TRANSCRIPTIONAL EFFECTS OF APOE4: RELEVANCE TO ALZHEIMER'S DISEASE
 
   

Transcriptional Effects of ApoE4:
Relevance to Alzheimer's Disease

This section is compiled by Frank M. Painter, D.C.
Send all comments or additions to:
    Frankp@chiro.org
 
   

FROM: Molecular Neurobiology 2018 (Jun); 55 (6): 5243–5254 ~ FULL TEXT

Veena Theendakara & Clare A. Peters-Libeu & Dale E. Bredesen

The Buck Institute for Research on Aging,
8001 Redwood Blvd,
Novato, CA, 94945, USA.

Easton Laboratories for Neurodegenerative Disease Research,
UCLA, Los Angeles, CA, 90025, USA.


The major genetic risk factor for sporadic Alzheimer's disease (AD) is the lipid binding and transporting carrier protein apolipoprotein E, epsilon 4 allele (ApoE4). One of the unsolved mysteries of AD is how the presence of ApoE4 elicits this age-associated, currently incurable neurodegenerative disease. Recently, we showed that ApoE4 acts as a transcription factor and binds to the promoters of genes involved in a range of processes linked to aging and AD disease pathogenesis.

These findings point to novel therapeutic strategies for AD and aging, resulting in an extension of human healthspan, the disease-free and functional period of life. Here, we review the effects and implications of the putative transcriptional role of ApoE4 and propose a model of Alzheimer's disease that focuses on the transcriptional nature of ApoE4 and its downstream effects, with the aim that this knowledge will help to define the role ApoE4 plays as a risk factor for AD, aging, and other processes such as inflammation and cardiovascular disease.

Keywords:   Alzheimer’s disease; ApoE4; Apolipoprotein E; Neurodegeneration; Transcription



From the FULL TEXT Article:

Introduction

Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder that affects over five million Americans, and at present, there are no pharmacological treatments known to cure or halt its progression. The biggest risk factor for AD is aging itself, but there are other determinants. One is a genetic variant termed APOE ε4, and interestingly, this variant is also linked to life span. However, the most common apolipoprotein E allele type is the APOE ε3, with a frequency range of 50– 90%, while APOE ε4 and APOE ε2 allele frequencies range from 5 to 35 and 1 to 5%, respectively. The risk for AD is associated with the APOE allele (ε4>ε3>ε2), and inheritance of one or two copies of APOE ε4 dramatically increases the late-onset AD risk approximately 3- or 12-fold, respectively [1, 2]. The protein structures of ApoE2, E3, and E4 are known. What has remained a mystery, however, is how subtle changes in the protein structure that distinguish the three isoforms lead to such profound changes in the risk and biology of AD.

Several genes, including Sirtuin1 (SirT1), have been linked to the normal aging process. Previous research from our group and from others discovered a link between apolipoprotein E, epsilon 4 allele (ApoE4) and SirT1, finding that most of the abnormalities associated with ApoE4 and AD in a cell culture model could be prevented by increasing the SirT1 activity [3]. These findings suggest that the activity of the SirT1 protein may be protective both against normal aging and AD. More recently, we provided new insights into the mechanism by which ApoE4 binds SirT1 and confers risk for AD [4], discovering that ApoE4 protein functions as a transcription factor, binding DNA and modulating the transcription of ~ 1700 genes, approximately half of which did not bind ApoE3. Notably, several of these genes have previously been linked to aging and AD pathogenesis and include genes involved in energy metabolism, inflammation, axon guidance, neuronal survival, and cell death [4]. Thus, it is noteworthy that the APOE ε4 allele is associated with aging and other agerelated diseases and that the associations between ApoE4, aging, and AD could be related mechanistically [5, 6].



ApoE4 and Neurodegeneration

The human ApoE protein is a 299 amino acid glycoprotein and is expressed in several organs, with the highest expression in the liver followed by the brain. ApoE is produced in abundance in the brain and serves as the principal lipid transport vehicle in CSF. In both human and rat brains, ApoE messenger RNA (mRNA) is abundant in the cerebral cortex, hippocampus, cerebellum, and medulla [7]. Non-neuronal cells, mainly astrocytes and microglia, are the major cell types that express ApoE in the brain [8–11]. Neurons can also produce ApoE under stressful conditions [7, 12].

ApoE functions as a ligand in receptor-mediated endocytosis of lipoprotein particles [13]. Unlike plasma HDL that contains ApoA-1 as its major apolipoprotein, the predominant apolipoprotein of HDL in the central nervous system (CNS) is ApoE [11]. Cholesterol released from ApoE-containing lipoprotein particles is used to support synaptogenesis and the maintenance of synaptic connections [14]. By the mid-1980s, several lines of evidence had been advanced that linked ApoE4 to neurodegeneration, including its role in AD, Parkinson’s disease (PD), cardiovascular diseases (CVD), multiple sclerosis (MS), type 2 diabetes mellitus (T2DM), vascular dementia (VD), and ischemic stroke (IS) [7, 15–17].

Several studies have shown a positive association between harboring the APOE ε4 allele and increased risk for VD [18–20], and a meta-analysis report has revealed evidence of an increased VD risk in individuals with ApoE4 compared with ApoE3 [19].

Similarly, epidemiologic studies have indicated a direct association between APOE and CVD as well as its impact on cholesterol levels [21]. In a study in middle-aged men, it was estimated that 40%of the ε4 carriers had an increased risk for CVD mortality compared with individuals with the APOE ε3/3 or ε2/2 genotypes [21–25], although other studies failed to find any association with ApoE4, and one study did show an increased CVD risk associated with the E2 allele [16, 25]. Thus, the exact role of ApoE in CVD remains enigmatic and needs to be explored further.

The study results of association of ApoE genotype with MS have been inconsistent [26, 27]. APOE ε4 allele may be a risk factor for cognitive impairment since patients with MS who possessed the E4 isoform were reported to present with verbal memory deficits [28–31].

While PD and AD have some overlapping clinical and neuropathological features, most studies have failed to report any association between ApoE4 genotype and susceptibility to PD or PD-associated dementia and studies focusing on the role of ApoE in PD have largely remained inconclusive [32–35].



ApoE and Alzheimer’s Disease

AD is a progressive, age-associated loss of cognitive function. Symptoms including memory losses typically appear after age 60, with some early-onset forms of the disease linked to a specific genetic defect. The disease is characterized by the presence of senile plaques and abnormal Tau tangles often starting in the hippocampal region. No cure exists for Alzheimer’s, and the drugs currently available to treat the disease address only its symptoms and with limited effectiveness. As the disease progresses, patients become totally dependent upon caregivers and eventually require comprehensive care. Thus, Alzheimer’s disease presents a considerable problem in patient management [36, 37]. One in eight older Americans has AD. An estimated 5.5 million Americans are living with Alzheimer’s dementia, although this number does not account for those individuals that are potential victims but not yet diagnosed with the disease. Thus, any therapeutic intervention that could postpone the onset or progression of AD would dramatically reduce the number of cases over the next 50 years [36, 37].

ApoE plays a critical role in transporting cholesterol in and out of the CNS and ApoE4 is the single most important genetic risk factor associated with AD. Individuals with two copies of the ApoE4 allele have an increased risk for sporadic or familial AD, with a significantly lower age of onset compared with AD patients not carrying this allele [7, 38–40]. Despite knowing for two decades that the ApoE4 allele is somehow contributory to the Alzheimer’s disease process, the precise molecular mechanisms underlying ApoE and β- amyloid precursor protein (APP) interactions, direct or indirect, resulting in ApoE4-mediated toxicity, remain unclear.



ApoE4 Effects on APP and AD Pathophysiology

The APP has been shown to function as a molecular switch: cleavage at the β, γ, and caspase sites results in the production of four pro-AD peptides—sAPPβ, Aβ, Jcasp, and C31—that mediate neurite retraction, synaptic reorganization, caspase activation, and ultimately programmed cell death. In contrast, cleavage at the α site produces the anti-AD trophic peptide sAPPα and the inhibitor of APP γ-site cleavage, α CTF [41]. The decision between these two proteolytic pathways is governed at least in part by ligand binding [41]. It has been shown that ApoE4 binds to APP with nanomolar affinity and decreases sAPPα secretion and reduces sAPPα/Aβ and sAPPα/sAPPβ ratios [3]. Furthermore, the level of CSF sAPPα is significantly lower in AD patients possessing one or two ApoE4 alleles than in those not possessing the ApoE4 allele [42]. In addition, treatment of human neuroblastoma cells with ApoE4 results in decreased secretion of sAPPα [43] and secretion of sAPPα is differentially affected by distinct ApoE isoforms as a functional consequence of the APPApoE interaction [44].

In addition to its effects on sAPPα reduction, several clinical studies have reported that cerebral Aβ deposition is positively associated with the ApoE4 genotype in cognitively normal subjects, mild cognitive impairment cases, and symptomatic AD patients [45–47], suggesting that ApoE4 may increase the risk of AD at least in part by initiating and accelerating Aβ accumulation, aggregation, and deposition in the brain [7, 38]. Data from several human studies confirm the ApoE genotype as a strong susceptibility factor for Aβ plaque accumulation and eventual development of AD [48, 49].

However, the mechanism by which ApoE4 directly promotes seeding and aggregation of Aβ in vivo remains unknown. Additionally, studies also suggest a role for ApoE4 in preventing Aβ clearance as a major pathogenic event for AD. Soluble Aβ can be removed from the brain by various clearance pathways. Lipidated ApoE can bind to Aβ (isoform-dependent with E3 >> E4) to form a ApoE–Aβ complex that is internalized by the cells through cell surface low density lipoprotein receptor-related protein 1 (LRP1) or lowdensity lipoprotein (LDL) receptors [48, 50]. While the endocytosed ApoE is recycled, Aβ is transported to the lysosomal compartment for degradation. Studies have shown that ApoE4 is less effective in promoting receptor-mediated Aβ clearance than ApoE3 [48, 51–54].

Furthermore, in the human brain, the macrophages have a critical role inAβ1–42 clearance and are extremely efficient at degrading soluble and insoluble Aβ that is also ApoE isoform dependent with E2 > E3 > E4 [55–57]. The ApoE-dependent, macrophage-induced Aβ degradation is blocked by the LDL receptor antagonist and receptor-associated protein (RAP) suggesting the involvement of one or more LDL receptors in mediating Aβ degradation and clearance by macrophages [56, 58]. ApoE4 may also prevent Aβ clearance by directly or indirectly blocking the activity of Aβ-degrading proteases including neprilysin (NEP), insulin-degrading enzyme (IDE), matrix metalloproteinases 2 and 9, and other proteases [59–65]. Hippocampal IDE protein is reduced by approximately 50% in AD patients with the ApoE4 genotype [63]. Studies also show that ApoE facilitates Aβ binding and lysosomal trafficking in an isoform-dependent manner, with ApoE3 more efficiently enhancing Aβ trafficking and degradation than ApoE4. ApoE3 enhances both the endolytic degradation of Aβ by NEP and extracellular Aβ degradation by IDE compared to ApoE4, although the mechanism remains unclear. Some studies have indicated the direct influence of ApoE4 in the regulation of IDE levels through the NMDA receptor pathway [64–66].



Other Effects of ApoE4 on AD Pathophysiology

Most of the APP and Aβ-independent effects of ApoE4 on AD pathophysiology have come from mouse models expressing human ApoE3 or ApoE4 that lack endogenous mouse ApoE [67]. Age and kainic acid-induced presynaptic and dendritic degeneration were exacerbated in NSE-ApoE4-Tg mice compared to NSE-ApoE3-Tg mice [68–70]. Furthermore, female NSE-ApoE4-Tg mice showed greater cognitive impairment compared to female NSE-ApoE3-Tg mice. ApoE4 is more susceptible to proteolytic cleavage than ApoE3 in vivo in these transgenic mouse models [68, 71–74]. The ApoE4 fragments accumulate in neurofibrillary tangles and amyloid plaques, disrupt cytoskeletal structure, and impair mitochondrial function [7, 68, 71, 73, 74]. Increased Tau phosphorylation is also observed in the ApoE4-Tg transgenic mice and in postmortem AD brains [3, 68, 73, 75, 76]. Morphological studies of these transgenic mouse lines demonstrate that human ApoE4 but not ApoE3 elicits neuronal and behavioral deficits in the absence of Aβ accumulation.

ApoE-TR mice were also developed to express each hApoE isoform (similar to the levels of ApoE isoforms in humans) and under the control of the endogenous mouse APOE promoter [67, 77–79]. ApoE4 levels were significantly lower in plasma, CSF, and brain homogenates than ApoE2 and ApoE3 [67]. The ApoE4-TR mice displayed decreased spine density and dendrite length, reduced excitatory transmission, and long-term potentiation compared to the ApoE3- TR mice. Female ApoE4-TR mice were cognitively impaired compared to female ApoE3-TR mice [78, 80]. ApoE4 and not ApoE3 inhibits neurite outgrowth, decreases dendritic spine density, and impairs adult hippocampal neurogenesis and neuronal plasticity in the different ApoE-Tg mouse models [7, 81–84].

Despite a wealth of data pointing to ApoE4’s manifold deleterious effects leading to AD pathophysiology, the mechanistic details of how ApoE4 causes all these effects leading to cellular toxicity remain incompletely defined. In addition, it is not clear if all of the previous effects stem from a direct interaction of ApoE4 with its target molecules or from an indirect mechanism.



ApoE4 Regulates Pathways Linking Aging and Alzheimer’s Disease

In addition to being a dominant risk factor for AD, the ApoE4 isoform is also linked to normal aging. We and others have shown that ApoE4 expression results in reduced expression and activity of Sirtuin1 (SirT1) in neural cells, ApoE4 TR mice, and AD postmortem tissue that featured at least one ApoE4 allele [3, 4, 85, 86]. SirT1, which belongs to the Sirtuin family of NAD-dependent protein deacetylases, has been linked to the normal aging process and also suppresses AD-related biochemical events in cells, primary neurons, and mouse models by directly activating transcription of ADAM10 and increasing the levels of the neuroprotective sAPPα [3, 4, 85, 87, 88].

SirT1 is interesting, since small molecules like resveratrol and other STACs (synthetic SIRT1 activating compounds) have been identified that activate this protein, which in model systems leads to life span extension, or improvements in healthspan [89–91]. Most of the abnormalities associated with ApoE4 and AD could also be prevented by increasing SirT1 activity [3, 4]. In addition to its effects on APP processing and signaling, ApoE4 and not ApoE3 triggered a marked reduction in the expression of SirT1 in cultured neural cells, cerebrospinal fluid, and in the brains of patients with AD [3, 4, 85, 86].

The ApoE4-mediated reduction in SirT1 mRNA levels correlated strongly with a reduction in SirT1 protein levels [3, 4]. Because SirT1 has been implicated in neuroprotection, this effect of ApoE4 may be important from both mechanistic and therapeutic development standpoints. It has been reported that SirT1 deficiency leads to hyperacetylation of Tau and accumulation of phosphorylated tau (p-Tau) and Aβ in mouse models of AD and in AD patients [92]. These findings suggest that increasing the SirT1 levels in an ApoE4 background may be protective both against normal aging and AD.



Indirect Transcriptional Role of ApoE

Several studies have now demonstrated an indirect role of ApoE4 in gene transcription. It has been demonstrated that the ApoE4 may act like a signaling molecule by increasing the nuclear translocation of histone deacetylases (HDACs) in human neurons that in turn reduce BDNF expression, in comparison to ApoE3, which increases histone-3 acetylation and upregulates BDNF expression. ApoE4 acts via LRP-1 receptors to modulate HDAC nucleo-cytoplasmic shuttling that is regulated by PKCε [93]. The exact mechanism underlying ApoE4 binding to LRP resulting in PKC activation, HDAC translocation to nucleus, and downregulation of BDNF expression remains unclear. In another recent study using human neurons derived from ES cells, Huang et al. showed that ApoE4 robustly stimulates APP transcription and Aβ production by activating a non-canonicalMAP kinase pathway involving DLK, MKK7, and ERK1/2. This cascade finally stimulates c-Fos phosphorylation and APP gene transcription, leading to increased APP and Aβ synthesis [94]. The factors that link the ApoE4 receptor complex to DLK activation remain unknown. In addition, it is not clear how APP transcription leads specifically to Aβ production without affecting the production of sAPPα or other related peptides.

In a report on the astrocyte transcriptome using astrocytes isolated from postmortem temporal cortex samples, Simpson et al. identified 237 genes that were differentially regulated by ApoE4. The 237 genes were categorized into six functional groups and included genes associated with signaling pathways, the cytoskeleton, DNA damage, metabolism, transcription, and the immune response [95]. While 164 genes were downregulated in ApoE4-associated astrocytes, the remainder of the genes was upregulated [95]. This differential regulation by ApoE4 could be a direct or indirect effect.

In another twist to its transcriptional role, Urfer et al. showed that the DNA sequence of the ApoE4 isoform itself controls the expression of other genes located in close proximity to the ApoE gene [96]. The ApoE4 DNA encompasses a de novo binding motif for the well-known transcription factor NRF1 whose targets include several neurodegenerative disease-related genes, such as PARK2, PINK1, PARK7, GPR37, PSENEN, and MAPK. The ApoE4/NRF1 complex offers a potential mechanism that links various triggers to an aberrant gene expression that influence the AD process. However, it is not clear from this study if the ApoE4/NRF1 DNA complex acts as a transcriptional activator, repressor, or both.



Direct Transcriptional Role of ApoE

One of the earliest reports indicating a transcriptional role of ApoE was reported by Zhang et al., when they described that ApoE selectively altered the type of steroid made by ovarian theca/interstitial cells by regulating the transcription of P450 17α-hydroxylase-C17–20 lyase mRNA [97]. Similarly, Levros et al. showed that ApoE constitutively bound to the apolipoprotein D (ApoD) promoter and repressed the ApoD promoter activity, thus demonstrating for the first time a transcriptional role for ApoE [98].

We too recently reported the transcriptional role of ApoE, first by showing that both ApoE3 and E4 bound to the Sirtuin1 (SirT1) promoter. Using several different procedures, we showed that ApoE4 and not ApoE3 significantly reduced SirT1 mRNA levels, SirT1 protein expression, and SirT1 enzyme activity [4]. Furthermore, we demonstrated that ApoE bound DNAwith high affinity (low nanomolar) in the promoter regions of over ~ 1700 genes. The genes associated with these promoters provided new insight into the mechanism by which AD risk is conferred by ApoE4, because they included genes associated with trophic support, programmed cell death, microtubule disassembly, synaptic function, aging, and insulin resistance, all processes that have been implicated in AD pathogenesis [4] (Figure 1).

While both ApoE3 and E4 regulate transcription, they have different outputs, with ApoE4 associated with promoters of genes linked to Alzheimer’s disease [4]. ApoE not only bound to the SirT1 promoter with high affinity but also repressed the promoter activity, suggesting a plausible role for ApoE4 in the nucleus as a transcriptional repressor in at least some settings. This conclusion was supported by the finding that four other genes—MADD, ADNP, COMMD6, and PPP2R5E (B56ε)— whose promoters also interacted with ApoE4, all showed reduced transcription in the presence of ApoE4 (and, to a lesser extent, in the presence of ApoE3) [4, 99]. There was a significant reduction in mRNA and protein levels of all four genes only in ApoE4-transfected cells compared with ApoE3- transfected cells or untransfected cells.

Immunohistochemical labeling of MADD, ADNP, and COMMD6 revealed greater expression of these proteins in the septal region of brains from 7-month-old ApoE3 TR mice compared with ApoE4 TR mice. MADD and COMMD6 regulate transcription by binding to the NFκB complex while ADNP functions as both an anti-apoptotic and anti-inflammatory gene [100–103]. Thus, the protein products of all three genes affect the inflammatory response, and, in each case, repression by ApoE4 would be expected to trigger inflammation and cell death, which is a hallmark of AD pathophysiology.

Protein phosphatase 2A (PP2A) is the principal tau dephosphorylating enzyme in the brain. Several abnormalities of PP2A have been reported in AD, including decreased protein levels of PP2A, decreased mRNA and protein levels of PP2AC and variable regulatory B subunits, and reduced methylation of PP2AC, all of which results in the reduction of the PP2A phosphatase activity [104–113]. Using a combination of human glioblastoma cells, ApoE3/4, and ApoE–/– NSC and human postmortem tissue, we showed that while both ApoE3 and E4 bound to the PPP2R5E promoter, only ApoE4 triggered a significant reduction in PP2A activity [99]. ApoE4 significantly repressed PPP2R5E mRNA levels and also reduced protein expression. Additionally, ApoE4 triggered demethylation of the PP2AC subunit of PP2A, resulting in the disruption of the PPP2R5E–PP2AC complex [99]. All of the previous PP2A modifications by ApoE4 provide an explanation for the reduction in PP2A activity in AD compared with ApoE3. This may also explain the elevated Tau phosphorylation in AD human brains featuring at least one ApoE4 allele.

Taking all these results together, the direct effects of ApoE4 on transcription of a small number of genes studied to date reveal only repressive effects; nevertheless, since we have not yet analyzed the interaction of ApoE with all of the gene promoters, we cannot fully substantiate the role of ApoE as a transcriptional repressor.



Nuclear Localization of ApoE4

If ApoE4 has a direct transcriptional role as mentioned previously, it indicates that it must enter the nucleus. The mechanism by which ApoE isoforms escape the secretory pathway and translocate to the cytosol or the nucleus is currently unclear. ApoE4 is known to trigger cellular stress including endoplasmic reticulum (ER) stress and cell death [3, 114, 115], and several reports have suggested that cellular and/or ER stress can result in mislocalization of proteins, resulting in their redistribution to the cytosol and nucleus [66, 116–121].

Similarly, several studies on the fate of misfolded protein clumps in the lysosomal/endosomal vesicles point to the escape of the cellular contents into the cytosolic compartment. According to these studies, misfolded protein clumps rupture intracellular vesicles following endocytosis allowing the proteins to then invade the cytoplasm and cause additional dysfunction [66, 120–122]. Interestingly, neurons fail to effectively degrade Aβ resulting in the formation of high molecular weight Aβ species in endosomal vesicles [123, 124] that could potentially rupture lysosomal/endosomal vesicles.

It has been established that ApoE accelerates neuronal Aβ uptake and lysosomal trafficking in an isoform-dependent manner with E3 >> E4 [66, 120, 121]. Any abnormalities or disturbance in the endocytoic pathway may result in the rupture of the endosomal vesicles causing the ApoE protein to accumulate in the cytosol [66, 120, 121, 125] (Fig. 1). From the cytosol, both full-length ApoE or fragments of ApoE isoforms could be transported into the nucleus by binding to specific nuclear proteins through their weak polybasic domains that may serve as nuclear signaling sequences (NLS) [98, 126–128]. Additionally, a subpopulation of ApoE on the cell surface may be a source of nuclear ApoE [119, 129] (Fig. 1). ApoE, which binds to the cell surface nucleolin may be transported to the nucleus as an ApoE–nucleolin complex similar to the transfer of cell surface midkine–nucleolin complex and other cell surface heparin binding proteins into the nucleus [119, 130–132].

The presence of ApoE in the nucleus has already been demonstrated in several cell types and tissues [98, 119, 127, 128, 133, 134] and may partly explain ApoE’s role as a transcription factor [4, 98]. Cancer cells are particularly prone to ER stress and mislocalization of proteins, with ER stress contributing to either survival or apoptosis of cancer cells depending on the setting [135]. Interestingly, several reports have described a positive correlation of cytosolic/nuclear ApoE immunoreactivity and clinical aggressiveness in prostate and ovarian cancers [134, 136].



Is ApoE an Authentic Transcription Factor?

Using the ChIP assay and SPR, we showed that both ApoE3 and ApoE4 bound to double-stranded DNAwith high affinity and that this binding required a specific region of DNA, present in the SirT1 promoter. Out of a total of 3080 promoter peaks examined, ~ 1700 were found to be associated with ApoE4 but not ApoE3 (Fig. 1). Because the kinetics of binding of ApoE3 or ApoE4 to DNAwere not significantly different, our data are consistent with the notion that the DNA binding site on ApoE is not influenced by either the sequence variation or the oligomerization state but by a different distribution of oligomers [4]. The genes associated with these promoters provided new insight into the mechanism by which AD risk is conferred by ApoE4, because they included genes associated with trophic support, programmed cell death, microtubule disassembly, synaptic function, aging, and insulin resistance, all processes that have been implicated in AD pathogenesis (Fig. 1). More specifically, ApoE not only bound to the SirT1 promoter with high affinity but also repressed its promoter activity, suggesting a plausible role for ApoE4 in the nucleus as a transcriptional repressor in at least some settings.

Thus, although the direct effects of ApoE4 on transcription of the small number of genes studied to date reveal only repressive effects, we cannot yet exclude the possibility that ApoE may in some cases turn out, with or without additional complex members, to function as a transcriptional activator. The estimated binding affinity of ApoE3 and ApoE4 (~ 10 nM) was in the range of known transcription factors [137, 138]. Both ApoE isoforms do not have high sequence similarity to the classical DNA binding proteins but do contain the helix–loop–helix secondary structure as well as high arginine content that is a characteristic of most DNA binding proteins [139, 140]. Binding of arginine residues to phosphate is very strong, and DNA-binding proteins use arginine to make critical interactions with the DNA backbone [141]. Because of its high arginine content, ApoE was originally named the arginine-rich protein [142], and these Arg residues might conceivably facilitate ApoE–DNA interaction.



Conclusions and Future Directions

It has been over two decades since the discovery that individuals carrying the ApoE4 allele are at increased risk for the development of AD in comparison to those possessing the ApoE3 or ApoE2 isoforms. Despite intensive research efforts that have revealed several important insights regarding ApoE4’s role in AD pathophysiology, a major unanswered question is the mechanism by which ApoE4 confers AD risk. Furthermore, not a single ApoE4-disease-modifying therapy has been approved for AD treatment thus far. Given that past failed clinical trials focused mostly on the same target — amyloid β-protein — it appears that targeting Aβ may not be enough to stop, reverse, or delay the disease. Genetic and biochemical research have revealed an extensive network of molecular interactions involved in AD pathogenesis, suggesting that a network-based therapeutics approach, rather than a single target-based approach, may be feasible and potentially more effective for the treatment of cognitive decline due to Alzheimer’s disease.

The fact that ApoE4 bound to a specific set of genes linked to the AD process suggests its increased vulnerability and as an AD risk factor. As discussed in the earlier sections, a relatively large body of literature now exists to support ApoE4’s roles in AD pathophysiology that are Aβ independent. The novel transcriptional role of ApoE4 needs more attention as it influences brain homeostasis beyond the known Aβ and Tau pathways [3, 4, 85]. The data supporting the transcriptional role of ApoE were mostly obtained using in vitro systems or using mouse models that are far from being physiological, although the effects were also observed in human AD brains. Indeed, human neuroblastoma and glioblastoma cells as well as human fibroblasts from AD patients do not reflect a system as complex as the human brain, and thus, as useful as the information obtained might be, it must be interpreted with caution. An essential next step in these studies is to determine whether the transcriptional effects of ApoE3 and ApoE4 are conserved in cells linked to AD, namely neurons and glial cells, especially when the proteins are expressed at endogenous levels. An attractive possibility to obtain a better understanding of ApoE’s transcriptional role in a physiological setting would be the use of isogenic iPSC-derived lineages expressing ApoE isoforms and exploring whether ApoE3/3 and ApoE4/4 neurons and glial cells differ transcriptionally from ApoE–/– controls.

Thus far, it appears that ApoE4, Tau and p-Tau, APP and p- APP, SirT1, PP2A, and inflammatory cytokines, including Il- 6 and Il-8, all may be part of a signaling network that is affected in AD [3–5, 57, 85], providing a high-throughput model for therapeutic candidate screening in AD drug discovery. Hence, this area needs much further exploration. Not discussed in this review is ApoE4’s additional role in triggering the immune response [4], which warrants further investigation.

In addition to ApoE4, the major risk factor for AD is aging itself, so future studies need to be directed at ApoE’s effects on specific cell populations (e.g., hippocampus and entorhinal cortex) in the brain as a consequence of aging itself. While most studies focus on animal models of AD, we need to be cautious about species differences between animals and humans in physiological function, lipid metabolism, the immune response, and cognition that might preclude mechanistic insights related to the disease. Human derived iPSCs and brain organoids would serve as the best models to complement the current animal models and pave the way toward a greater and clearer understanding of ApoE’s relationship with AD pathophysiology for effective therapies.


Acknowledgements

We thank the members of the DEB laboratory for helpful comments and discussions and Rowena Abulencia for administrative assistance. Our research work is supported in part by grants from the Buck-Impact Circle Funds (R.V.R), the Lucas Brothers Foundation (R.V.R), the Four Winds Foundation (D.E.B), the Marin Community Foundation (D.E.B), The John and Bonnie Strauss Foundation (R.V.R and D.E.B), and The Katherine Gehl Foundation (R.V.R and D.E.B).


Abbreviations

AD = Alzheimer’s disease
APP = β-amyloid precursor protein
ApoE = Apolipoprotein E
SirT1 = Sirtuin1
PP2A = Protein phosphatase 2A


Competing Interests

The authors declare that they have no competing interests.



References:

  1. Mahley RW, Rall SC Jr (2000) Apolipoprotein E: far more than a lipid transport protein. Annu Rev Genomics Hum Genet 1:507– 537. https://doi.org/10.1146/annurev.genom.1.1.507

  2. Verghese PB, Castellano JM, Holtzman DM (2011) Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol 10(3):241–252. https://doi.org/10.1016/ S1474-4422(10)70325-2

  3. Theendakara V, Patent A, Peters Libeu CA, Philpot B, Flores S, Descamps O, Poksay KS, Zhang Q et al (2013) Neuroprotective Sirtuin ratio reversed by ApoE4. Proc Natl Acad Sci U S A 110(45):18303–18308. https://doi.org/10.1073/pnas.1314145110

  4. Theendakara V, Peters-Libeu CA, Spilman P, Poksay KS, Bredesen DE, Rao RV (2016) Direct transcriptional effects of apolipoprotein E. J Neurosci 36(3):685–700. https://doi.org/10. 1523/JNEUROSCI.3562-15.2016

  5. Bredesen DE, Amos EC, Canick J, Ackerley M, Raji C, Fiala M, Ahdidan J (2016)
    Reversal of Cognitive Decline in Alzheimer's Disease
    Aging (Albany NY). 2016 (Jun); 8 (6): 1250–1258

  6. Raichlen DA, Alexander GE (2014) Exercise, APOE genotype, and the evolution of the human lifespan. Trends Neurosci 37(5): 247–255. https://doi.org/10.1016/j.tins.2014.03.001

  7. Huang Y, Mahley RW (2014) Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. NeurobiolDis 72(Pt A):3–12. https://doi.org/10.1016/j.nbd. 2014.08.025

  8. Huang Y, Weisgraber KH, Mucke L, Mahley RW (2004) Apolipoprotein E: diversity of cellular origins, structural and biophysical properties, and effects in Alzheimer’s disease. J Mol Neurosci 23(3):189–204. https://doi.org/10.1385/JMN:23:3:189

  9. Grehan S, Tse E, Taylor JM (2001) Two distal downstream enhancers direct expression of the human apolipoprotein E gene to astrocytes in the brain. J Neurosci 21(3):812–822

  10. Ignatius MJ, Gebicke-Haerter PJ, Pitas RE, Shooter EM (1987) Apolipoprotein E in nerve injury and repair. Prog Brain Res 71: 177–184

  11. Pitas RE, Boyles JK, Lee SH, Foss D, Mahley RW (1987) Astrocytes synthesize apolipoprotein E and metabolize apolipoprotein E-containing lipoproteins. Biochim Biophys Acta 917(1): 148–161

  12. Xu PT, Schmechel D, Qiu HL, Herbstreith M, Rothrock-Christian T, Eyster M, Roses AD, Gilbert JR (1999) Sialylated human apolipoprotein E (apoEs) is preferentially associated with neuronenriched cultures from APOE transgenic mice. Neurobiol Dis 6(1):63–75. https://doi.org/10.1006/nbdi.1998.0213

  13. Mahley RW,Weisgraber KH, HuangY (2006) Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, includingAlzheimer’s disease. Proc Natl Acad Sci U S A 103(15):5644– 5651. https://doi.org/10.1073/pnas.0600549103

  14. Pfrieger FW (2003) Cholesterol homeostasis and function in neurons of the central nervous system. Cell Mol Life Sci 60(6):1158– 1171. https://doi.org/10.1007/s00018-003-3018-7

  15. Liao F, Yoon H, Kim J (2017) Apolipoprotein E metabolism and functions in brain and its role in Alzheimer’s disease. Curr Opin Lipidol 28(1):60–67. https://doi.org/10.1097/MOL. 0000000000000383

  16. Giau VV, Bagyinszky E, An SS, Kim SY (2015) Role of apolipoprotein E in neurodegenerative diseases. Neuropsychiatr Dis Treat 11:1723–1737. https://doi.org/10.2147/NDT.S84266

  17. Helisalmi S, Linnaranta K, Lehtovirta M, Mannermaa A, Heinonen O, Ryynanen M, Riekkinen P Sr, Soininen H (1996) Apolipoprotein E polymorphism in patients with different neurodegenerative disorders. Neurosci Lett 205(1):61–64

  18. Baum L, Lam LC, Kwok T, Lee J, Chiu HF, Mok VC, Wong A, Chen X et al (2006) Apolipoprotein E epsilon4 allele is associated with vascular dementia. Dement Geriatr Cogn Disord 22(4):301– 305. https://doi.org/10.1159/000095246

  19. Yin YW, Li JC, Wang JZ, Li BH, Pi Y, Yang QW, Fang CQ, Gao CYet al (2012) Association between apolipoprotein E gene polymorphism and the risk of vascular dementia: a meta-analysis. Neurosci Lett 514(1):6–11. https://doi.org/10.1016/j.neulet.2012. 02.031

  20. Chuang YF, Hayden KM, Norton MC, Tschanz J, Breitner JC, Welsh-Bohmer KA, Zandi PP (2010) Association between APOE epsilon4 allele and vascular dementia: The Cache County study. Dement Geriatr Cogn Disord 29(3):248–253. https://doi. org/10.1159/000285166

  21. Stengard JH, Weiss KM, Sing CF (1998) An ecological study of association between coronary heart disease mortality rates in men and the relative frequencies of common allelic variations in the gene coding for apolipoprotein E. Hum Genet 103(2):234–241

  22. Stengard JH, Pekkanen J, Sulkava R, Ehnholm C, Erkinjuntti T, Nissinen A (1995) Apolipoprotein E polymorphism, Alzheimer’s disease and vascular dementia among elderly Finnish men. Acta Neurol Scand 92(4):297–298

  23. Stengard JH, Zerba KE, Pekkanen J, Ehnholm C, Nissinen A, Sing CF (1995) Apolipoprotein E polymorphism predicts death from coronary heart disease in a longitudinal study of elderly Finnish men. Circulation 91(2):265–269

  24. Lehtinen S, Lehtimaki T, Sisto T, Salenius JP, Nikkila M, Jokela H, Koivula T, Ebeling F et al (1995) Apolipoprotein E polymorphism, serum lipids, myocardial infarction and severity of angiographically verified coronary artery disease in men and women. Atherosclerosis 114(1):83–91

  25. Eichner JE, Kuller LH, Orchard TJ, Grandits GA, McCallum LM, Ferrell RE, Neaton JD (1993) Relation of apolipoprotein E phenotype to myocardial infarction and mortality from coronary artery disease. Am J Cardiol 71(2):160–165

  26. van der Walt A, Stankovich J, Bahlo M, Taylor BV, van der Mei IA, Foote SJ, Kilpatrick TJ, Rubio JP et al (2009) Apolipoprotein genotype does not influence MS severity, cognition, or brain atrophy. Neurology 73(13):1018–1025. https://doi.org/10.1212/ WNL.0b013e3181b9c85e

  27. Lill CM, Liu T, Schjeide BM, Roehr JT, Akkad DA, Damotte V, Alcina A, Ortiz MA et al (2012) Closing the case of APOE in multiple sclerosis: no association with disease risk in over 29 000 subjects. J Med Genet 49(9):558–562. https://doi.org/10.1136/ jmedgenet-2012-101175

  28. Savettieri G, Messina D, Andreoli V, Bonavita S, Caltagirone C, Cittadella R, Farina D, Fazio MC et al (2004) Gender-related effect of clinical and genetic variables on the cognitive impairment in multiple sclerosis. J Neurol 251(10):1208–1214. https://doi.org/ 10.1007/s00415-004-0508-y

  29. PinholtM, Frederiksen JL, ChristiansenM(2006) The association between apolipoprotein E and multiple sclerosis. Eur J Neurol 13(6):573–580. https://doi.org/10.1111/j.1468-1331.2006.01360. x

  30. Shi J, Zhao CB, Vollmer TL, Tyry TM, Kuniyoshi SM (2008) APOE epsilon 4 allele is associated with cognitive impairment in patients with multiple sclerosis. Neurology 70(3):185–190. https://doi.org/10.1212/01.wnl.0000264004.62612.44

  31. Koutsis G, Panas M, Giogkaraki E, Potagas C, Karadima G, Sfagos C, Vassilopoulos D (2007) APOE epsilon4 is associated with impaired verbal learning in patients with MS. Neurology 68(8):546–549. https://doi.org/10.1212/01.wnl.0000254468. 51973.44

  32. Ezquerra M, Campdelacreu J, Gaig C, Compta Y,Munoz E, Marti MJ, Valldeoriola F, Tolosa E (2008) Lack of association of APOE and tau polymorphisms with dementia in Parkinson’s disease. Neurosci Lett 448(1):20–23. https://doi.org/10.1016/j.neulet. 2008.10.018

  33. Ryu HG, Kwon OD (2010) Apolipoprotein E epsilon 4 allele is not associated with age at onset or MMSE of Parkinson’s disease in a Korean study. Parkinsonism Relat Disord 16(9):615–617. https://doi.org/10.1016/j.parkreldis.2010.06.015

  34. Federoff M, Jimenez-Rolando B, NallsMA, Singleton AB (2012) A large study reveals no association between APOE and Parkinson’s disease. Neurobiol Dis 46(2):389–392. https://doi. org/10.1016/j.nbd.2012.02.002

  35. Harhangi BS, de Rijk MC, van Duijn CM, Van Broeckhoven C, Hofman A, BretelerMM(2000) APOE and the risk of PD with or without dementia in a population-based study. Neurology 54(6): 1272–1276

  36. Alzheimer’s Association (2015) Alzheimer’s disease Facts and Figures. Alzheimers Dement 11(3):332–384

  37. Alzheimer’s Association (2017) Alzheimer’s disease Facts and Figures. Alzheimers Dement 13(4):325–373

  38. Huang Y (2011) Roles of apolipoprotein E4 (ApoE4) in the pathogenesis of Alzheimer’s disease: lessons from ApoE mouse models. Biochem Soc Trans 39(4):924–932. https://doi.org/10. 1042/BST0390924

  39. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P et al (2000) Inflammation and Alzheimer’s disease. Neurobiol Aging 21(3):383–421

  40. Finch CE, Morgan TE (2007) Systemic inflammation, infection, ApoE alleles, and Alzheimer disease: a position paper. Curr Alzheimer Res 4(2):185–189

  41. Bredesen DE (2009)
    Neurodegeneration in Alzheimer's disease: Caspases and Synaptic Element Interdependence
    Molecular Neurodegeneration 2009 (Jun 26);   4:   27

  42. Olsson A, Hoglund K, Sjogren M, Andreasen N, Minthon L, Lannfelt L, Buerger K, Moller HJ et al (2003) Measurement of alpha- and beta-secretase cleaved amyloid precursor protein in cerebrospinal fluid from Alzheimer patients. Exp Neurol 183(1): 74–80

  43. Cedazo-Minguez A, Wiehager B, Winblad B, Huttinger M, Cowburn RF (2001) Effects of apolipoprotein E (apoE) isoforms, beta-amyloid (Abeta) and apoE/Abeta complexes on protein kinase C-alpha (PKC-alpha) translocation and amyloid precursor protein (APP) processing in human SH-SY5Y neuroblastoma cells and fibroblasts. Neurochem Int 38(7):615–625

  44. Vincent B, Smith JD (2001) Astrocytes down-regulate neuronal beta-amyloid precursor protein expression and modify its processing in an apolipoprotein E isoform-specific manner. Eur J Neurosci 14(2):256–266

  45. Gonneaud J, Arenaza-Urquijo EM, FouquetM, Perrotin A, Fradin S, de La Sayette V, Eustache F, Chetelat G (2016) Relative effect of APOE epsilon4 on neuroimaging biomarker changes across the lifespan. Neurology 87(16):1696–1703. https://doi.org/10.1212/ WNL.0000000000003234

  46. Kantarci K, Lowe V, Przybelski SA, Weigand SD, Senjem ML, Ivnik RJ, Preboske GM, Roberts R et al (2012) APOE modifies the association between Abeta load and cognition in cognitively normal older adults. Neurology 78(4):232–240. https://doi.org/10. 1212/WNL.0b013e31824365ab

  47. Murphy KR, Landau SM, Choudhury KR, Hostage CA, Shpanskaya KS, Sair HI, Petrella JR, Wong TZ et al (2013) Mapping the effects of ApoE4, age and cognitive status on 18Fflorbetapir PET measured regional cortical patterns of betaamyloid density and growth. NeuroImage 78:474–480. https:// doi.org/10.1016/j.neuroimage.2013.04.048

  48. Huynh TV, Davis AA, Ulrich JD, Holtzman DM (2017) Apolipoprotein E and Alzheimer’s disease: the influence of apolipoprotein E on amyloid-beta and other amyloidogenic proteins. J Lipid Res 58(5):824–836. https://doi.org/10.1194/jlr.R075481

  49. Morris JC, Roe CM, Xiong C, Fagan AM, Goate AM, Holtzman DM, Mintun MA (2010) APOE predicts amyloid-beta but not tau Alzheimer pathology in cognitively normal aging. Ann Neurol 67(1):122–131. https://doi.org/10.1002/ana.21843

  50. Tokuda T, Calero M, Matsubara E, Vidal R, Kumar A, Permanne B, Zlokovic B, Smith JD et al (2000) Lipidation of apolipoprotein E influences its isoform-specific interaction with Alzheimer’s amyloid beta peptides. Biochem J 348(Pt 2):359–365

  51. Deane R, Sagare A, Hamm K, Parisi M, Lane S, Finn MB, Holtzman DM, Zlokovic BV (2008) apoE isoform-specific disruption of amyloid beta peptide clearance from mouse brain. J Clin Invest 118(12):4002–4013. https://doi.org/10.1172/ JCI36663

  52. LaDu MJ, FaldutoMT,Manelli AM, Reardon CA, Getz GS, Frail DE (1994) Isoform-specific binding of apolipoprotein E to betaamyloid. J Biol Chem 269(38):23403–23406

  53. Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, Bu G, Frieden C et al (2013) ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci U S A 110(19): E1807–E1816. https://doi.org/10.1073/pnas.1220484110

  54. Raber J,Wong D, ButtiniM, Orth M, Bellosta S, Pitas RE,Mahley RW, Mucke L (1998) Isoform-specific effects of human apolipoprotein E on brain function revealed in ApoE knockout mice: increased susceptibility of females. Proc Natl Acad Sci U S A 95(18):10914–10919

  55. Vitek MP, Brown CM, Colton CA (2009) APOE genotypespecific differences in the innate immune response. Neurobiol Aging 30(9):1350–1360. https://doi.org/10.1016/ j . neurobiolaging.2007.11.014

  56. Zhao L, Lin S, Bales KR, Gelfanova V, Koger D, Delong C, Hale J, Liu F et al (2009) Macrophage-mediated degradation of betaamyloid via an apolipoprotein E isoform-dependent mechanism. J Neurosci 29(11):3603–3612. https://doi.org/10.1523/ JNEUROSCI.5302-08.2009

  57. Famenini S, Rigali EA, Olivera-Perez HM, Dang J, Chang MT, Halder R, Rao RV, Pellegrini M et al (2017) Increased intermediate M1-M2 macrophage polarization and improved cognition in mild cognitive impairment patients on omega-3 supplementation. FASEB J 31(1):148–160. https://doi.org/10.1096/fj. 201600677RR

  58. Kanekiyo T, Bu G (2014) The low-density lipoprotein receptorrelated protein 1 and amyloid-beta clearance in Alzheimer’s disease. Front Aging Neurosci 6:93. https://doi.org/10.3389/fnagi. 2014.00093

  59. Leissring MA (2008) The AbetaCs of Abeta-cleaving proteases. J Biol Chem 283(44):29645–29649. https://doi.org/10.1074/jbc. R800022200

  60. Leissring MA (2016) Abeta-degrading proteases: therapeutic potential in Alzheimer disease. CNS Drugs 30(8):667–675. https:// doi.org/10.1007/s40263-016-0364-1

  61. Turner AJ, Nalivaeva NN (2007) New insights into the roles of metalloproteinases in neurodegeneration and neuroprotection. Int Rev Neurobiol 82:113–135. https://doi.org/10.1016/S0074- 7742(07)82006-X

  62. Iwata N, Tsubuki S, Takaki Y, Shirotani K, Lu B, Gerard NP, Gerard C, Hama E et al (2001) Metabolic regulation of brain Abeta by neprilysin. Science 292(5521):1550–1552. https://doi. org/10.1126/science.1059946

  63. Cook DG, Leverenz JB, McMillan PJ, Kulstad JJ, Ericksen S, Roth RA, Schellenberg GD, Jin LWet al (2003) Reduced hippocampal insulin-degrading enzyme in late-onset Alzheimer’s disease is associated with the apolipoprotein E-epsilon4 allele. Am J Pathol 162(1):313–319

  64. Du J, Chang J, Guo S, Zhang Q, Wang Z (2009) ApoE 4 reduces the expression of Abeta degrading enzyme IDE by activating the NMDA receptor in hippocampal neurons. Neurosci Lett 464(2): 140–145. https://doi.org/10.1016/j.neulet.2009.07.032

  65. Jiang Q, Lee CY, Mandrekar S,Wilkinson B, Cramer P, Zelcer N, Mann K, Lamb B et al (2008) ApoE promotes the proteolytic degradation of Abeta. Neuron 58(5):681–693. https://doi.org/10. 1016/j.neuron.2008.04.010

  66. Li J, Kanekiyo T, Shinohara M, Zhang Y, LaDu MJ, Xu H, Bu G (2012) Differential regulation of amyloid-beta endocytic trafficking and lysosomal degradation by apolipoprotein E isoforms. J Biol Chem 287(53):44593–44601. https://doi.org/10.1074/jbc. M112.420224

  67. Tai LM, Youmans KL, Jungbauer L, Yu C, Ladu MJ (2011) Introducing human APOE into Abeta transgenic mouse models. Int J Alzheimers Dis 2011:810981. https://doi.org/10.4061/2011/ 810981

  68. Brecht WJ, Harris FM, Chang S, Tesseur I, Yu GQ, Xu Q, Dee Fish J,Wyss-Coray T et al (2004) Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice. J Neurosci 24(10):2527–2534. https:// doi.org/10.1523/JNEUROSCI.4315-03.2004

  69. Buttini M, Akeefe H, Lin C, Mahley RW, Pitas RE, Wyss-Coray T, Mucke L (2000) Dominant negative effects of apolipoprotein E4 revealed in transgenic models of neurodegenerative disease. Neuroscience 97(2):207–210

  70. Buttini M,OrthM, Bellosta S, Akeefe H, Pitas RE,Wyss-Coray T, Mucke L, Mahley RW (1999) Expression of human apolipoprotein E3 or E4 in the brains of Apoe–/– mice: isoform-specific effects on neurodegeneration. J Neurosci 19(12):4867–4880

  71. Harris FM, BrechtWJ, Xu Q, Tesseur I, Kekonius L,Wyss-Coray T, Fish JD, Masliah E et al (2003) Carboxyl-terminal-truncated apolipoprotein E4 causes Alzheimer’s disease-like neurodegeneration and behavioral deficits in transgenic mice. Proc Natl Acad Sci U S A 100(19):10966–10971. https://doi.org/10.1073/pnas. 1434398100

  72. Huang Y, Liu XQ,Wyss-Coray T, Brecht WJ, Sanan DA,Mahley RW (2001) Apolipoprotein E fragments present in Alzheimer’s disease brains induce neurofibrillary tangle-like intracellular inclusions in neurons. Proc Natl Acad Sci U S A 98(15):8838–8843. https://doi.org/10.1073/pnas.151254698

  73. Andrews-Zwilling Y, Bien-Ly N, Xu Q, Li G, Bernardo A, Yoon SY, Zwilling D, Yan TX et al (2010) Apolipoprotein E4 causes age- and Tau-dependent impairment of GABAergic interneurons, leading to learning and memory deficits in mice. J Neurosci 30(41):13707–13717. https://doi.org/10.1523/JNEUROSCI. 4040-10.2010

  74. Andrews-Zwilling Y, Gillespie AK, Kravitz AV, Nelson AB, Devidze N, Lo I, Yoon SY, Bien-Ly N et al (2012) Hilar GABAergic interneuron activity controls spatial learning and memory retrieval. PLoS One 7(7):e40555. https://doi.org/10. 1371/journal.pone.0040555

  75. Tesseur I, Van Dorpe J, Spittaels K, Van den Haute C, Moechars D, Van Leuven F (2000) Expression of human apolipoprotein E4 in neurons causes hyperphosphorylation of protein tau in the brains of transgenic mice. Am J Pathol 156(3):951–964. https:// doi.org/10.1016/S0002-9440(10)64963-2

  76. Tesseur I, Van Dorpe J, Bruynseels K, Bronfman F, Sciot R, Van Lommel A, Van Leuven F (2000) Prominent axonopathy and disruption of axonal transport in transgenic mice expressing human apolipoprotein E4 in neurons of brain and spinal cord.AmJ Pathol 157(5):1495–1510. https://doi.org/10.1016/S0002-9440(10) 64788-8

  77. Sullivan PM, Han B, Liu F, Mace BE, Ervin JF, Wu S, Koger D, Paul S et al (2011) Reduced levels of human apoE4 protein in an animal model of cognitive impairment. Neurobiol Aging 32(5): 791–801. https://doi.org/10.1016/j.neurobiolaging.2009.05.011

  78. Bour A, Grootendorst J, Vogel E, Kelche C, Dodart JC, Bales K, Moreau PH, Sullivan PM et al (2008) Middle-aged human apoE4 targeted-replacement mice show retention deficits on a wide range of spatial memory tasks. Behav Brain Res 193(2):174–182. https://doi.org/10.1016/j.bbr.2008.05.008

  79. Dumanis SB, Tesoriero JA, Babus LW, Nguyen MT, Trotter JH, Ladu MJ, Weeber EJ, Turner RS et al (2009) ApoE4 decreases spine density and dendritic complexity in cortical neurons in vivo. J Neurosci 29(48):15317–15322. https://doi.org/10.1523/ JNEUROSCI.4026-09.2009

  80. Grootendorst J, Bour A, Vogel E, Kelche C, Sullivan PM, Dodart JC, Bales K, Mathis C (2005) Human apoE targeted replacement mouse lines: h-apoE4 and h-apoE3 mice differ on spatial memory performance and avoidance behavior. Behav Brain Res 159(1):1– 14. https://doi.org/10.1016/j.bbr.2004.09.019

  81. Holtzman DM, Bales KR, Tenkova T, Fagan AM, Parsadanian M, Sartorius LJ, Mackey B, Olney J et al (2000) Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 97(6):2892–2897. https://doi.org/10.1073/pnas.050004797

  82. Holtzman DM, Fagan AM, Mackey B, Tenkova T, Sartorius L, Paul SM, Bales K, Ashe KH et al (2000) Apolipoprotein E facilitates neuritic and cerebrovascular plaque formation in an Alzheimer’s disease model. Ann Neurol 47(6):739–747

  83. Ji Y, Gong Y, Gan W, Beach T, Holtzman DM, Wisniewski T (2003) Apolipoprotein E isoform-specific regulation of dendritic spine morphology in apolipoprotein E transgenic mice and Alzheimer’s disease patients. Neuroscience 122(2):305–315

  84. Li G, Bien-Ly N, Andrews-Zwilling Y, Xu Q, Bernardo A, Ring K, Halabisky B, Deng C et al (2009) GABAergic interneuron dysfunction impairs hippocampal neurogenesis in adult apolipoprotein E4 knockin mice. Cell Stem Cell 5(6):634–645. https:// doi.org/10.1016/j.stem.2009.10.015

  85. Lattanzio F, Carboni L, Carretta D, Rimondini R, Candeletti S, Romualdi P (2014) Human apolipoprotein E4 modulates the expression of Pin1, Sirtuin 1, and Presenilin 1 in brain regions of targeted replacement apoE mice. Neuroscience 256:360–369. https://doi.org/10.1016/j.neuroscience.2013.10.017

  86. Kumar R, Chaterjee P, Sharma PK, Singh AK, Gupta A, Gill K, Tripathi M, Dey AB et al (2013) Sirtuin1: a promising serum protein marker for early detection of Alzheimer’s disease. PLoS One 8(4):e61560. https://doi.org/10.1371/journal.pone.0061560

  87. Lee HR, Shin HK, Park SY, Kim HY, Lee WS, Rhim BY, Hong KW, Kim CD (2014) Cilostazol suppresses beta-amyloid production by activating a disintegrin and metalloproteinase 10 via the upregulation of SIRT1-coupled retinoic acid receptor-beta. J Neurosci Res 92(11):1581–1590. https://doi.org/10.1002/jnr. 23421

  88. Herskovits AZ, Guarente L (2014) SIRT1 in neurodevelopment and brain senescence. Neuron 81(3):471–483. https://doi.org/10. 1016/j.neuron.2014.01.028

  89. Porquet D, Casadesus G, Bayod S, Vicente A, Canudas AM, Vilaplana J, Pelegri C, Sanfeliu C et al (2013) Dietary resveratrol prevents Alzheimer’s markers and increases life span in SAMP8. Age 35(5):1851–1865. https://doi.org/10.1007/s11357-012-9489- 4

  90. Rege SD, Geetha T, Griffin GD, Broderick TL, Babu JR (2014) Neuroprotective effects of resveratrol in Alzheimer disease pathology. Front Aging Neurosci 6:218. https://doi.org/10.3389/fnagi. 2014.00218

  91. Hubbard BP, Sinclair DA (2014) Smallmolecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci 35(3):146–154. https://doi.org/10.1016/j.tips. 2013.12.004

  92. Min SW, Cho SH, Zhou Y, Schroeder S, Haroutunian V, Seeley WW, Huang EJ, Shen Yet al (2010) Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67(6):953–966. https://doi.org/10.1016/j.neuron.2010.08.044

  93. Sen A, Nelson TJ, Alkon DL (2015) ApoE4 and Abeta oligomers reduce BDNF expression via HDAC nuclear translocation. J Neurosci 35(19):7538–7551. https://doi.org/10.1523/ JNEUROSCI.0260-15.2015

  94. Huang YA, Zhou B, Wernig M, Sudhof TC (2017) ApoE2, ApoE3, and ApoE4 differentially stimulate APP transcription and Abeta secretion. Cell 168(3):427–441 e421. https://doi.org/ 10.1016/j.cell.2016.12.044

  95. Simpson JE, Ince PG, Shaw PJ, Heath PR, Raman R, Garwood CJ, Gelsthorpe C, Baxter L et al (2011) Microarray analysis of the astrocyte transcriptome in the aging brain: relationship to Alzheimer’s pathology and APOE genotype. Neurobiol Aging 32(10):1795–1807. https://doi.org/10.1016/j.neurobiolaging. 2011.04.013

  96. Urfer-Buchwalder A, Urfer R (2017) Identification of a nuclear respiratory factor 1 recognition motif in the apolipoprotein E variant APOE4 linked to Alzheimer’s disease. Sci Rep 7:40668. https://doi.org/10.1038/srep40668

  97. Zhang G, Curtiss LK,Wade RL, Dyer CA (1998) An apolipoprotein E synthetic peptide selectively modulates the transcription of the gene for rat ovarian theca and interstitial cell P450 17alphahydroxylase, C17-20 lyase. J Lipid Res 39(12):2406–2414

  98. Levros LC Jr, Labrie M, Charfi C, Rassart E (2013) Binding and repressive activities of apolipoprotein E3 and E4 isoforms on the human ApoD promoter. Mol Neurobiol 48(3):669–680. https:// doi.org/10.1007/s12035-013-8456-0

  99. Theendakara V, Bredesen DE, Rao RV (2017) Downregulation of protein phosphatase 2A by apolipoprotein E: implications for Alzheimer’s disease. Mol Cell Neurosci. https://doi.org/10.1016/ j.mcn.2017.07.002

  100. Del Villar K, Miller CA (2004) Down-regulation of DENN/MADD, a TNF receptor binding protein, correlates with neuronal cell death in Alzheimer’s disease brain and hippocampal neurons. Proc Natl Acad Sci U S A 101(12):4210–4215. https:// doi.org/10.1073/pnas.0307349101

  101. Quintana FJ, Zaltzman R, Fernandez-Montesinos R, Herrera JL, Gozes I, Cohen IR, Pozo D (2006) NAP, a peptide derived from the activity-dependent neuroprotective protein, modulates macrophage function. Ann N YAcad Sci 1070:500–506. https://doi.org/ 10.1196/annals.1317.069

  102. de Bie P, van de Sluis B, Burstein E, Duran KJ, Berger R, Duckett CS, Wijmenga C, Klomp LW (2006) Characterization of COMMD protein-protein interactions in NF-kappaB signalling. Biochem J 398(1):63–71. https://doi.org/10.1042/BJ20051664

  103. Fernandez-Montesinos R, Torres M, Baglietto-Vargas D, Gutierrez A, Gozes I, Vitorica J, Pozo D (2010) Activity-dependent neuroprotective protein (ADNP) expression in the amyloid precursor protein/presenilin 1 mouse model of Alzheimer’s disease. J Mol Neurosci 41(1):114– 120. https://doi.org/10.1007/s12031-009-9300-x

  104. Janssens V, Longin S, Goris J (2008) PP2A holoenzyme assembly: in cauda venenum (the sting is in the tail). Trends Biochem Sci 33(3):113–121. https://doi.org/10.1016/j.tibs.2007.12.004

  105. Torrent L, Ferrer I (2012) PP2A and Alzheimer disease. Curr Alzheimer Res 9(2):248–256

  106. Sontag JM, Sontag E (2014) Protein phosphatase 2A dysfunction in Alzheimer’s disease. Front Mol Neurosci 7:16. https://doi.org/ 10.3389/fnmol.2014.00016

  107. Martin L, Latypova X, Wilson CM, Magnaudeix A, Perrin ML, Terro F (2013) Tau protein phosphatases in Alzheimer’s disease: the leading role of PP2A. Ageing Res Rev 12(1):39–49. https:// doi.org/10.1016/j.arr.2012.06.008

  108. Iqbal K, Liu F, Gong CX (2016) Tau and neurodegenerative disease: the story so far. Nat Rev Neurol 12(1):15–27. https://doi.org/ 10.1038/nrneurol.2015.225

  109. Sommer LM, Cho H, Choudhary M, Seeling JM (2015) Evolutionary analysis of the B56 gene family of PP2A regulatory subunits. Int J Mol Sci 16(5):10134–10157. https://doi.org/10. 3390/ijms160510134

  110. Sangodkar J, Farrington CC, McClinch K, Galsky MD, Kastrinsky DB, Narla G (2016) All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J 283(6):1004–1024. https://doi.org/10.1111/febs.13573

  111. Vogelsberg-Ragaglia V, Schuck T, Trojanowski JQ, Lee VM (2001) PP2A mRNA expression is quantitatively decreased in Alzheimer’s disease hippocampus. Exp Neurol 168(2):402–412. https://doi.org/10.1006/exnr.2001.7630

  112. Sontag E, Luangpirom A, Hladik C, Mudrak I, Ogris E, Speciale S, White CL 3rd (2004) Altered expression levels of the protein phosphatase 2A ABalphaC enzyme are associated with Alzheimer disease pathology. J Neuropathol Exp Neurol 63(4):287–301

  113. Braithwaite SP, Stock JB, Lombroso PJ, Nairn AC (2012) Protein phosphatases and Alzheimer’s disease. Prog Mol Biol Transl Sci 106:343–379. https://doi.org/10.1016/B978-0-12-396456-4. 00012-2

  114. Zhong N, Ramaswamy G,Weisgraber KH (2009) Apolipoprotein E4 domain interaction induces endoplasmic reticulum stress and impairs astrocyte function. J Biol Chem 284(40):27273–27280. https://doi.org/10.1074/jbc.M109.014464

  115. Cash JG, Kuhel DG, Basford JE, Jaeschke A, Chatterjee TK, Weintraub NL, Hui DY (2012) Apolipoprotein E4 impairs macrophage efferocytosis and potentiates apoptosis by accelerating endoplasmic reticulum stress. J Biol Chem 287(33):27876– 27884. https://doi.org/10.1074/jbc.M112.377549

  116. Rao RV, Bredesen DE (2004) Misfolded proteins, endoplasmic reticulum stress and neurodegeneration. Curr Opin Cell Biol 16(6):653–662

  117. Drori A, Misaghi S, Haimovich J, Messerle M, Tirosh B (2010) Prolonged endoplasmic reticulum stress promotes mislocalization of immunoglobulins to the cytoplasm. Mol Immunol 47(9):1719– 1727. https://doi.org/10.1016/j.molimm.2010.03.006

  118. Miesbauer M, Rambold AS, Winklhofer KF, Tatzelt J (2010) Targeting of the prion protein to the cytosol: mechanisms and consequences. Curr Issues Mol Biol 12(2):109–118

  119. Quinn CM, Kagedal K, Terman A, Stroikin U, Brunk UT, Jessup W, Garner B (2004) Induction of fibroblast apolipoprotein E expression during apoptosis, starvation-induced growth arrest and mitosis. Biochem J 378(Pt 3):753–761. https://doi.org/10.1042/ BJ20031352

  120. Cataldo AM, Peterhoff CM, Troncoso JC, Gomez-Isla T, Hyman BT, Nixon RA (2000) Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer’s disease and Down syndrome: differential effects of APOE genotype and presenilin mutations. Am J Pathol 157(1):277–286

  121. Ji ZS, Miranda RD, Newhouse YM, Weisgraber KH, Huang Y, Mahley RW (2002) Apolipoprotein E4 potentiates amyloid beta peptide-induced lysosomal leakage and apoptosis in neuronal cells. J Biol Chem 277(24):21821–21828. https://doi.org/10. 1074/jbc.M112109200

  122. FlavinWP, Bousset L, Green ZC, Chu Y, Skarpathiotis S, Chaney MJ, Kordower JH, Melki R et al (2017) Endocytic vesicle rupture is a conserved mechanism of cellular invasion by amyloid proteins. Acta Neuropathol. https://doi.org/10.1007/s00401-017- 1722-x

  123. Zerbinatti CV, Wahrle SE, Kim H, Cam JA, Bales K, Paul SM, Holtzman DM, Bu G (2006) Apolipoprotein E and low density lipoprotein receptor-related protein facilitate intraneuronal Abeta42 accumulation in amyloid model mice. J Biol Chem 281(47):36180–36186. https://doi.org/10.1074/jbc.M604436200

  124. Hu X, Crick SL, Bu G, Frieden C, Pappu RV, Lee JM (2009) Amyloid seeds formed by cellular uptake, concentration, and aggregation of the amyloid-beta peptide. Proc Natl Acad Sci U S A 106(48):20324–20329. https://doi.org/10.1073/pnas. 0911281106

  125. Lee CY, Tse W, Smith JD, Landreth GE (2012) Apolipoprotein E promotes beta-amyloid trafficking and degradation by modulating microglial cholesterol levels. J Biol Chem 287(3):2032–2044. https://doi.org/10.1074/jbc.M111.295451

  126. Kim WS, Elliott DA, KockxM, Kritharides L, Rye KA, Jans DA, Garner B (2008) Analysis of apolipoprotein E nuclear localization using green fluorescent protein and biotinylation approaches. Biochem J 409(3):701–709. https://doi.org/10.1042/BJ20071261

  127. Rohn TT (2013) Proteolytic cleavage of apolipoprotein E4 as the keystone for the heightened risk associated with Alzheimer’s disease. Int J Mol Sci 14(7):14908–14922. https://doi.org/10.3390/ ijms140714908

  128. Love JE, Day RJ, Gause JW, Brown RJ, Pu X, Theis DI, Caraway CA, Poon WWet al (2017) Nuclear uptake of an amino-terminal fragment of apolipoprotein E4 promotes cell death and localizes within microglia of the Alzheimer’s disease brain. Int J Physiol Pathophysiol Pharmacol 9(2):40–57

  129. Fazio S, Linton MF, Swift LL (2000) The cell biology and physiologic relevance of ApoE recycling. Trends Cardiovasc Med 10(1):23–30

  130. Semenkovich CF, Ostlund RE Jr, Olson MO, Yang JW (1990) A protein partially expressed on the surface of HepG2 cells that binds lipoproteins specifically is nucleolin. Biochemistry 29(41): 9708–9713

  131. Said EA, Krust B, Nisole S, Svab J, Briand JP, Hovanessian AG (2002) The anti-HIV cytokine midkine binds the cell surfaceexpressed nucleolin as a low affinity receptor. J Biol Chem 277(40):37492–37502. https://doi.org/10.1074/jbc.M201194200

  132. Shibata Y, Muramatsu T, Hirai M, Inui T, Kimura T, Saito H, McCormick LM, Bu G et al (2002) Nuclear targeting by the growth factor midkine. Mol Cell Biol 22(19):6788–6796

  133. Panin LE, Russkikh GS, Polyakov LM (2000) Detection of apolipoprotein A-I, B, and E immunoreactivity in the nuclei of various rat tissue cells. Biochemistry (Mosc) 65(12):1419–1423

  134. Chen YC, Pohl G,Wang TL,Morin PJ, Risberg B, Kristensen GB, Yu A, Davidson B et al (2005) Apolipoprotein E is required for cell proliferation and survival in ovarian cancer. Cancer Res 65(1): 331–337

  135. Wang M, Kaufman RJ (2014) The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat Rev Cancer 14(9):581–597. https://doi.org/10.1038/nrc3800

  136. Venanzoni MC, Giunta S, Muraro GB, Storari L, Crescini C, Mazzucchelli R, Montironi R, Seth A (2003) Apolipoprotein E expression in localized prostate cancers. Int J Oncol 22(4):779– 786

  137. Majka J, Speck C (2007) Analysis of protein-DNA interactions using surface plasmon resonance. Adv Biochem Eng Biotechnol 104:13–36

  138. Jantz D, Berg JM (2010) Probing the DNA-binding affinity and specificity of designed zinc finger proteins. Biophys J 98(5):852– 860. https://doi.org/10.1016/j.bpj.2009.11.021

  139. Rohs R, West SM, Sosinsky A, Liu P, Mann RS, Honig B (2009) The role of DNA shape in protein-DNA recognition. Nature 461(7268):1248–1253. https://doi.org/10.1038/nature08473

  140. LuscombeNM, Laskowski RA, Thornton JM (2001)Amino acidbase interactions: a three-dimensional analysis of protein-DNA interactions at an atomic level. Nucleic Acids Res 29(13):2860– 2874

  141. Rohs R, Jin X, West SM, Joshi R, Honig B, Mann RS (2010) Origins of specificity in protein-DNA recognition. Annu Rev Biochem 79:233–269. https://doi.org/10.1146/annurev-biochem- 060408-091030

  142. Danielsson B, Ekman R, Johansson BG,Nilsson-Ehle P, Petersson BG (1978) Isolation of a high density lipoprotein with high contents of arginine-rich apoprotein (apoE) from rat plasma. FEBS Lett 86(2):299–302

  143. Jaeger S, Pietrzik CU (2008) Functional role of lipoprotein receptors in Alzheimer’s disease. Curr Alzheimer Res 5(1):15–25

Return to ALZHEIMER's

Since 4-12-2020

                  © 1995–2024 ~ The Chiropractic Resource Organization ~ All Rights Reserved