipoprotein E. Biochemistry 1996; 35:12623-12628.
54Olesen OF, Dagø L. High density lipoprotein inhibits assembly of amyloid beta-peptides into fibrils. Biochem Biophys Res Commun 2000; 270:62-66.
55Koudinov AR, Berezov TT, Koudinova NV. The levels of soluble amyloid beta in different high density lipoprotein subfractions distinguish Alzheimer’s and normal aging cerebrospinal fluid: implication for brain cholesterol pathology? Neurosci Lett 2001; 314:115-118.
56Fan J, Donkin J, Wellington C. Greasing the wheels of Abeta clearance in Alzheimer’s disease: the role of lipids and apolipoprotein E. Biofactors 2009; 35:239-248.
57Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer’s disease. Neuron 2009; 63:287-303.
58Kim J, Castellano JM, Jiang H, Basak JM, Parsadanian M, Pham V, Mason SM, Paul SM, Holtzman DM. Overexpression of low-density lipoprotein receptor in the brain markedly inhibits amyloid deposition and increases extracellular A beta clearance. Neuron 2009; 64:632-644.
59Stocker A. Molecular mechanisms of vitamin E transport. Ann NY Acad Sci 2004; 1031:44-59.
60Murch O, Collin M, Hinds CJ, Thiemermann C. High-density lipoproteins in sepsis and septic shock: metabolism, actions, and therapeutic applications. Intensive Care Med 2007; 33:13-24.
61Jansen PJ, Lutjohann D, Thelen KM, von Bergmann K, van Leuven F, Ramaekers FC, Mulder M. Absence of apoE upregulates murine brain apoD and ABCA1 levels, but does not affect brain sterol levels, while human apoE3 and human apoE4 upregulate brain cholesterol precursor levels. J Alzheimers Dis 2009; 18:319-329.
62Zannis VI, Cole FS, Jackson CL, Kurnit DM, Karathanasis SK. Distribution of apolipoprotein A-I, C-II, C-II, and E mRNA in fetal human tissues. Time-dependent induction of apolipoprotein E mRNA by cultures of human monocyte-macrophages. Biochemistry 1985; 24:4450-4455.
63Beffert U, Danik M, Krzywkowski P, Ramassamy C, Berrada F, Poirier J. The neurobiology of apolipoproteins and their receptors in the CNS and Alzheimer's disease. Brain Res Brain Res Rev 1998; 27:119-42.
64Wahrle SE, Jiang H, Parsadanian M, Legleiter J, Han X, Fryer JD, Kowalewski T, Holtzman DM. ABCA1 is required for normal central nervous system apoE levels and for lipidation of astrocyte-secreted apoE. J Biol Chem 2004; 279:40987-40993.
65Diedrich JF, Minnigan H, Carp RI, Whitaker JN, Race R, Frey W 2nd, Haase AT. Neuropathological changes in scrapie and Alzheimer’s disease are associated with increased expression of apolipoprotein E and cathepsin D in astrocytes. J Virol 1991; 65:4759-4768.
66Poirier J, Hess M, May PC, Finch CE. Astrocytic apolipoprotein E mRNA and GFAP mRNA in hippocampus after entorhinal cortex lesioning. Brain Res Mol Brain Res 1991; 11:97-106.
67Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y. Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci 2006; 26:4985-4994.
68Xu Q, Walker D, Bernardo A, Brodbeck J, Balestra ME, Huang Y. Intron-3 retention/splicing controls neuronal expression of apolipoprotein E in the CNS. J Neurosci 2008 28:1452-1459.
69Harris FM, Tesseur I, Brecht WJ, Xu Q, Mullendorff K, Chang S, Wyss-Coray T, Mahley RW, Huang Y. Astroglial regulation of apolipoprotein E expression in neuronal cells. Implications for Alzheimer’s disease. J Biol Chem 2004; 279:3862-3868.
70Dupont-Wallois L, Soulie C, Seargeant N, Wavrant-de Wrieze N, Chartier-Harlin MC, Delacourte A, Caillet-Boudin ML. ApoE synthesis in human neuroblastoma cells. Neurobiol Dis 1997; 4:356-364.
71Elliot DA, Kim WS, Jans DA, Garner B. Apoptosis induces neuronal apolipoprotein E synthesis and localization in apoptotic bodies. Neurosci Lett 2007; 416:206-210.
72Dekroon RM, Armati PJ. Endocytosis of apoE-EGFP by primary human brain cultures. Cell Biol Int 2002; 26:761-770.
73Rellin L, Heeren J, Beisiegel U. Recycling of apolipoprotein E is not associated with cholesterol efflux in neuronal cells. Biochim Biophys Acta 2008; 1781:232-238.
74De Chaves EP, Narayanaswami V. Apolipoprotein E and cholesterol in aging and disease in the brain. Future Lipidol 2008; 3:505-530.
75Rogers JT, Weeber EJ. Reelin and apoE actions on signal transduction, synaptic function and memory formation. Neuron Glia Biol 2009; 13:1-12.
76Boyles JK, Zoellner CD, Anderson LJ, Kosik LM, Pitas RE, Weisgraber KH, Hui DY, Mahley RW. A role for apolipoprotein E, apolipoprotein A-I, and low density lipoprotein receptors in cholesterol transport during regeneration and remyelination of the rat sciatic nerve. J Clin Invest 1989; 83:1015-1031.
77Rothe T, Müller HW. Uptake of endoneurial lipoprotein into Schwann cells and sensory neurons is mediated by low density lipoprotein receptors and stimulated after axonal injury. J Neurochem 1991; 57:2016-2025.
78Vance JE, Karten B, Hayashi H. Lipid dynamics in neurons. Biochem Soc Trans 2006; 34:399-403.
79Wang H, Durham L, Dawson H, Song P, Warner DS, Sullivan PM, Vitek MP, Laskowitz DT. An apolipoprotein E-based therapeutic improves outcome and reduces Alzheimer’s disease pathology following close head injury: evidence of pharmacogenomic interaction. Neuroscience 2007; 144:1324-1333.
80Eto M, Watanabe K, Ishii K. Reciprocal effects of apolipoprotein E alleles (epsilon 2 and epsilon on plasma lipid levels in normolipidemic subjects. Clin Genet 1986; 29:477–484.
81Emi M, Wu LL, Robertson MA, Myers RL, Hegele RA, Williams RR, Whit R, Lalouel JM. Genotyping and sequence analysis of apolipoprotein E isoforms. Genomics 1988; 3:373-379.
82Mahley RW, Rall SC Jr. Apolipoprotein E: far more than a lipid transport protein. Annu Rev Genomics Hum Genet 2000; 1:507-537.
83Strittmatter WJ, Bova Hill C. Molecular biology of apolipoprotein E. Curr Opin Lipidol 2002; 13:119-123.
84Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science 1988; 240:622-630.
85Huang Y, Weisgraber KH, Mucke L, Mahley RW. Apolipoprotein E: diversity of cellular origins, structural and biophysical properties, and effects in Alzheimer’s disease. J Mol Neurosci 2004; 23:189-204.
86Zhong N, Weisgraber KH. Understanding the association of apolipoprotein E4 with Alzheimer disease: clues from its structure. J Biol Chem 2009; 284:6027-6031.
87Mahley RW, Innerarity TL, Rall SC Jr, Weiisgraber KH, Taylor JM. Apolipoprotein E: genetic variants provide insights into tis structure and function. Curr Opin Lipidology 1990; 1:87-85.
88Mahley RW, Weisgraber KH, Huang Y. Apolipoprotein E: structure determines function, from atherosclerosis to Alzheimer’s disease to AIDS. J Lipid Res 2009; 50:S183-S188.
89Minagawa H, Gong JS, Jung CG, Watanabe A, Lund-Katz S, Phillips MC, Saito H, Michikawa M. Mechanism underlying apolipoprotein E (apoE) isoform-dependent lipid efflux from neural cells in culture. J Neurosci Res 2009; 87:2498-2508.
90Du J, Chang J, Guo S, Zhang Q, Wang Z. ApoE4 reduces expression of Abeta degrading enzyme IDE by activating the NMDA receptor in hippocampal neurons. Neurosci Lett 2009; 464:140-145.
91Korwek KM, Trotter JH, LaDu MJ, Sullivan PM, Weeber EJ. ApoE isoform-dependent changes in hippocampal synaptic function. Mol Neurodegener 2009; 27:4-21.
92Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 1993; 261: 921-23.
93Bedlack RS, Strittmatter WJ, Morgenlander JC. Apolipoprotein E and neuromuscular disease: a critical review of the literature. Arch Neurol 2000; 57:1561-1565.
94O'Donnell HC, Rosand J, Knudsen KA, Furie KL, Segal AZ, Chiu RI, Ikeda D, Greenberg SM. Apolipoprotein E genotype and the risk of recurrent lobar intracerebral hemorrhage. N Engl J Med 2000; 342:240–245.
95Fazekas F, Strasser-Fuchs S, Kollegger H, Berger T, Kristoferitsch W, Schmidt H, Enzinger C, Schiefermeier M, Schwarz C, Kornek B, Reindl M, Huber K, Grass R, Wimmer G, Vass K, Pfeiffer KH, Hartung HP, Schmidt R. Apolipoprotein E epsilon 4 is associated with rapid progression of multiple sclerosis. Neurology 2001; 57:853–857
96Enzinger C, Ropele S, Smith S, Strasser-Fuchs S, Poltrum B, Schmidt H, Matthews PM, Fazekas F. Accelerated evolution of brain atrophy and "black holes" in MS patients with APOE-epsilon 4. Ann Neurol 2004; 55:563–569.
97Pankratz N, Byder L, Halter C, Rudolph A, Shults CW, Conneally PM, Foroud T, Nichols WC. Presence of an APOE4 allele results in significantly earlier onset of Parkinson's disease and a higher risk with dementia. Mov Disord 2006; 21:45–49.
98Blázquez L, Otaegui D, Sáenz A, Paisán-Ruiz C, Emparanza JI, Ruiz-Martinez J, Moreno F, Marti Massó JF, López de Munain A. Apolipoprotein E epsilon4 allele in familial and sporadic Parkinson’s disease. Neurosci Lett 2006; 406:235-239.
99Raber J. Role of apolipoprotein E in anxiety. Neural Past 2007; 2007:91236.
100Hui DY, Basford JE. Distinct signaling mechanisms for apoE inhibition of cell migration and proliferation. Neurobiol Aging 2005; 26:317-333.
101Fukumoto H, Ingelsson M, Garevik N, Wahlund LO, Nukina N, Yaguchi Y, Shibata M, Hyman BT, Rebeck GW, Irizarry MC. APOE epsilon 3/epsilon 4 heterozygotes have an elevated proportion of apolipoprotein E4 in cerebrospinal fluid relative to plasma, independent of Alzheimer’s disease diagnosis. Exp Neurol 2003; 183:249-253.
102Riddell DR, Zhou H, Atchison K, Warwick HK, Atkinson PJ, Jefferson J, Xu L, Aschmies S, Kirksey Y, Hu Y, Wagner E, Parratt A, Xu J, Li Z, Zaleska MM, Jacobsen JS, Pangalos MN, Reinhart PH. Impact of apolipoprotein E (apoE) polymorphism on brain apoE levels. J Neurosci 2008; 28:11445-11453.
103Higgins GA, Large CH, Rupniak HT, Barnes JC. Apolipoprotein E and Alzheimer’s disease: a review of recent studies. Pharmacol Biochem Behav 1997; 56:675-685.
104Corder EH, Ghebremedhin E, Taylor MG, Thal DR, Ohm TG, Braak H. The biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphism. Ann N Y Acad Sci 2004; 1019:24–28.
105Artiga MJ, Bullido MJ, Frank A, Sastre I, Recuero M, Garcia MA, Lendon CL, Han SW, Morris JC, Vazquez J, Goate A, Valdivieso F. Risk of Alzheimer’s disease correlates with transcriptional activity of the APOE gene. Hum Mol Genet 1998; 7:1887-1892.
106Baum L, Chen L, Ng HK, Pang CP. Apolipoprotein E isoforms in Alzheimer’s disease pathology and etiology. Microsc Res Tech 2000; 50:278-281.
107Wisniewski T, Frangione B. Apolipoprotein E: a pathological chaperone protein in patients with cerebral and systemic amyloid. Neurosci Lett 1992; 135:235-238.
108Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, Roses AD. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA 1993; 90: 1977-1981.
109Nathan BP, Bellosta S, Sanan DA, Weisgraber KH, Mahley RW, Pitas RE. Differential effects of apolipoproteins E3 and E4 on neuronal growth in vitro. Science 1994; 264:850–852.
110Genis I, Gordon I, Sehayek E, Michaelson DM. Phosphorylation of tau in apolipoprotein E-deficient mice. Neurosci Lett 1995; 199:5-8.
111Nagy Z, Esiri MM, Jobst KA, Johnston C, Litchfield S, Sim E, Smith AD. Influence of the apolipoprotein E genotype on amyloid deposition and neurofibrillary tangle formation in Alzheimer's disease. Neuroscience 1995; 69:757-61.
112Näslund J, Thyberg J, Tjernberg LO, Wernstedt C, Karlström AR, Bogdanovic N, Gandy SE, Lannfelt L, Terenius L, Nordstedt C. Characterization of stable complexes involving apolipoprotein E and the amyloid beta peptide in Alzheimer’s disease brain. Neuron 1995; 15:219-228.
113Roses AD, Einstein G, Gilbert J, Goedert M, Han SH, Huang D, Hulette C, Masliah E, Pericak-Vance MA, Saunders AM, Schmechel DE, Strittmatter WJ, Weisgraber KH, Xi PT. Morphological, biochemical, and genetic support for an apolipoprotein E effect on microtubular metabolism. Ann N Y Acad Sci. 1996; 777:146-57.
114Arendt T, Schindler C, Brückner MK, Eschrich K, Bigl V, Zedlick D, Marcova L. Plastic neuronal remodeling is impaired in patients with Alzheimer’s disease carrying apolipoprotein epsilon 4 allele. J Neurosci 1997; 17:516-529.
115Puttfarcken PS, Manelli AM, Falduto MT, Getz GS, LaDu MJ. Effect of apolipoprotein E on neurite outgrowth and beta-amyloid-induced toxicity in develop in rat primary hippocampal cultures. J Neurochem 1997; 68:760-769.
116Chapman S, Michaelson DM. Specific neurochemical derangements of brain projecting neurons in apolipoprotein E-deficient mice. J Neurochem 1998; 70:708-714.
117Wang X, Luebbe P, Gruenstein E, Zemlan F. Apolipoprotein E (ApoE) peptide regulates tau phosphorylation via two different signaling pathways. J Neurosci Res 1998; 51:658-65.
118Pillot T, Goethals M, Najib J, Labeur C, Lins L, Chambaz J, Brasseur R, Vandekerskhove J, Fosseneu M. Beta-amyloid peptide interacts specifically with the carboxy-terminal domain of human apolipoprotein E: relevance to Alzheimer’s disease. J Neurochem 1999; 72:230-237.
119Gibb GM, Pearce J, Betts JC, Lovestone S, Hoffmann MM, Maerz W, Blackstock WP, Anderton BH. Differential effects of apolipoprotein E isoforms on phosphorylation at specific sites on tau by glycogen synthase kinase-3 beta identified by nano-electrospray mass spectrometry. FEBS Lett 2000; 485:99-103.
120Ji Y, Gong Y, Gan W, Beach T, Holtzman DM, Wisniewski T. Apolipoprotein E isoform-specific regulation of dendritic spine morphology in apolipoprotein E transgenic mice and Alzheimer's disease patients. Neuroscience 2003; 122:305–315.
121Brecht WJ, Harris FM, Chang S, Tesseur I, Yu GQ, Xu Q, Dee Fish J, Wyss-Coray T, Buttni M, Mucke L, Mahley RW, Huang Y. Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice. J Neurosci 2004; 24:2527-2534.
122DeMattos RB, Cirrito JR, Parsadanian M, May PC, O’Dell MA, Taylor JW, Harmony JA, Aronow BJ, Bales KR, Paul SM, Holtzman DM. ApoE and clusterin cooperatively suppress Abeta levels and deposition: evidence that apoE regulates extracellular Abeta metabolism in vivo. Neuron 2004; 41:193-202.
123Irizarry MC, Deng A, Leo A, Berezovska O, Von Arnim CA, Martin-Rehrmann M, Manelli A, LaDu MJ, Hyman BT, Rebeck GW. Apolipoprotein E modulates gamma-secretase cleavage of the amyloid precursor protein. J Neurochem 2004; 90:1132-1143.
124Champagne D, Rochford J, Poirier J. Effect of apolipoprotein E deficiency on reactive sprouting in the dentate gyrus of the hippocampus following entorhinal cortex lesion: role of the astroglial response. Exp Neurol 2005; 194:31-42.
125Hoe HS, Freeman J, Rebeck GW. Apolipoprotein E decreases tau kinases and phosphor-tau levels in primary neurons. Mol Neurodegener 2006; 1:18.
126Ohkubo N, Lee YD, Morishima A, Terashima T, Kikkawa S, Tohyama M, Sakanaka M, Tanaka J, Maeda N, Vitek MP, Mitsuda N. Apolipoprotein E and Reelin ligands modulate tau phosphorylation through an apolipoprotein E receptor/disabled-1/glycogen synthase kinase-3beta cascade. FASEB J 2003; 17:295-297.
127Lai MK, Tsang SW, Garcia-Alloza M, Minger SL, Nicoll JA, Esiri MM, Wong PT, Chen CP, Ramirez MJ, Francis PT. Selective effects of the APOE epsilon4 allele on presynaptic cholinergic markers in the neocortex of Alzheimer's disease. Neurobiol Dis 2006; 22:555–561.
128Vatassery GT, Lam C, Smith WE, Quach HT. Apolipoprotein E exerts selective and differential control over vitamin E concentrations in different areas of mammalian brain. J Neurosci Res 2006; 84:1335-1342.
129Bell RD, Sagare AP, Friedman AE, Bedi GS, Holtzman DM, Deane R, Zlokovic BV. Transport pathways for clearance of human Alzheimer’s amyloid beta-peptide and apolipoprotein E and J in the mouse central nervous system. J Cereb Blood flow Metab 2007; 27:909-918.
130Jiang Q, Lee CY, Mandrekar S, Wilkinson B, Cramer P, Zelcer N, Mann K, Lamb B, Willson TM, Collins JL, Richardson JC, Smith JD, Comery TA, Riddell D, Holtzman DM, Tontonoz P, Landreth GE. ApoE promotes the proteolytic degradation of Abeta. Neuron 2008; 58:681-693.
131Teasdale GM, Nicoll JA, Murray G, Fiddes M. Association of apolipoprotein E polymorphism with outcome after head injury. Lancet 1997; 350:1069-1071.
132Horsburgh K, Graham DI, Stewart J, Nicoll JA. Influence of apolipoprotein E genotype on neuronal damage and apoE immunoreactivity in human hippocampus following global ischemia. J Neuropathol Exp Neurol 1999; 58:227-234.
133He J, Farias S, Martinez O, Reed B, Mungas D, Decarli C. Differences in brain volume, hippocampal volume, cerebrovascular risk factors, and apolipoprotein E4 among mild cognitive impairment subtypes. Arch Neurol 2009; 66:1393-1399.
134Lanterna LA, Biroli F. Significance of apolipoprotein E in subarachnoid hemorrhage: neuronal injury, repair and therapeutic perspectives – a review. J Stroke Cerebrovasc Dis 2009; 18:116-123.
135Ott BR, Cohen RA, Gongvatana A, Okonkwo OC, Johanson CE, Stopa EG, Donahue JE, Silverberg GD, The Alzheimer's Disease Neuroimaging Initiative. Brain ventricular volume and cerebrospinal fluid biomarkers of Alzheimer’s disease. J Alzheimers Dis 2010 In Press.
136Reinvang I, Winjevoll IL, Rootwelt H, Espeseth T. Working memory deficits in healthy APOE epsilon 4 carriers. Neuropsychologia 2010; 48:566-573.
137Thambisetty M, Beason-Held L, An Y, Kraut MA, Resnick SM. APOE epsilon4 genotype and longitudinal changes in cerebral blood flow in normal aging. Arch Neurol 2010; 67:93-98.
138Vemuri P, Wiste HJ, Weigand SD, Knopman DS, Shaw LM, Trojanowski JQ, Aisen PS, Weiner M, Petersen RC, Jack CR Jr; Alzheimer's Disease Neuroimaging Initiative.Effect of apolipoprotein E on biomarkers of amyloid load and neuronal pathology in Alzheimer disease. Ann Neurol 2010; 67:308-316.
139Chen Y, Lomnitski L, Michaelson DM, Shohami E. Motor and cognitive deficits in apolipoprotein E-deficient mice after closed head injury. Neuroscience 1997; 80:1255-1262.
140Genden EM, Watanabe O, Mackinnon SE, Hunter DA, Strasberg SR. Peripheral nerve regeneration in the apolipoprotein-E-deficient mouse. J Reconstr Mircosurg 2002; 18:495-502.
141Seitz A, Kragol M, Aglow E, Showe L, Heber-Katz E. Apolipoprotein E expression after spinal cord injury in the mouse. J Neurosci Res 2003; 71: 417-426
142Iwata A, Browne KD, Chen XH, Yuguchi T, Smith DH. Traumatic brain injury induces biphasic upregulation of apoE and apoJ protein in rats. J Neurosci Res 2005; 82:103-114.
143Horsburgh K, Kelly S, McCulloch J, Higgins GA, Roses AD, Nicoll JA. Increased neuronal damage in apolipoprotein E-deficient mice following global ischaemia. Neuroreport 1999; 10:837-841.
144Masliah E, Samuel W, Veinbergs I, Mallory M, Mante M, Saitoh T. Neurodegeneration and cognitive impairment in apoE-deficient mice is ameliorated by infusion of recombinant apoE. Brain Res 1997; 751:307–314.
145Horsburgh K, McCulloch J, Nilsen M, McCracken E, Large C, Roses AD, Nicoll JA. Intraventricular infusion of apolipoprotein E ameliorates acute neuronal damage after global cerebral ischemia in mice. J Cereb Blood Flow Metab 2000; 20:458-462.
146Levi O, Lutjohann D, Devir A, von Bergmann K, Hartmann T, Michaelson DM. Regulation of hippocampal cholesterol metabolism by apoE and environmental stimulation. J Neurochem 2005; 95:987-997.
147Heininger K. A unifying hypothesis of Alzheimer’s disease. III. Risk factors. Hum Psychopharmacol 2000; 15:1-70.
148Arab L, Sabbagh MN. Are certain lifestyle habits associated with lower Alzheimer’s disease risk? J Alzheimers Dis 2010 In Press.
149Qiu C, Xu W, Fratiglioni L. Vascular and psychosocial factors in Alzheimer’s disease: epidemiological evidence toward intervention. J Alzheimers Dis 2010 In Press.
150Weiler-Güttler H, Sommerfeldt M, Papandrikopoulou A, Mischek U, Bonitz D, Frey A, Grupe M, Scheerer J, Gassen HG. Synthesis of apolipoprotein A-1 in pig brain microvascular endothelial cells. J Neurochem 1990; 54:444-450.
151Fuji H, Saito K, Hamakawa H, Maekawa N, Fujigaki S, Wada H, Shimokawa K, Seishima M, Takami T. Immunohistochemical localization and mRNA expression of apolipoprotein A-I in rat spinal cord. J Atheroscler Thromb 2002; 9:93-98.
154Karasinska JM, Rinninger F, Lutjohann D, Ruddle P, Franciosi S, Kruit JK, Singaraja RR, Hirsch-Reinshagen V, Fan J, Brunham LR, Bissada N, Ramakrishnan R, Wellington CL, Parks JS, Hayden MR. Specific loss of brain ABCA1 increases brain cholesterol uptake and influences neuronal structure and function. J Neurosci 2009; 29:3579-3589.
155Krimbou L, Denis M, Haidar B, Carrier M, Marcil M, Genest J Jr. Molecular interactions between apoE and ABCA1: impact on apoE lipidation. J Lipid Res 2004; 45:839-848.
156Vedhachalam C, Narayanaswami V, Neto N, Forte TM, Phillips MC, Lund-Katz S, Bielicki JK. The C-terminal lipid-binding domain of apolipoprotein E is a highly efficient mediator of ABCA1-dependent cholesterol efflux that promotes the assembly of high-density lipoproteins. Biochemistry 2007; 46:2583-2593.
157Wang ES, Sun Y, Guo JG, Gao X, Hu JW, Zhou L, Hu J, Jiang CC. Tetranectin and apolipoprotein A-I in cerebrospinal fluid as potential biomarkers for Parkinson’s disease. Acta Neurol Scand 2010 In Press.
158Huang JT, Wang L, Prabakaran S, Wengenroth M, Lockstone HE, Koethe D, Gerth CW, Gross S, Schreiber D, Lilley K, Wayland M, Oxley D, Leweke FM, Bahn S. Independent protein-profiling studies show a decrease in apolipoprotein A-I levels in schizophrenia CSF, brain and peripheral tissues. Mol Psychiatry 2008; 13:1118-1128.
159Handattu SP, Garber DW, Monroe CE, van Groen T, Kadish I, Nayyar G, Cao D, Palgunachari MN, Li L, Anantharamaiah GM. Oral apolipoprotein A-I mimetic peptide improves cognitive function and reduces amyloid burden in a mouse model of Alzheimer’s disease. Neurobiol Dis 2009; 34:525-534.
160Williamson R, Lee D, Hagaman J, Maeda N. Marked reduction of high density lipoprotein cholesterol in mice genetically modified to lack apolipoprotein A-I. Proc Natl Aca Sci USA 1992; 89:7134-7138.
161Weech PK, Provost P, Tremblay NM, Carnato RN, Milne RW, Marcel YL, Rassart E. Apolipoprotein D – an atypical apolipoprotein. Prog Lipid Res 1991; 30:259-266.
162Morais Cabrai JH, Atkins GL, Sánches LM, López-Boado YS, López-Otin C, Sawyer L. Arachidonic acid binds to apolipoprotein D: implications for the protein’s function. FEBS Lett 1995; 366:53-56.
163Kim WS, Wong J, Weickert CS, Webster MJ, Bahn S, Garner B. Apolipoprotein-D expression is increased during development and maturation of the human prefrontal cortex. J Neurochem 2009; 109:1053-1066.
164Smith KM, Lawn RM, Wilcox JN. Cellular localization of apolipoprotein D and lecithin:cholesterol acyltransferase mRNA in rhesus monkey tissues by in situ hybridization. J Lipid Res 1990; 31:995-1004.
165Patel SC, ASotra K, Patel YC, McConathy WJ, Patel RC, Suresh S. Astrocytes synthesize and secrete the lipophilic ligand carrier apolipoprotein D. Neuroreport 1995; 6:653-657.
166Kalman J, McConathy W, Araoz C, Kasa P, Lacko AG. Apolipoprotein D in the aging brain and in Alzheimer’s dementia. Neurol Res 2000; 22:330-336.
167Boyles JK, Notterpek LM, Anderson LJ. Accumulation of apolipoproteins in the regenerating and remyelinating mammalian peripheral nerve. Identification of apolipoprotein D, apolipoprotein A-IV, apolipoprotein E, and apolipoprotein A-I. J Biol Chem 1990; 265:17805-17815.
168Franz G, Reindl M, Patel SC, Beer R, Unterrichter I, Berger T, Schumtzhard E, Poewe W, Kampfl A. Increased expression of apolipoprotein D following experimental traumatic brain injury. J Neurochem 1999; 73:1615-1625.
169Terrisse L, Séguin D, Bertrand P, Poirier J, Milne R, Rassart E. Modulation of apolipoprotein D and apolipoprotein E expression in rat hippocampus after entorhinal cortex lesion. Brain Res Mol Brain Res 1999; 70:26-35.
170Søiland H, Søreide K, Janssen EA, Körner H, Baak JP, Søreide JA. Emerging concepts of apolipoprotein D with possible implications for breast cancer. Cell Oncol 2007; 29:195-209.
171Ganfornina MD, Do Carmo S, Lora JM, Torres-Schumann S, Vogel M, Allhorn M, González C, Bastiani MJ, Rassart E, Sanchez D. Apolipoprotein D is involved in the mechanisms regulating protection from oxidative stress. Aging Cell 2008; 7:506-515.
172Do Carmo S, Jacomy H, Talbot PJ, Rassart E. Neuroprotective effect of apolipoprotein D against coronarvirus OC43-induced encephalitis in mice. J Neurosci 2008; 28:10330-10338.
173Rickhag M, Deierborg T, Patel S, Ruscher K, Wieloch T. Apolipoprotein D is elevated in oligodendrocytes in the peri-infarct region after experimental stroke: influence of enriched environment. J Cereb Blood Flow Metab 2008; 28:551-562.
174Sanchez D, Lopez-Arias B, Torroja L, Canal I, Wang X, Bastiani MJ, Ganfornina MD. Loss of glial lazarillo, a homolog of apolipoprotein D, reduces lifespan and stress resistance in Drosophila. Curr Biol 2006; 16:680-686.
175Walker DW, Muffat J, Rundel C, Benzer S. Overexpression of a Drosophila homolog of apolipoprotein D leads to increased stress resistance and extended lifespan. Curr Biol 2006; 16:674-679.
176Terrisse L, Poirier J, Bertrand P, Merched A, Visvikis S, Siest G, Milne R, Rassart E. Increased levels of apolipoprotein D in cerebrospinal fluid and hippocampus of Alzheimer’s patients. J Neurochem 1998; 71:1643-1650.
177Reindl M, Knipping G, Wicher I, Dilitz E, Egg R, Deisenhammer F, Berger T. Increased intrathecal production of apolipoprotein D in multiple sclerosis. J Neuroimmunol 2001; 119:327-332.
178Thomas EA, Copolov DL, Sutcliffe JG. From pharmacotherapy to pathophysiology: emerging mechanisms of apolipoprotein D in psychiatric disorders. Curr Mol Med 2003; 3:408-418.
179Glöckner F; Ohm TG. Hippocampal apolipoprotein D level depends on Braak stage and apoE genotype. Neuroscience 2003; 122:103-110.
180Desai PP, Ikonomovic MD, Abrahamson EE, Hamilton RL, Isanski BA, Hope CE, Klunk WE, DeKosky ST, Kamboh MI. Apolipoprotein D is a component of compact but not diffuse amyloid-beta plaques in Alzheimer’s disease temporal cortex. Neurobiol Dis 2005; 20:574-582.
181Bellander BM, von Holst H, Fredman P, Svensson M. Activation of the complement cascade and increase of clusterin in the brain following a cortical contusion in the adult rat. J Neurosurg 1996; 85: 468-475
182Pucci S, Mazzarelli P, Missiroli F, Regine F, Ricci F. Neuroprotection: VEGF, IL-6, and clusterin: the dark side of the moon. Porg Brain Res 2008; 173:555-573.
183May PC, Lampert-Etchells M, Johnson SA, Poirier J, Masters JN, Finch CE. Dynamics of gene expression for a hippocampal glycoprotein elevated in Alzheimer’s disease and in response to experimental lesions in rat. Neuron 1990; 5:831-839.
184Leskov KS, Klokov DY, Li J, Kinsella TJ, Boothman DA. Synthesis and functional analyses of nuclear clusterin, a cell death protein. J Biol Chem 2003; 278:11590-11600.
185Pasinetti GM, Johnson SA, Oda T, Rozovsky I, Finch CE. Clusterin (SPG-: a multifunctional glycoprotein with regional expression in astrocytes and neurons of the adult rat brain. J Comp Neurol 1994; 339:387-400.
186Saura J, Petegnief V, Wu X, Liang Y, Paul SM. Microglial apolipoprotein E and astroglial apolipoprotein J expression in vitro: opposite effects of lipopolysaccharide. J Neurochem 2003; 85:1455-1467.
187Pucci S, Bonanno E, Pichiorri F, Angeloni C, Spognoli LG. Modulation of different clusterin isoforms in human colon tumorigenesis. Oncogene 2004; 23:2298-2304.
188 Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R, Wang CY. Clusterin inhibits apoptosis by interacting with activated Bax. Nat Cell Biol 2005;7:909–915.
189Nizard P, Tetley S, Le Dréan Y, Watrin T, Le Goff P, Wilson MR, Michel D. Stress-induced retrotranslocation of clusterin/apoJ into the cytosol. Traffic 2007; 8-554-565.
190Yang CR, Leskov K, Hosley-Eberlein K, Criswell T, Pink JJ, Kinsella TJ, Boothman DA. Nuclear clusterin/XIP8, an x-ray-induced Ku70-binding protein that signals cell death. Proc Natl Acad Sci USA 2000;97:5907–5912.
191 Santilli G, Aronow BJ, Sala A. Essential requirement of apolipoprotein J (clusterin) signaling for IkappaB expression and regulation of NF-kappaB activity. J Biol Chem 2003;278:38214–38219.
192 Kang SW, Shin YJ, Shim YJ, Jeong SY, Park IS, Min BH. Clusterin interacts with SCLIP (SCG10-like protein) and promotes neurite outgrowth of PC12 cells. Exp Cell Res 2005; 309:305–315.
193 Debure L, Vayssiere JL, Rincheval V, Loison F, Le Drean Y, Michel D. Intracellular clusterin causes juxtanuclear aggregate formation and mitochondrial alteration. J Cell Sci 2003;116:3109–3121.
194Giannakopoulos P, Kovari E, French LE, Viard I, Hof PR, Bouras C. Possible neuroprotective role of clusterin in Alzheimer's disease: a quantitative immunocytochemical study. Acta Neuropathol (Berl) 1998; 95: 387-394
195Hammad SM, Ranganathan S, Loukinova E, Twai WO, Argraves WS. Interction of apolipoprotein J-amyloid beta-peptide complex with low density lipoprotein receptor-related protein-2/megalin. A mechanism to prevent pathological accumulation of amyloid beta-peptide. J Biol Chem 1997; 272:18644-18649.
196Yerbury JJ, Wilson MR. Extracellular chaperones modulate the effects of Alzheimer’s patient cerebrospinal fluid on Abeta(1- toxicity and uptake. Cell Stress Chaperones 2010; 15:115-121.
197Nuutinen T, Suuronen T, Kauppinen A, Salminen A. Clusterin: a forgotten player in Alzheimer’s disease. Brain res Rev 2009; 61:89-104.
198Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, Letenneur L, Bettens K, Berr C, Pasquier F, Fiévet N, Barberger-Gateau P, Engelborghs S, De Deyn P, Mateo I, Franck A, Helisalmi S, Porcellini E, Hanon O; European Alzheimer's Disease Initiative Investigators, de Pancorbo MM, Lendon C, Dufouil C, Jaillard C, Leveillard T, Alvarez V, Bosco P, Mancuso M, Panza F, Nacmias B, Bossù P, Piccardi P, Annoni G, Seripa D, Galimberti D, Hannequin D, Licastro F, Soininen H, Ritchie K, Blanché H, Dartigues JF, Tzourio C, Gut I, Van Broeckhoven C, Alpérovitch A, Lathrop M, Amouyel P. Genome-wide association study identifies variants of CLU and CR1 associated with Alzheimer’s disease. Nat Genet 2009; 41:1094-1099.
199Gong Y, Xi G, Wan S, Gu Y, Keep RF, Hua Y. Effects of aging on complement activation and neutrophil infiltration after intracerebral hemorrhage. Acta Neurochir Suppl 2008; 105:67-70.
200Wiggins AK, Shen PJ, Gundlach AL. Delayed, but prolonged increases in astrocytic clusterin (apoJ) mRNA expression following acute cortical spreading depression in the rat: evidence for a role of clusterin in ischemic tolerance. Brain Res Mol Brain Res 2003; 114:20-30.
201Dati G, Quattrini A, Bernasconi L, Malagati MC, Antonsson B, Nicoletti F, Alliod C, Di Marco R, Sagot Y, Vitte PA, Hiver A, Greco B, Roach A, Zaratin PF. Beneficial effects of r-h-CLU on disease severity in different animal models of peripheral neuropathy. J Neuroimmunol 2007; 190:8-17.
202Sasaki K, Doh-ura K, Wakisaka Y, Iwaki T. Clusterin/apolipoprotein J is associated with cortical Lewy bodies: immunohistochemical study in cases with alpha-synucleinopathies. Acta Neuropathol 2002; 104:225-230.
203Vanhollebeke B, Pays E. The function of apolipoproteins L. Cell Mol Life 2006; 63:1937-1944.
204Duchateau PN, Pullinger CR, Cho MH, Eng C, Kane JP. Apolipoprotein L gene family: tissue-specific expression, splicing, promoter regions, discovery of new gene. J Lipid Res 2001; 42:620-630.
205Mimmack ML, Ryan M, Baba H, Navarro-Ruiz J, Iritani S, Faull RL, McKenna PJ, Jones PB, Arai H, Starkey M, Emson PC, Bahn S. Gene expression analysis in schizophrenia: reproducible up-regulation of several members of the apolipoprotein L family located in a high-susceptibility locus for schizophrenia on chromosome 22. Proc Natl Acad Sci USA 2002; 99:4680-4685.
206Page NM, Butlin DJ, Lomthaison K, Lowry PJ. The human apolipoprotein L gene cluster: identification, classification, and sites of distribution. Genomics 2001; 74:71-78.
207Duchateau PN, Pullinger CR, Orellana RE, Kunitake ST, Naya-Vigne J, O’Connor PM, Malloy MJ, Kane JP. Apolipoprotein L, a new human high density lipoprotein apolipoprotein expressed by the pancreas. Identification, cloning, characterization, and plasma distribution of apolipoprotein L. J Biol Chem 1997; 272:25576-25582.
208Duchateau PN, Movsesyan I, Yamashita S, Sakai N, Hirano KI, Schoenhaus SA, O’Connor-Kearns PM, Spencer SJ, Jaffe RB, Redberg RF, Ishida BY, Matsuzawa Y, Kane JP, Malloy MJ. Plasma apolipoprotein L concentrations correlate with plasma triglycerides and cholesterol levels in normolipidemic, hyperlipidemic, and diabetic subjects. J Lipid Res 2000; 41:1231-1236.
209Li Q, Fan P, Bai H, Liu R, Huang Y, Wang X, Wu H, Liu Y, Liu B. Distribution and effect of apoL1 genotype on plasma lipid and apolipoprotein levels in Chinese normolipidemic and endogenous hypertriglyceridemic subjects. Clin Chim Acta 2009; 401:152-155.
210Vanhollebeke B, Nielsen MJ, Watanabe Y, Truc P, Vanhamme L, Nakajima K, Moestrup SK, Pays E. Distinct roles of haptoglobin-related protein and apolipoprotein L-1 in trypanolysis by human serum. Proc Natl Acad Sci USA 2007; 104:4118-4123.
211Sutcliffe JG, Thomas EA. The neurobiology of apolipoproteins in psychiatric diseases. Mol Neurobiol 2002; 26:369-388.
212Takahashi S, Cui YH, Han YH, Fagerness JA, Galloway B, Shen YC, Kojima T, Uchiyama M, Faraone SV, Tsuang MT. Association of SNPs and haplotypes in apoL1, 2 and 4 with schizophrenia. Schizophr Res 2008; 104:153-164.
213Lehrmann E, Colantuoni C, Deep-Soboslay A, Becker KG, Lowe R, Huestis MA, Hyde TM, Kleinman JE, Freed WJ. Transcriptional changes common to human cocaine, cannabis and phencyclidine abuse. PLoS One 2006; 1:e114.
214Mahadik SP, Evans DR. Is schizophrenia a metabolic brain disorder? Membrane phospholipid dysregulation and its therapeutic implications. Psychiatr Clin North Am 2003; 26:85-102.
215Mahadik SP, Mukherjee S, Horrobin DF, Jenkins K, Correnti EE, Scheffer RE. Plasma membrane phospholipid fatty acid composition of cultured skin fibroblasts from schizophrenic patients: comparison with bipolar patients and normal subjects. Psychiatry Res 1996; 63:133-142.
216Pap M, Cooper GM. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-kinase/Akt cell survival pathway. J Biol Chem 1998; 273:19929-19932.
217Bu G. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat Rev Neurosci 2009; 10:333-344.
218Herz J. Apolipoprotein E receptors in the nervous system. Curr Opin Lipidol 2009; 190-196.
219Oram JF, Wolfbauer G, Vaughan AM, Tang C, Albers JJ. Phospholipid transfer protein interacts with and stabilizes ATP-binding cassette transporter A1 and enhances cholesterol efflux from cells. J Biol Chem 2003; 278:52379-52385.
220Oram JF, Wolfbauer G, Tang C, Davidson WS, Albers JJ. An amphipathic helical region of the N-terminal of phospholipid transfer protein is critical for ABCA1-dependent cholesterol efflux. J Biol Chem 2008; 283:11541-11549.
221Okuhira K, Fitzgerald ML, Tamehiro N, Ohoka N, Suzuki K, Sawada JI, Naito M, Nishimaki-Mogami T. Binding of PDZ-RhoGEF to ATP-binding cassette transporter A1 (ABCA induces cholesterol efflux through RhoA activation and prevention of transporter degradation. J Biol Chem 2010 In Press.
222Hayashi H, Campenot RB, Vance DE, Vance JE. Apolipoprotein E-containing lipoproteins protect neurons from apoptosis via a signaling pathway involving low-density lipoprotein receptor-related protein-1. J Neurosci 2007; 27:1983-1941.
223Vaughan AM, Oram JF. ABCA1 and ABCG1 or ABCG4 act sequentially to remove cellular cholesterol and generate cholesterol-rich HDL. J Lipid Res 2006; 47:2433-2443.
224Lorenzi I, von Eckardstein A, Radosavljevic S, Rohrer L. Lipidation of apolipoprotein A-I by ATP-binding cassette transporter (ABC) A1 generates an interaction partner for ABCG1 but not for scavenger receptor B1. Biochim Biophys Acta 2008; 178:306-313.
225Calero M, Tokuda T, Rostagno A, Kumar A, Zlokovic B, Frangione B, Ghiso J. Functional and structural properties of lipid-associated apolipoprotein J (clusterin). Biochem J 1999; 344:375-383.
226Ammar H, Closset JL. Clusterin activates survival through the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem 2008; 19:12851-12361.
227Bingle CD, Craven CJ. Meet the relatives: a family of BPI- and LBP-related proteins. Trends Immunol 2004; 25:53-55.
228Agranoff BW, Benjamin JA, Hajra AK. Lipids; in Basic neurochemistry, molecular, cellular, and medical aspects (Siegel GJ, Agranoff BW, Albers RW, Fisher SK, Uhler MD editors); Lipincott-Raven Publishers, Philadelphia – New York 1999; Chapter 3:p38.
229Kostner GM, Oettl K, Jauhiainen M, Ehnholm C, Esterbauer H, Dieplinger H. Human plasma phospholipid transfer protein accelerates exchange/transfer of alpha-tocopherol between lipoproteins and cells. Biochem J. 1995; 305:659-667.
230Rao R, Albers JJ, Wolfbauer G, Pownall HJ. Molecular and macromolecular specificity of human plasma phospholipid transfer protein. Biochemistry 1997; 36:3645-3653.
231Wolfbauer G, Albers JJ, Oram JF. Phospholipid transfer protein enhances removal of cellular cholesterol and phospholipids by high-density lipoprotein apolipoproteins. Biochimica et Biophysica Acta 1999; 1439:65-76.
232Desrumaux C, Risold PY, Schroeder H, Deckert V, Masson D, Athias A, Laplanche H, Le Guern N, Blache D, Jiang XC, Tall AR, Desor D, Lagrost L. Phospholipid transfer protein (PLTP) deficiency reduces brain vitamin E content and increases anxiety in mice. FASEB J 2005; 19:296-297.
233Albers JJ and Cheung MC. Emerging roles for phospholipid transfer protein in lipid and lipoprotein metabolism. Curr Opin Lipidol 2004; 15:255-260.
234Desrumaux C, Deckert V, Athias A, Masson D, Lizrd G, Palleau V, Gambert P, Labrost L. Plasma phospholipid transfer protein prevents vascular endothelium dysfunction by delivering alpha-tocopherol to endothelial cells. FASEB J 1999; 13:883-892.
235Kono S, Otsuji A, Hattori H, Shirikawa K, Suzuki H, Miyajima H. Ataxia with vitamin E deficiency with a mutation in a phospholipid transfer protein gene. J Neurol 2009; 256:1180-1181.
236Vuletic S, Jin LW, Marcovina SM, Peskind ER, Moller T, Albers JJ. Widespread distribution of PLTP in human CNS: evidence for PLTP synthesis by glia and neurons, and increased levels in Alzheimer's disease. J Lipid Res 2003; 44:1113-23.
237Krapfenbauer K, Yoo BC, Kim SH, Cairns N, Lubec G. Differential display reveals downregulation of the phospholipid transfer protein (PLTP) at the mRNA level in brains of patients with Down syndrome. Life Sci 2001; 68:2169-2179.
238Vuletic S, Peskind ER, Marcovina SM, Quinn JF, Cheung MC, Kennedy H, Kaye JA, Jin LW, Albers JJ. Reduced CSF PLTP activity in Alzheimer's disease and other neurologic diseases; PLTP induces ApoE secretion in primary human astrocytes in vitro. J Neurosci Res 2005; 80:406-13.
239Dong W, Albers JJ, Vuletic S. 2009. Phospholipid transfer protein reduces phosphorylation of tau in human neuronal cells. J Neurosci Res 2009; 87:3176-3185.
240Vuletic S, Dong W, Wolfbauer G, Day JR, Albers JJ. PLTP is present in the nucleus, and its nuclear export is CRM1-dependent. Biochim Biophys Acta 2009; 1793:584-591.