Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Aug 10.
Published in final edited form as: Neurosci Lett. 2019 May 28;707:134285. doi: 10.1016/j.neulet.2019.134285

The role of APOE in transgenic mouse models of AD

Deebika Balu 1,#, Aimee James Karstens 1,2,#, Efstathia Loukenas 1, Juan Maldonado Weng 1, Jason M York 1, Ana Carolina Valencia-Olvera 1, Mary Jo LaDu 1,*
PMCID: PMC6717006  NIHMSID: NIHMS1533097  PMID: 31150730

Abstract

Identified in 1993, APOE4 is the greatest genetic risk factor for Alzheimer’s disease (AD), increasing risk up to 15-fold compared to the common variant APOE3. Since the mid 1990’s, transgenic (Tg) mice have been developed to model AD pathology and progression, primarily via expression of the familial AD (FAD) mutations in the presence of mouse-APOE (m-APOE). APOE4, associated with enhanced amyloid-β (Aβ) accumulation, has rarely been the focus in designing FAD-Tg mouse models. Initially, FAD-Tg mice were crossed with human (h)-APOE driven by heterologous promoters to identify an APOE genotype-specific AD phenotype. These models were later supplemented with FAD-Tg mice crossed with APOE-knockouts (APOE−/− or APOE-KO) and h-APOE-targeted replacement (h-APOE-TR) mice, originally generated to study the role of APOE genotype in peripheral lipid metabolism and atherosclerotic lesion development. Herein, we compare the m- and h-APOE multi-gene clusters, and then critically review the relevant history and approaches to developing a Tg mouse model to characterize APOE-dependent AD pathology, in combination with genetic (sex, age) and modifiable (e.g., inflammation, obesity) risk factors. Finally, we present recent data from the EFAD mice, which express 5xFAD mutations with the expression of the human apoE isoforms (E2FAD, E3FAD E4FAD). This includes a study of 6- and 18-month-old male and female E3FAD and E4FAD, a comparison that enables examination of the interaction among the main AD risk factors: age, APOE genotype and sex. While no single transgenic mouse can capture the effects of all modifiable and genetic risk factors, going forward, a conscious effort needs to be made to include the factors that most significantly modulate AD pathology.

Keywords: Alzheimer's disease (AD), familial AD transgenic mice (FAD-Tg), apolipoprotein E, sex and AD risk, APOE4 and AD risk, aging FAD-Tg mice, EFAD-Tg mouse model

Introduction

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with no cure and few palliative treatments [1]. Since the mid 90’s, transgenic (Tg) mice have been developed to model the phenotypic pathology and progression of AD, including amyloid plaques (amyloid-β peptide/Aβ), intracellular neurofibrillary tangles (tau; NFT), neuroinflammation, neuritic dystrophy, frank neuronal loss, and learning/memory deficits (reviewed in [75, 76]. Although it accounts for only 2-3% of AD cases, familial AD (FAD) is the result of autosomal dominant mutations in amyloid precursor protein (APP), presenilin-1 (PS1) or presenilin-2 (PS2) that result in an increase in Aβ42 or the ratio of Aβ42/Aβ40 [82]. As wild type (wt) mouse APP/PS1/PS2 expression does not lead to plaque deposition, FAD mutations in the human genes are commonly used to induce AD pathology in Tg mouse models (FAD-Tg mice) (for review, [76]). As summarized in Figure 1, the first generation of FAD-Tg mouse models were developed to understand the progression of Aβ pathology, including either single (Fig. 1: #2,3) [21, 25] or multiple FAD mutations (Fig. 1: #7,9,10,11,12) [29, 52, 55, 56, 62]. Mutations in the MAPT gene expressing microtubule-associated protein were later combined with the FAD mutations to induce tau pathology (Fig. 1: #10) [56]. In 1993, APOE4 was identified as the greatest genetic risk factor for AD and found to be associated with accelerated Aβ accumulation [16], both as amyloid and soluble oligomeric forms of Aβ (oAβ), the latter considered a proximal neurotoxin [15, 20, 23, 31, 32, 36, 64, 65, 80]. Despite the 5- to 15-fold increased risk in APOE4 carriers, many FAD-Tg mice fail to model the effects of human apoE isoforms. Specifically, FAD-Tg models continue to use murine (m)-APOE instead of human (h)-APOE or limit the spatial and temporal expression of h-APOE via heterologous promoters (Fig. 1: #6,8) Consequently, FAD-Tg models fail to capture the scope of the APOE genotype-specific effects, particularly on Aβ deposition and neuroinflammation. In addition, female sex profoundly increases the APOE4 related risk for AD, though the nature of this synergistic risk remains unclear [6, 19]. Thus, an ideal Tg model for AD would exhibit the relevant spatial and temporal progression of AD pathology, mirroring the APOE4- and female sex-based risk in humans (reviewed in [75]). Herein we critically review the relevant history and approaches to APOE-dependent AD pathology in Tg-mouse models.

Figure 1. Timeline of development of FAD- and APOE-transgenic mouse models.

Figure 1.

Example of FAD-, APOE- and crossed-transgenic mouse models developed over time (1992-2018). Boxes are numbered in chronological order for reference in text. Boxes are labeled: line 1- common name of transgenic model; line 2- genotype of mouse; line 3-promotor. Attributions: last name of first author, common abbreviation of journal, year of publication. NOTE: This timeline is not comprehensive but illustrative.

Murine vs human APOE: APOE−/− mice

APOE encodes the protein apolipoprotein E (apoE), the primary apolipoprotein expressed in the brain. While apolipoprotein J and apolipoprotein D are also expressed in the brain and are components of lipoproteins, these apolipoproteins do not support the synthesis or survival of a lipoprotein [17, 34]. In humans, apoE has 3 naturally occurring isoforms that differ by a single amino acid change at residues 112 and 158 (apoE2Cys,Cys, apoE3Cys,Arg, apoE4Arg,Arg). In contrast, m-apoE is expressed as a single isoform and differs from h-apoE at the protein level by ~100/300 amino acids. Based on the pioneering work of John Taylor and colleagues [4, 5, 18, 37, 57], h-APOE is now understood to be part of a ~58Kb multi-gene cluster on chromosome 19 that contains TOMM40/APOE/APOCI/APOCI’/APOCCIV/APOCII, while m-APOE is part of a ~37Kb gene cluster on chromosome 7 that contains TOMM40/APOE/APOCI/APOCCIV/APOCII [24, 83] (Fig. 2). Thus, APOE transcription is regulated by transcription factors in the promotor located up to −1000bp from the APOE transcription start site (TSS; reviewed in [47, 48]), as well as the interaction between these elements and far distal enhancers [3, 12, 58, 68-71]. These distal elements include the hepatic control regions (HCR) and multienhancer regions (ME) that contain, for example, liver X receptor (LXR) response elements (LXRE) that dictate cell-specific expression ([35, 67]; reviewed in [86]). Indeed, one of the unique aspects of APOE transcriptional regulation is that in the absence of distal enhancers, the promotor cannot direct gene transcription in vitro [35, 46, 79] or in vivo [3, 22, 35, 67, 71]. Recent concern has focused on the difference in the transcriptional regulation of the m- and h-APOE genes [47, 48], driven, in part, by the growing popularity of FAD-Tg mice with h-APOE, where h-APOE expression is driven by the m-APOE promotor. Of particular concern is the role of nuclear transcription factors in the expression of APOE and thus the number of, for example, LXRE consensus sequences in m- and h-APOE [13, 49, 86]. While these are important points to consider, a comprehensive review of this cluster regulation is beyond the scope of this review.

Figure 2. TOMM40/APOE/APOCI/APOCI’/APOCIV/APOCII multi-gene cluster.

Figure 2.

Schematic representation of the multi-gene clusters containing the h-APOE and m-APOE genes. The size of the clusters are defined by the 5’ transcription start site (TSS) of TOMM40 and 3’ end of the coding sequence of APOCII. The 5’ promotor regions and distal regulatory elements, including the hepatic control regions (HCR) and multi enhancers (ME), are identified. The +1 TSS of the h- and m-APOE genes are the basis for the location of other genes in the cluster. The locations of the HCR and ME are based on consensus sequences from www.ensembl.org and www.ncbi.nlm.nih.gov/gene. The exons and introns of the h- and m-APOE gene are from www.ncbi.nlm.nih.gov/IEB/Research/Acembly/index.html. All sizes are approximate, scaled based on gene sequence. In general, this figure is designed to be illustrative and not comprehensive.

Initially, APOE-Tg mice, including APOE-knockouts (APOE−/− or APOE-KO) (Fig. 1: #1) and APOE-targeted replacement (APOE-TR) (Fig. 1: #4) mice, were generated primarily to understand the role of apoE in peripheral lipid metabolism and atherosclerotic lesion development [73, 87]. Many FAD-Tg models continue to use murine (m)-APOE instead of human (h)-APOE or limit the spatial and temporal expression of h-APOE via heterologous promoters. However, APOE−/− and APOE-TR mice are being exploited in FAD-Tg models to study the role of apoE in the CNS (for review, [76]). Genetic deletion of m-APOE in FAD-Tg mice delays Aβ accumulation and amyloid deposition, as first demonstrated in the landmark paper with PDAPP+/+ APOE−/− mice (Fig. 1: #5) [8], and replicated using other FAD-Tg mouse models. Introducing h-APOE into FAD-Tg/m-APOE−/− mice further delays Aβ accumulation/amyloid deposition, with Aβ pathology eventually appearing in the sequence of h-apoE isoforms: apoE4 > apoE3 > apoE2. This temporal delay in pathology can be considerable, as with the PDAPP/APOE-TR mice where plaque deposition is delayed from 6- to 18-months (M) (Fig. 1: #13) [7]. These results underscore functional differences between m- and h-APOE on Aβ accumulation. Similar to amyloid deposition, apoE functioning varies in m-apoE vs. h-apoE in most phenotypic AD processes, including Aβ clearance, neuroinflammation and synaptic integrity (reviewed in [75]). Thus, in this comparison, the APOE−/− mouse is functionally similar to mouse models with h-APOE compared to m-APOE; thus, FAD-Tg mouse models expressing endogenous m-APOE are less relevant and predictive of human AD pathology.

Role of apoE in Tau pathology

As reviewed by Rebeck and colleagues, the deposition of Aβ is apoE isoform-specific, while the accumulation and progression of tau pathology is not apoE isoform-specific [63]. While Aβ plaques found in FAD-Tg mouse brains are structurally similar to those found in human AD brains, these mice do not develop NFTs composed of tau aggregates comparable to those found in human AD brains. Therefore, the role of microtubule associated protein tau (MAPT) has been well studied in frontal temporal dementia (FTD) and other variants of frontotemporal lobar degeneration (e.g., progressive supranuclear palsy) [27, 60, 72]. In FTD-Tg mouse models with mutations in human tau (e.g., Tau P301L, Tau P301S), NFTs form by 3-6M and may be accompanied by motor deficits [77, 84]. The MAPT mutations used to induce NFTs and tauopathy in mice do not increase AD risk but are associated with the rare FTD’s in humans [30]. Recently, in TauP301S × APOE-TR mice (Fig. 1: #18), it was demonstrated that in the presence of apoE4, tau levels increased with apoE4 > apoE3 = apoE2 [66]. In contrast, adeno-associated viral (AAV) injections of human tauP301L in neonatal APOE-TR mice show exacerbated tau pathology and behavioral deficits in the presence of apoE2 but not apoE4 [88]. Although inconsistent, the exacerbation of tau pathology by h-APOE4 in the TauP301S mouse is likely a response to the neurodegeneration and neuroinflammation induced by the mutant tau, not the product of a direct interaction between apoE4 with tau. Indeed, apoE4 likely acts as a “second hit” in cases where the CNS is already compromised by the “first hit” from causes that include TBI, neurodegeneration and even recovery from anesthesia [44]. Further evidence to consider in regard to apoE/tau interactions includes: 1. ApoE is not likely to influence tau pathology via a direct interaction under normal conditions in vivo as tau and apoE are not present in the same intracellular compartment. As a classic secretory protein, exocytosis of nascent apoE does not predict an interaction with tau, primarily a microtubule-associated protein found in the cytosol. 2. FAD-Tg mice do not mirror the temporal and pathological progression of tau pathology. Indeed, the only evidence for an apoE isoform-specific effect on tau hyperphosphorylation (apoE4 > apoE3) is in NSE-APOE mice (Fig. 1: #8), with relevance compromised by the use of a heterologous promoter that targets APOE expression to neurons, while apoE is primarily secreted by glial cells (for review, [76]). Regardless, these mice do not develop NFTs [61]. 3. Incorporating tauopathy in AD mouse models may not mirror human AD as MAPT mutations used to induce tauopathy in mice do not increase AD risk but are associated with the rare FTD’s in humans, as discussed above [30]. In conclusion, the utility of incorporating tau mutations in AD-Tg mouse models to determine if apoE exerts an isoform-specific effect on the development of tau pathology requires a careful interpretation and may have a limited relevance to AD as mutant tau causes FTD.

Temporal regulation of apoE expression (AAV delivery vs. Inducible models)

Crossing APOE-TR mice that express h-apoE isoforms with FAD-Tg mice results in apoE expression at physiological levels under the control of the m-apoE regulatory sequences. However, age-dependent mechanisms and critical periods for apoE isoform-specific modulation of AD pathology remain unclear. To address the stage of amyloid deposition where apoE4 has its greatest effect, both AAV-mediated gene delivery and Cre-mediated mouse models can be activated at different stages of pathology. AAV4-mediates overexpression of h-apoE in ependymal cells and astrocytes in the subventricular zone of FAD-Tg mouse brains via the ependymal system, while AAV8 driven by GFAP promotor leads to overexpression of h-apoE in all astrocytes of APOE-TR mice [26, 38]. Interestingly, in both studies Aβ-associated AD pathology was highest in apoE4-expressing mice. However, AAV-mediated CNS gene transfer in mice is not fully understood due to limited characterization of biodistribution profiles and immune responses with certain AAV serotypes. This limits the interpretation of these and future findings that use AAV-mediated gene delivery to study the effect of apoE expression on amyloid pathology at various stages. Crossing mice with the Cre transgene driven by the GFAP promotor with inducible apoE3 or apoE4 in a FAD-Tg mice provides astrocyte-specific expression (Fig. 1: #19). Thus, expression of apoE3 or apoE4 can be activated at different stages of AD pathology based on the Cre expression [41]. Alternatively, anti-sense oligos (ASOs) that target APOE3 or APOE4 mRNA expression in FAD-Tg mice lower apoE expression, again either before or after significant amyloid deposition [28]. In both studies, expression of apoE4 increased Aβ-associated AD pathology in the brain when expressed early in the development of AD pathology but not in the later stages. Thus, differential modulation of Aβ aggregation highlights the critical interaction between two AD risk factors: age and APOE.

Recent applications of APOE-Tg mouse models

In addition to the EFAD mice, a number of novel FAD/APOE-Tg models have been developed (Fig. 1). Other publications have relied on a comparison between FAD/m-APOE+/+ and FAD/m-APOE−/− to identify the function of “apoE,” actually m-apoE. The latter is concerning as this comparison, initially made by Bales in 1997 as discussed above, does not address the differential isoform-specific functions of h-APOE (for example [81]). Similarly, using a FAD/APOE-Tg mouse with mixed apoE4/m-apoE, differential effects on cerebral amyloid angiopathy (CAA) and parenchymal plaques were observed, suggesting opposing functions for m-apoE versus h-apoE4 (Fig. 1: #16) [40]. The question remains: What can be learned by comparing m-apoE to h-apoE4 that will inform the difference between the human apoE isoforms. With the FAD/APOE-Tg models, APOE4 has been linked to enhanced Aβ-induced deficits in insulin signaling [11] and support the use of an anti-h-apoE4 antibody to reduce apoE aggregation and plaque formation [39] [Fig. 1: #17]. Data from Koldamova and colleagues demonstrate the interactive effects of diet, APOE and sex on the immune response (Fig. 1: #15) [54]. Compared to normal diet, a high fat diet (HFD) increased amyloid deposition in ♂ and ♀ APOE4 carrier mice vs. ♂ and ♀ APOE3 carrier mice [53]. In addition, the HFD ♀APOE4 carriers also had the greatest microglial coverage surrounding plaques, thus establishing a link among diet, APOE, sex and the neuroimmune response [54].

EFAD mice: effects of modifiable and genetic risk factors on AD pathology

The EFAD-Tg mouse model (carriers: 5xFAD+/−/APOE+/+; non-carriers: 5xFAD−/−APOE++) was designed as a tractable preclinical FAD-Tg-mouse model for identifying the mechanisms underlying apoE isoform-specific modulation of AD pathology (Fig. 1: #14) [85]. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research [2, 9, 10, 14, 33, 42, 43, 51, 75, 89]. Recent publications have expanded the characterization of this model to include measures of cerebrovascular integrity and inflammation [50, 78]. In LPS-challenged ♂E4FAD carrier mice, peripheral inflammation increased cognitive decline and decreased post-synaptic protein levels [50]. Furthermore, chronic peripheral inflammation resulted in cerebrovascular deficits, suggesting that the interaction among APOE4, Aβ, and peripheral inflammation induced cerebrovascular damage and cognitive decline. Recent developments with the EFAD-Tg mouse presented in this review including the moderation effect of modifiable (e.g., inflammation, obesity) and genetic (e.g., sex, age) risk factors on AD pathology. Epidemiological evidence suggests that vascular risk and lifestyle factors (e.g., obesity, Western diet) increase AD risk in humans with increasing attention on the interplay of APOE genotype and sex [59]. In ♂EFAD carrier mice on a control diet, metabolic profiles for E3FAD = E4FAD, while the Western diet was associated with Aβ-associated pathology with E4FAD > EFAD3 [51]. This relationship differs by sex as recent findings show that in ♀EFAD carrier mice, Western diet yields E3FAD > E4FAD for select measures of both metabolic dysfunction and Aβ accumulation [14]. These results demonstrate a significant interaction between diet, APOE genotype and sex on AD pathology and an important gene-environment interaction [51].

Aged male and female EFAD mice

As an update on the EFAD mice, a recently completed study of an 18M cohort of ♀ and ♂ E3FAD and E4FAD mice was compared to 6M mice (n=8-12). The 6M male EFAD mouse data was adapted from [75]. All protocols follow the UIC Institutional Animal Care and Use Committee protocols. Breeding and colony maintenance was conducted at UIC. All the biochemical and immunohistochemical methods have been published previously [85]. Kaplan Meier survival curves for only the 18M cohort revealed a significance difference in survival: ♀E4FAD < (♂E4FAD ~ ♀E3FAD < (♂E3FAD (Fig. 3). Results from MWM in the 18M cohort demonstrated a significant memory loss in the ♀E4FAD compared to (♂E4FAD, ♀E3FAD, and (♂E3FAD (data not shown). The apoE and Aβ extraction profiles, using a 3-step sequential extraction with TBS, TBSX (TBS + 0.1 % TritonX) and FA (formic acid) were compared between 6M and 18M (Fig. 4 & 5). For apoE, total levels increased significantly from 6 to 18M in the 4 groups but with no difference between genotype and sex (Fig. 4A). However, in the TBSX fraction, the fraction where lipoprotein-associated apoE elutes, there was no age-induced increase in the ♀E4FAD mice, and ♀E4FAD < (♂E4FAD mice, and while there was an age-induced increase in the ♀E3FAD, ♀E3FAD < ♂E3FAD (Fig. 4B). Total Aβ increased from 6 to 18M in the 4 groups, with ♀E3FAD > ♂E3FAD and ♀E4FAD > (♂E4FAD (Fig. 5A). Importantly, soluble oAβ exhibited a similar increase from 6 to 18M, with the response at 18M: ♀E3FAD > ♂E3FAD and ♀E4FAD > (♂E4FAD (Fig 5B). There was no difference in the soluble Aβ42 levels at 18M among the 4 groups (Fig 5C). Immunohistochemistry for astrogliosis (GFAP) and Aβ deposition (Aβ mAb MOAB2) (Fig. 6A) revealed that at 18M, astrogliosis in the ♀E4FAD was > 6M, > (♂E4FAD and > ♀E3FAD, thus demonstrating significant age, sex and genotype effects (Fig. 6B). Aβ deposition in the cortex at 18M was also greatest in ♀E4FAD mice (Fig. 6C). These results indicate that measures of AD pathology in the EFAD mice develop with age, with 18M the calculated 75% survival rate for mice on their genetic background (https://www.jax.org/research-and-faculty/research-labs/the-harrison-lab/gerontology/life-span-as-a-biomarker), confirmed by the Kaplan Meier survival curve (Fig. 3). Both biochemical and immunohistochemical measures of AD pathology demonstrate significant interactive effects among age, sex and APOE. Aging studies with these mice are ongoing.

Figure 3. Kaplan Meier survival curves for 18M EFAD mice.

Figure 3.

Survival rates of 18M ♀E4FAD, (♂E4FAD, ♀E3FAD, and ♂E3FAD plotted as % survival. n = 8-12 mice per cohort.

Figure 4. ApoE levels in 6M and 18M EFAD mice.

Figure 4.

A) Total apoE levels; B) extraction profiles of apoE using a three-step sequential protein extraction (TBS, TBSX and FA) in the cortex of 6 and 18M EFAD mice measured by ELISA. Data expressed as mean ± S.E.M. and analyzed by two-way ANOVA followed by Bonferroni’s multiple comparison post hoc analysis. n=8-12, § p < 0.05 vs. 6M within sex and genotype (age effect); * p < 0.05 vs. males within genotype (sex effect); # p < 0.05 vs. E3FAD within sex (APOE genotype effect).

Figure 5. Aβ levels in 6M and 18M EFAD mice.

Figure 5.

Levels of: A) total Aβ42; B) soluble oligomeric Aβ; and C) soluble Aβ42 in the cortex of 6-and 18M EFAD mice measured by ELISA. Data expressed as mean ± S.E.M. and analyzed by two-way ANOVA followed by Bonferroni’s multiple comparison post hoc analysis. n=8-12, § p < 0.05 vs. 6M within sex and genotype (age effect); * p < 0.05 vs. males within genotype (sex effect); # p < 0.05 vs. E3FAD within sex (APOE genotype effect).

Figure 6. Aβ-associated neuroinflammation in 6M and 18M EFAD mice.

Figure 6.

A) Representative images of sagittal brain sections from 6- and 18M EFAD mice immunostained for astrogliosis (GFAP, green) and Aβ (MOAB2, red), x10 magnification (scale bar = 500 μm). Quantification of % area in cortex and hippocampus + subiculum covered by: B) astrocytes; C) Aβ deposition. Data expressed as mean ± S.E.M. and analyzed by two-way ANOVA followed by Bonferroni’s multiple comparison post hoc analysis. n=8-12, § p < 0.05 vs. 6M within sex and genotype (age effect); * p < 0.05 vs. males within genotype (sex effect); # p < 0.05 vs. E3FAD within sex (APOE genotype effect).

Conclusion

FAD-Tg mouse models are evolving in an effort to replicate the diverse pathology present in sporadic AD. However, information gleaned from these models is limited if basic risks that include age, APOE genotype, and sex are not considered. This is particularly critical for preclinical testing as the Tg model should mirror the human population, which includes, for example, APOE4 females. Furthermore, characterization of these effects should not ignore the potential contributions of peripheral phenotypes (e.g., Cre, AAV). In addition, the physiological relevance of certain models is still understudied and controversial. For example, while the use of inducible APOE-Tg models is a promising approach, it is still understudied. Comparisons between models with m-APOE and APOE−/− are not mechanistically informative or predictive of future therapeutic approaches as a case study of an APOE−/− human demonstrated deficits in learning, memory, language, visuospatial abilities, and executive functions as well as extensive xanthomas and a lipidemic plasma lipoprotein profile [45]. Further, m-apoE is not equivalent to the human apoE isoforms, thus requiring the introduction of h-APOE. In the end, no single transgenic mouse can capture the effects of all modifiable and genetic risk factors. Going forward, a conscious effort needs to be made to include the factors that most significantly modulate AD pathology. This approach will help to: 1) allow full interpretation of findings; and 2) validate a mouse model for testing therapeutics that translate to clinical trials.

Highlights:

  • APOE transgenic mouse models of Alzheimer’s disease have evolved overtime to include various methods for deleting or introducing the expression of apolipoprotein E.

  • Limitations of current APOE transgenic models include comparison with FAD/m-APOE with FAD/APOE−/− mice to determine the effect of APOE on AD pathology.

  • Few recent models are phenotypically characterized by APOE isoform with genetic (sex, age) and modifiable (e.g., inflammation, obesity) Alzheimer’s disease risk factors.

  • New aging data presented that further characterizes the EFAD mice (5xFAD+/−/APOE3+/+ or 5xFAD+/−/APOE4+/+) supports the interactive and synergistic effects of APOE genotype with sex on FAD-induced pathology.

Acknowledgements

This work was supported by NIH (UH2NS100127, R01AG057008, and RF1AG058067), Alzheimer’s Association SAGA and institutional funding from University of Illinois College of Medicine. AJK is currently supported by her T32 training grant (1T32AG057468-01). The authors have nothing to disclose.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].2017 Alzheimer's disease facts and figures, Alzheimer's & Dementia: The Journal of the Alzheimer's Association 13 325–373. [Google Scholar]
  • [2].Abdullah L, Evans JE, Emmerich T, Crynen G, Shackleton B, Keegan AP, Luis C, Tai L, LaDu MJ, Mullan M, Crawford F, Bachmeier C, APOE epsilon4 specific imbalance of arachidonic acid and docosahexaenoic acid in serum phospholipids identifies individuals with preclinical Mild Cognitive Impairment/Alzheimer's Disease, Aging (Albany NY) 9 (2017) 964–985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Allan CM, Taylor S, Taylor JM, Two hepatic enhancers HCR.1 and HCR.2, coordinate the liver expression of the entire human apolipoprotein E/C-I/C-IV/C-II gene cluster, J Biol Chem 272 (1997) 29113–29119. [DOI] [PubMed] [Google Scholar]
  • [4].Allan CM, Walker D, Segrest JP, Taylor JM, Identification and characterization of a new human gene (APOC4) in the apolipoprotein E, C-I, and C-II gene locus, Genomics 28 (1995) 291–300. [DOI] [PubMed] [Google Scholar]
  • [5].Allan CM, Walker D, Taylor JM, Evolutionary duplication of a hepatic control region in the human apolipoprotein E gene locus. Identification of a second region that confers high level and liver-specific expression of the human apolipoprotein E gene in transgenic mice, J Biol Chem 270 (1995) 26278–26281. [DOI] [PubMed] [Google Scholar]
  • [6].Altmann A, Tian L, Henderson VW, Greicius MD, Sex modifies the APOE-related risk of developing Alzheimer disease, Ann Neurol 75 (2014) 563–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Bales KR, Liu F, Wu S, Lin S, Koger D, DeLong C, Hansen JC, Sullivan PM, Paul SM, Human APOE isoform-dependent effects on brain beta-amyloid levels in PDAPP transgenic mice, The Journal of neuroscience 29 (2009) 6771–6779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Bales KR, Verina T, Dodel RC, Du Y, Altstiel L, Bender M, Hyslop P, Johnstone EM, Little SP, Cummins DJ, Piccardo P, Ghetti B, Paul SM, Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition, Nat Genet 17 (1997) 263–264. [DOI] [PubMed] [Google Scholar]
  • [9].Cacciottolo M, Christensen A, Moser A, Liu J, Pike CJ, Smith C, LaDu MJ, Sullivan PM, Morgan TE, Dolzhenko E, Charidimou A, Wahlund LO, Wiberg MK, Shams S, Chiang GC, I Alzheimer's Disease Neuroimaging, C.E. Finch, The APOE4 allele shows opposite sex bias in microbleeds and Alzheimer's disease of humans and mice, Neurobiol Aging 37 (2016) 47–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Cacciottolo M, Wang X, Driscoll I, Woodward N, Saffari A, Reyes J, Serre ML, Vizuete W, Sioutas C, Morgan TE, Gatz M, Chui HC, Shumaker SA, Resnick SM, Espeland MA, Finch CE, Chen JC, Particulate air pollutants, APOE alleles and their contributions to cognitive impairment in older women and to amyloidogenesis in experimental models, Transl Psychiatry 7 (2017) e1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Chan ES, Shetty MS, Sajikumar S, Chen C, Soong TW, Wong BS, ApoE4 expression accelerates hippocampus-dependent cognitive deficits by enhancing Abeta impairment of insulin signaling in an Alzheimer's disease mouse model, Sci Rep 6 (2016) 26119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Chang DJ, Paik YK, Leren TP, Walker DW, Howlett GJ, Taylor JM, Characterization of a human apolipoprotein E gene enhancer element and its associated protein factors, J Biol Chem 265 (1990) 9496–9504. [PubMed] [Google Scholar]
  • [13].Chen XF, Zhang YW, Xu H, Bu G, Transcriptional regulation and its misregulation in Alzheimer's disease, Mol Brain 6 (2013) 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Christensen A, Pike CJ, APOE genotype affects metabolic and Alzheimer-related outcomes induced by Western diet in female EFAD mice, FASEB J (2018) fj201801756R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Cline EN, Bicca MA, Viola KL, Klein WL, The Amyloid-beta Oligomer Hypothesis: Beginning of the Third Decade, J Alzheimers Dis 64 (2018) S567–S610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Corder 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 261 (1993) 921–923. [DOI] [PubMed] [Google Scholar]
  • [17].Elliott DA, Weickert CS, Garner B, Apolipoproteins in the brain: implications for neurological and psychiatric disorders, Clin Lipidol 51 (2010) 555–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Elshourbagy NA, Liao WS, Mahley RW, Taylor JM, Apolipoprotein E mRNA is abundant in the brain and adrenals, as well as in the liver, and is present in other peripheral tissues of rats and marmosets, Proc. Natl. Acad. Sci. USA 82 (1985) 203–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, Myers RH, Pericak-Vance MA, Risch N, van Duijn CM, Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium, JAMA 278 (1997) 1349–1356. [PubMed] [Google Scholar]
  • [20].Ferreira ST, Lourenco MV, Oliveira MM, De Felice FG, Soluble amyloid-beta oligomers as synaptotoxins leading to cognitive impairment in Alzheimer's disease, Front Cell Neurosci 9 (2015) 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Games D, Adams D, Alessandrini R, Barbour R, Berthelette P, Blackwell C, Carr T, Clemens J, Donaldson T, Gillespie F, Guido T, Hagopian S, Johnson-Wood K, Khan K, Lee M, Leibowitz P, Lieberburg I, Little S, Masliah E, McConlogue L, Montoya-Zavala, Mucke L, Paganini L, Penniman E, Power M, Schenk D, Seubert P, Snyder B, Soriano F, Tan H, Vitale J, Wadsworth S, Wolozin B, Zhao J, Alzheimer-type neuropathology in transgenic mice overexpressing V717F ß-amyloid precursor protein, Nature 373 (1995) 523–527. [DOI] [PubMed] [Google Scholar]
  • [22].Grehan S, Tse E, Taylor JM, Two distal downstream enhancers direct expression of the human apolipoprotein E gene to astrocytes in the brain, J Neurosci 21 (2001) 812–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Hayden EY, Conovaloff JL, Mason A, Bitan G, Teplow DB, Preparation of pure populations of covalently stabilized amyloid beta-protein oligomers of specific sizes, Anal Biochem 518 (2017) 78–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Hoffer MJ, Hofker MH, van Eck MM, Havekes LM, Frants RR, Evolutionary conservation of the mouse apolipoprotein e-c1-c2 gene cluster: structure and genetic variability in inbred mice, Genomics 15 (1993) 62–67. [DOI] [PubMed] [Google Scholar]
  • [25].Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G, Correlative Memory Deficits, Aβ Elevation, and Amyloid Plaques in Transgenic Mice, Science 274 (1996) 99–103. [DOI] [PubMed] [Google Scholar]
  • [26].Hudry E, Dashkoff J, Roe AD, Takeda S, Koffie RM, Hashimoto T, Scheel M, Spires-Jones T, Arbel-Ornath M, Betensky R, Davidson BL, Hyman BT, Gene transfer of human apoe isoforms results in differential modulation of amyloid deposition and neurotoxicity in mouse brain, Sci Transl Med 5 (2013) 212ra161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, Pickering-Brown S, Chakraverty S, Isaacs A, Grover A, Hackett J, Adamson J, Lincoln S, Dickson D, Davies P, Petersen RC, Stevens M, de Graaff E, Wauters E, van Baren J, Hillebrand M, Joosse M, Kwon JM, Nowotny P, Che LK, Norton J, Morris JC, Reed LA, Trojanowski J, Basun H, Lannfelt L, Neystat M, Fahn S, Dark F, Tannenberg T, Dodd PR, Hayward N, Kwok JB, Schofield PR, Andreadis A, Snowden J, Craufurd D, Neary D, Owen F, Oostra BA, Hardy J, Goate A, van Swieten J, Mann D, Lynch T, Heutink P, Association of missense and 5'-splice-site mutations in tau with the inherited dementia FTDP-17, Nature 393 (1998) 702–705. [DOI] [PubMed] [Google Scholar]
  • [28].Huynh TV, Liao F, Francis CM, Robinson GO, Serrano JR, Jiang H, Roh J, Finn MB, Sullivan PM, Esparza TJ, Stewart FR, Mahan TE, Ulrich JD, Cole T, Holtzman DM, Age-Dependent Effects of apoE Reduction Using Antisense Oligonucleotides in a Model of beta-amyloidosis, Neuron 96 (2017) 1013–1023 e1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Jankowsky JL, Slunt HH, Ratovitski T, Jenkins NA, Copeland NG, Borchelt DR, Co-expression of multiple transgenes in mouse CNS: a comparison of strategies, Biomolecular engineering 17 (2001) 157–165. [DOI] [PubMed] [Google Scholar]
  • [30].Jankowsky JL, Zheng H, Practical considerations for choosing a mouse model of Alzheimer's disease, Mol Neurodegener 12 (2017) 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Klein WL, Stine WB Jr., Teplow DB, Small assemblies of unmodified amyloid beta-protein are the proximate neurotoxin in Alzheimer's disease, Neurobiol Aging 25 (2004) 569–580. [DOI] [PubMed] [Google Scholar]
  • [32].Klyubin I, Cullen WK, Hu NW, Rowan MJ, Alzheimer's disease Abeta assemblies mediating rapid disruption of synaptic plasticity and memory, Mol Brain 5 (2012) 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Kunzler J, Youmans KL, Yu C, Ladu MJ, Tai LM, APOE modulates the effect of estrogen therapy on Abeta accumulation EFAD-Tg mice, Neurosci Lett 560 (2014) 131–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].LaDu MJ, Gilligan SM, Lukens JR, Cabana VG, Reardon CA, Van Eldik LJ, Holtzman DM, Nascent astrocyte particles differ from lipoproteins in CSF, J Neurochem 70 (1998) 2070–2081. [DOI] [PubMed] [Google Scholar]
  • [35].Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, Tontonoz P, LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes, Proc Natl Acad Sci U S A 98 (2001) 507–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Larson ME, Lesne SE, Soluble Abeta oligomer production and toxicity, J Neurochem 120 Suppl 1 (2012) 125–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Lauer SJ, Walker D, Elshourbagy NA, Reardon CA, Levy-Wilson B, Taylor JM, Two copies of the human apolipoprotein C-I gene are linked closely to the apolipoprotein E gene, J Biol Chem 263 (1988) 7277–7286. [PubMed] [Google Scholar]
  • [38].Liao F, Hori Y, Hudry E, Bauer AQ, Jiang H, Mahan TE, Lefton KB, Zhang TJ, Dearborn JT, Kim J, Culver JP, Betensky R, Wozniak DF, Hyman BT, Holtzman DM, Anti-ApoE antibody given after plaque onset decreases Abeta accumulation and improves brain function in a mouse model of Abeta amyloidosis, J Neurosci 34 (2014) 7281–7292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Liao F, Li A, Xiong M, Bien-Ly N, Jiang H, Zhang Y, Finn MB, Hoyle R, Keyser J, Lefton KB, Robinson GO, Serrano JR, Silverman AP, Guo JL, Getz J, Henne K, Leyns CE, Gallardo G, Ulrich JD, Sullivan PM, Lerner EP, Hudry E, Sweeney ZK, Dennis MS, Hyman BT, Watts RJ, Holtzman DM, Targeting of nonlipidated, aggregated apoE with antibodies inhibits amyloid accumulation, J Clin Invest 128 (2018) 2144–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Liao F, Zhang TJ, Jiang H, Lefton KB, Robinson GO, Vassar R, Sullivan PM, Holtzman DM, Murine versus human apolipoprotein E4: differential facilitation of and co-localization in cerebral amyloid angiopathy and amyloid plaques in APP transgenic mouse models, Acta Neuropathol Commun 3 (2015) 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Liu CC, Zhao N, Fu Y, Wang N, Linares C, Tsai CW, Bu G, ApoE4 Accelerates Early Seeding of Amyloid Pathology, Neuron 96 (2017) 1024–1032 e1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Liu DS, Pan XD, Zhang J, Shen H, Collins NC, Cole AM, Koster KP, Ben Aissa M, Dai XM, Zhou M, Tai LM, Zhu YG, LaDu M, Chen XC, APOE4 enhances age-dependent decline in cognitive function by down-regulating an NMDA receptor pathway in EFAD-Tg mice, Mol Neurodegener 10 (2015) 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Luo J, Lee SH, VandeVrede L, Qin Z, Ben Aissa M, Larson J, Teich AF, Arancio O, D'Souza Y, Elharram A, Koster K, Tai LM, LaDu MJ, Bennett BM, Thatcher GR, A multifunctional therapeutic approach to disease modification in multiple familial mouse models and a novel sporadic model of Alzheimer's disease, Mol Neurodegener 11 (2016) 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Mahley RW, Weisgraber KH, Huang Y, Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, including Alzheimer's disease, Proc Natl Acad Sci U S A 103 (2006) 5644–5651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Mak AC, Pullinger CR, Tang LF, Wong JS, Deo RC, Schwarz JM, Gugliucci A, Movsesyan I, Ishida BY, Chu C, Poon A, Kim P, Stock EO, Schaefer EJ, Asztalos BF, Castellano JM, Wyss-Coray T, Duncan JL, Miller BL, Kane JP, Kwok PY, Malloy MJ, Effects of the Absence of Apolipoprotein E on Lipoproteins, Neurocognitive Function, and Retinal Function, JAMA Neurol (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Mak PA, Laffitte BA, Desrumaux C, Joseph SB, Curtiss LK, Mangelsdorf DJ, Tontonoz P, Edwards PA, Regulated expression of the apolipoprotein E/C-I/C-IV/C-II gene cluster in murine and human macrophages. A critical role for nuclear liver X receptors alpha and beta, J Biol Chem 277 (2002) 31900–31908. [DOI] [PubMed] [Google Scholar]
  • [47].Maloney B, Ge YW, Alley GM, Lahiri DK, Important differences between human and mouse APOE gene promoters: limitation of mouse APOE model in studying Alzheimer's disease, J Neurochem 103 (2007) 1237–1257. [DOI] [PubMed] [Google Scholar]
  • [48].Maloney B, Ge YW, Petersen RC, Hardy J, Rogers JT, Perez-Tur J, Lahiri DK, Functional characterization of three single-nucleotide polymorphisms present in the human APOE promoter sequence: Differential effects in neuronal cells and on DNA-protein interactions, Am J Med Genet B Neuropsychiatr Genet 153B (2010) 185–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Mandrekar-Colucci S, Landreth GE, Nuclear receptors as therapeutic targets for Alzheimer's disease, Expert opinion on therapeutic targets 15 (2011) 1085–1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Marottoli FM, Katsumata Y, Koster KP, Thomas R, Fardo DW, Tai LM, Peripheral Inflammation, Apolipoprotein E4, and Amyloid-beta Interact to Induce Cognitive and Cerebrovascular Dysfunction, ASN Neuro 9 (2017) 1759091417719201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Moser VA, Pike CJ, Obesity Accelerates Alzheimer-Related Pathology in APOE4 but not APOE3 Mice, eNeuro 4 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Mucke L, Masliah E, Yu GQ, Mallory M, Rockenstein EM, Tatsuno G, Hu K, Kholodenko D, Johnson-Wood K, McConlogue L, High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation, J Neurosci 20 (2000) 4050–4058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Nam KN, Mounier A, Wolfe CM, Fitz NF, Carter AY, Castranio EL, Kamboh HI, Reeves VL, Wang J, Han X, Schug J, Lefterov I, Koldamova R, Effect of high fat diet on phenotype, brain transcriptome and lipidome in Alzheimer's model mice, Sci Rep 7 (2017) 4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Nam KN, Wolfe CM, Fitz NF, Letronne F, Castranio EL, Mounier A, Schug J, Lefterov I, Koldamova R, Integrated approach reveals diet, APOE genotype and sex affect immune response in APP mice, Biochim Biophys Acta Mol Basis Dis 1864 (2018) 152–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar R, Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer's disease mutations: potential factors in amyloid plaque formation, J Neurosci 26 (2006) 10129–10140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Oddo S, Caccamo A, Kitazawa M, Tseng BP, LaFerla FM, Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer's disease, Neurobiol Aging 24 (2003) 1063–1070. [DOI] [PubMed] [Google Scholar]
  • [57].Paik YK, Chang DJ, Reardon CA, Davies GE, Mahley RW, Taylor JM, Nucleotide sequence and structure of the human apolipoprotein E gene, Proc Natl Acad Sci U S A 82 (1985) 3445–3449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Paik YK, Chang DJ, Reardon CA, Walker MD, Taxman E, Taylor JM, Identification and characterization of transcriptional regulatory regions associated with expression of the human apolipoprotein E gene, J Biol Chem 263 (1988) 13340–13349. [PubMed] [Google Scholar]
  • [59].Pike CJ, Sex and the development of Alzheimer's disease, J Neurosci Res 95 (2017) 671–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Poorkaj P, Bird TD, Wijsman E, Nemens E, Garruto RM, Anderson L, Andreadis A, Wiederholt WC, Raskind M, Schellenberg GD, Tau is a candidate gene for chromosome 17 frontotemporal dementia, Ann Neurol 43 (1998) 815–825. [DOI] [PubMed] [Google Scholar]
  • [61].Raber J, Wong D, Buttini M, Orth M, Bellosta S, Pitas RE, Mahley RW, Mucke L, 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 (1998) 10914–10919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Radde R, Bolmont T, Kaeser SA, Coomaraswamy J, Lindau D, Stoltze L, Calhoun ME, Jaggi F, Wolburg H, Gengler S, Haass C, Ghetti B, Czech C, Holscher C, Mathews PM, Jucker M, Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology, EMBO Rep 7 (2006) 940–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Rebeck GW, Kindy M, LaDu MJ, Apolipoprotein E and Alzheimer's disease: the protective effects of ApoE2 and E3, J Alzheimers Dis 4 (2002) 145–154. [DOI] [PubMed] [Google Scholar]
  • [64].Roychaudhuri R, Huynh TV, Whitaker TR, Hodara E, Condron MM, Teplow DB, A Critical Role of Ser26 Hydrogen Bonding in Abeta42 Assembly and Toxicity, Biochemistry 56 (2017) 6321–6324. [DOI] [PubMed] [Google Scholar]
  • [65].Roychaudhuri R, Zheng X, Lomakin A, Maiti P, Condron MM, Benedek GB, Bitan G, Bowers MT, Teplow DB, Role of Species-Specific Primary Structure Differences in Abeta42 Assembly and Neurotoxicity, ACS Chem Neurosci 6 (2015) 1941–1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Shi Y, Yamada K, Liddelow SA, Smith ST, Zhao L, Luo W, Tsai RM, Spina S, Grinberg LT, Rojas JC, Gallardo G, Wang K, Roh J, Robinson G, Finn MB, Jiang H, Sullivan PM, Baufeld C, Wood MW, Sutphen C, McCue L, Xiong C, Del-Aguila JL, Morris JC, Cruchaga C, Alzheimer's Disease Neuroimaging I, Fagan AM, Miller BL, Boxer AL, Seeley WW, Butovsky O, Barres BA, Paul SM, Holtzman DM, ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy, Nature 549 (2017) 523–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Shih SJ, Allan C, Grehan S, Tse E, Moran C, Taylor JM, Duplicated downstream enhancers control expression of the human apolipoprotein E gene in macrophages and adipose tissue, J Biol Chem 275 (2000) 31567–31572. [DOI] [PubMed] [Google Scholar]
  • [68].Simonet WS, Bucay N, Lauer SJ, Pitas RE, Weisgraber KH, Taylor JM, Downstream regulatory elements stimulate expression of the human apolipoprotein E gene in the liver and suppress expression in the kidney of transgenic mice, Trans Assoc Am Physicians 103 (1990) 119–128. [PubMed] [Google Scholar]
  • [69].Simonet WS, Bucay N, Lauer SJ, Taylor JM, A far-downstream hepatocyte-specific control region directs expression of the linked human apolipoprotein E and C-I genes in transgenic mice, J Biol Chem 268 (1993) 8221–8229. [PubMed] [Google Scholar]
  • [70].Simonet WS, Bucay N, Lauer SJ, Wirak DO, Stevens ME, Weisgraber KH, Pitas RE, Taylor JM, In the absence of a downstream element, the apolipoprotein E gene is expressed at high levels in kidneys of transgenic mice, J Biol Chem 265 (1990) 10809–10812. [PubMed] [Google Scholar]
  • [71].Simonet WS, Bucay N, Pitas RE, Lauer SJ, Taylor JM, Multiple tissue-specific elements control the apolipoprotein E/C-I gene locus in transgenic mice, J Biol Chem 266 (1991) 8651–8654. [PubMed] [Google Scholar]
  • [72].Spillantini MG, Murrell JR, Goedert M, Farlow MR, Klug A, Ghetti B, Mutation in the tau gene in familial multiple system tauopathy with presenile dementia, Proc Natl Acad Sci U S A 95 (1998) 7737–7741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Sullivan PM, Mezdour H, Aratani Y, Knouff C, Najib J, Reddick RL, Quarfordt SH, Maeda N, Targeted replacement of the mouse apolipoprotein E gene with the common human APOE3 allele enhances diet-induced hypercholesterolemia and atherosclerosis, J Biol Chem 272 (1997) 17972–17980. [DOI] [PubMed] [Google Scholar]
  • [74].Sun Y, Wu S, Bu G, Onifade MK, Patel SN, LaDu MJ, Fagan AM, Holtzman DM, Glial fibrillary acidic protein-apolipoprotein E (apoE) transgenic mice: astrocyte-specific expression and differing biological effects of astrocyte-secreted apoE3 and apoE4 lipoproteins, J Neurosci 18 (1998) 3261–3272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ, EFAD Transgenic Mice as a Human APOE Relevant Preclinical Model of Alzheimer's Disease, J Lipid Res (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Tai LM, Youmans KL, Jungbauer L, Yu C, Ladu MJ, Introducing Human APOE into Abeta Transgenic Mouse Models, Int J Alzheimers Dis 2011 (2011) 810981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Terwel D, Lasrado R, Snauwaert J, Vandeweert E, Van Haesendonck C, Borghgraef P, Van Leuven F, Changed conformation of mutant Tau-P301L underlies the moribund tauopathy, absent in progressive, nonlethal axonopathy of Tau-4R/2N transgenic mice, J Biol Chem 280 (2005) 3963–3973. [DOI] [PubMed] [Google Scholar]
  • [78].Thomas R, Morris AWJ, Tai LM, Epidermal growth factor prevents APOE4-induced cognitive and cerebrovascular deficits in female mice, Heliyon 3 (2017) e00319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Trusca VG, Fuior EV, Florea IC, Kardassis D, Simionescu M, Gafencu AV, Macrophage-specific up-regulation of apolipoprotein E gene expression by STAT1 is achieved via long range genomic interactions, J Biol Chem 286 (2011) 13891–13904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Tu S, Okamoto S, Lipton SA, Xu H, Oligomeric Abeta-induced synaptic dysfunction in Alzheimer's disease, Mol Neurodegener 9 (2014) 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Ulrich JD, Ulland TK, Mahan TE, Nystrom S, Nilsson KP, Song WM, Zhou Y, Reinartz M, Choi S, Jiang H, Stewart FR, Anderson E, Wang Y, Colonna M, Holtzman DM, ApoE facilitates the microglial response to amyloid plaque pathology, J Exp Med 215 (2018) 1047–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Van Cauwenberghe C, Van Broeckhoven C, Sleegers K, The genetic landscape of Alzheimer disease: clinical implications and perspectives, Genetics in medicine : official journal of the American College of Medical Genetics 18 (2016) 421–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].van Eck MM, Hoffer MJ, Havekes LM, Frants RR, Hofker MH, The apolipoprotein C2-linked (Acl) gene: a new gene within the mouse apolipoprotein e-c1-c2 gene cluster, Genomics 21 (1994) 110–115. [DOI] [PubMed] [Google Scholar]
  • [84].Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, Maeda J, Suhara T, Trojanowski JQ, Lee VM, Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model, Neuron 53 (2007) 337–351. [DOI] [PubMed] [Google Scholar]
  • [85].Youmans KL, Tai LM, Nwabuisi-Heath E, Jungbauer L, Kanekiyo T, Gan M, Kim J, Eimer WA, Estus S, Rebeck GW, Weeber EJ, Bu G, Yu C, Ladu MJ, APOE4-specific Changes in Abeta Accumulation in a New Transgenic Mouse Model of Alzheimer Disease, J Biol Chem 287 (2012) 41774–41786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Zannis VI, Kan HY, Kritis A, Zanni E, Kardassis D, Transcriptional regulation of the human apolipoprotein genes, Front Biosci 6 (2001) D456–504. [DOI] [PubMed] [Google Scholar]
  • [87].Zhang SH, Reddick RL, Piedrahita JA, Maeda N, Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E, Science (New York, N.Y 258 (1992) 468–471. [DOI] [PubMed] [Google Scholar]
  • [88].Zhao N, Liu CC, Van Ingelgom AJ, Linares C, Kurti A, Knight JA, Heckman MG, Diehl NN, Shinohara M, Martens YA, Attrebi ON, Petrucelli L, Fryer JD, Wszolek ZK, Graff-Radford NR, Caselli RJ, Sanchez-Contreras MY, Rademakers R, Murray ME, Koga S, Dickson DW, Ross OA, Bu G, APOE epsilon2 is associated with increased tau pathology in primary tauopathy, Nat Commun 9 (2018) 4388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Zhou M, Huang T, Collins N, Zhang J, Shen H, Dai X, Xiao N, Wu X, Wei Z, York J, Lin L, Zhu Y, LaDu MJ, Chen X, APOE4 induces site-specific tau phosphorylation through calpain-CDK5 signaling pathway in EFAD-Tg mice, Curr Alzheimer Res (2016). [DOI] [PubMed] [Google Scholar]

RESOURCES