Skip to main content
International Journal of Alzheimer's Disease logoLink to International Journal of Alzheimer's Disease
. 2011 Oct 19;2011:810981. doi: 10.4061/2011/810981

Introducing Human APOE into Aβ Transgenic Mouse Models

Leon M Tai 1, Katherine L Youmans 1, Lisa Jungbauer 1, 2,2, Chunjiang Yu 1, Mary Jo LaDu 1,*
PMCID: PMC3199079  PMID: 22028984

Abstract

Apolipoprotein E (apoE) and apoE/amyloid-β (Aβ) transgenic (Tg) mouse models are critical to understanding apoE-isoform effects on Alzheimer's disease risk. Compared to wild type, apoE−/− mice exhibit neuronal deficits, similar to apoE4-Tg compared to apoE3-Tg mice, providing a model for Aβ-independent apoE effects on neurodegeneration. To determine the effects of apoE on Aβ-induced neuropathology, apoE−/− mice were crossed with Aβ-Tg mice, resulting in a significant delay in plaque deposition. Surprisingly, crossing human-apoE-Tg mice with apoE−/−/Aβ-Tg mice further delayed plaque deposition, which eventually developed in apoE4/Aβ-Tg mice prior to apoE3/Aβ-Tg. One approach to address hAPOE-induced temporal delay in Aβ pathology is an additional insult, like head injury. Another is crossing human-apoE-Tg mice with Aβ-Tg mice that have rapid-onset Aβ pathology. For example, because 5xFAD mice develop plaques by 2 months, the prediction is that human-apoE/5xFAD-Tg mice develop plaques around 6 months and 12 months before other human-apoE/Aβ-Tg mice. Thus, tractable models for human-apoE/Aβ-Tg mice continue to evolve.

1. Introduction

Alzheimer's disease (AD) is the most common form of dementia and represents a serious economic and social burden worldwide. The familial form of AD (FAD) is caused by autosomal dominant mutations that increase levels of the 42 amino acid isoform of the amyloid-beta 42 (Aβ42) peptide [1, 2]. The primary genetic risk factor for AD is inheritance of the APOE4 gene for apolipoprotein E (apoE), compared to APOE3, with APOE2 reducing risk [35]. The mechanism(s) by which apoE and Aβ affect pathogenesis of the disease is unclear (reviewed [35]). However, evidence suggests that apoE may isoform-specifically modulate Aβ-induced neurotoxicity [4, 5]. To address potential mechanisms in vivo, several transgenic (Tg) mouse models have been developed to assess the structural and functional interactions between apoE and Aβ. However, each of these models has potential drawbacks that affect the interpretation and physiological relevance of the results. This paper will summarize the current models resulting from the introduction of human-APOE into Aβ-Tg mice. The continued development and characterization of both apoE and apoE/Aβ-Tg mouse models is critical to understanding the apoE-isoform effects on AD pathology.

1.1. Amyloid-β

Traditional diagnosis of AD is based on pathology that includes extracellular amyloid plaques, composed primarily of Aβ42, intraneuronal neurofibrillary tangles, hyperphosphorylated tau, neuroinflammation, and neuronal cell loss. Aβ is a 39–43 amino acid (4 kDa) peptide produced via sequential proteolysis of amyloid precursor protein (APP) by β-secretase/BACE followed by γ-secretase (composed of presenilins [PS] 1 and 2), to produce Aβ peptides primarily 40 and 42 amino acids in length. Genetic and experimental evidence indicates that Aβ42 is the cause of AD pathogenesis [1, 2]: (1) FAD, although only 3–5% of all AD cases is caused by autosomal dominant mutations in APP, PS1, or PS2 that increase levels of Aβ42 or the Aβ42 : 40 ratio; (2) Down syndrome is caused by trisomy of chromosome 21 (the location of the APP gene), and is characterized by plaque deposition and dementia by the age 40; (3) Aβ42 is neurotoxic in vitro and in vivo. Aβ, particularly the more toxic Aβ42, aggregates to form a variety of higher-order assemblies including oligomers, protofibrils, fibrils, and amyloid plaques [6]. Amyloid plaques themselves exist in different conformations including compact plaques (composed of a dense Thioflavin-S- (ThioS-) positive core), neuritic plaques (identified as ThioS-positive plaques surrounded by a ring of dystrophic neurites), and diffuse plaques (characterized by amorphous wisps of amyloid that lack a central core and are not neurotoxic) [710]. While total plaque burden does not directly correlate with dementia [11], it is an indication of increased Aβ42 levels, and compact or dense core plaques may be disease relevant [7, 12].

1.2. ApoE

ApoE is the only apolipoprotein synthesized within the blood-brain barrier (BBB) and is the primary apolipoprotein associated with lipoprotein particles in the central nervous system (CNS), as peripheral apoE is unable to cross the BBB or blood-cerebrospinal fluid barrier. While the majority of apoE in the CNS is secreted by glial cells, particularly astrocytes, neuronal production of apoE has been observed under specific pathological conditions [13]. CNS lipoproteins are critical for lipid homeostasis, particularly as cholesterol and phospholipids are required for neuronal growth, repair, and synaptogenesis (reviewed [35]). In humans, three apoE polymorphic alleles exist (APOE2, APOE3, and APOE4) which encode three protein isoforms (apoE2, apoE3, and apoE4). Although human-apoE (h-apoE) is a 299 amino acid protein, the three apoE-isoforms differ by a single amino acid substitution at residues 112 or 158: apoE2 contains Cys112,158, apoE3 contains Cys112Arg118, and apoE4 contains Arg112,158 [5]. In the general population, APOE3 is the most common allele (77%), followed by APOE4 (15%) and APOE2 (8%). In contrast, 40% of AD patients express at least one APOE4 allele. Compared to APOE3, inheritance of one or two copies of the APOE4 allele increases the risk for developing AD by 4- and 12-fold, whereas APOE2 decreases risk by 2- and 4-fold [5, 14]. Amyloid plaque deposition is greatest in AD patients with an APOE4 allele(s) [14]. In addition to AD, APOE4 is a risk factor for cerebral amyloid angiopathy (CAA; amyloid deposition in blood vessels) and impairs recovery from cerebral insults such as stroke, cerebral hemorrhage, and brain injury [4]. While apoE knock-out mice (apoE−/−) and h-apoE-Tg models demonstrate that apoE affects neuronal viability independent of Aβ-induced neurotoxicity, the focus of this paper is on the synergism between the h-apoE isoforms and Aβ on neuropathology. To address the latter in vivo, apoE/Aβ-Tg models were subsequently developed to specifically address the isoform-specific effects of h-apoE on Aβ deposition.

2. Transgenic Mice Expressing Human ApoE (Table 1)

Table 1.

ApoE transgenic models. Summary of the effects induced by deletion of mouse apoE (apoE−/−) or the introduction of h-apoE isoforms on markers of neuronal degeneration and behavior. Key: HC: hippocampus; CX: cortex; DG: dentate gyrus; ChAT: choline acetyl transferase; ICH: intracerebral hemorrhage; LTP: long-term potentiation; TR: targeted replacement (knock-in); F: female; M: male; m: month.

ApoE model [ApoE] Neurodegeneration Behavioral phenotype Gender effects on behavior References
ApoE−/− NA Spine density and dendritic length:
WT > KO (HC/DG, 12 m)
Water maze:
WT > KO (6 m, F and M)
Water maze:
M > F (6 m)
[1521]
ChAT:
WT > KO (HC, 6 m, M)
Anxiety vertical exploratory:
KO > WT (6 m, F)

GFAP-apoE Matched Spine length and density:
E3 > E4 (HC, 12 m)
Water maze:
E3 > E4 (6 m, F)
Water maze:
M > F (apoE4 only)
[17, 2224]
Radial arm maze:
E3 > E4 (11–14 m, M)

NSE-apoE Matched Synaptophysin:
E3 > E4 (HC, 5 m)
Water maze:
E3 > E4 (6 m, F)
Water maze: M > F (apoE4 only, 6 m)
ChAT activity:
E3 > E4 = KO (12 m):
[2527]
Tau phosphorylation:
E4 > E3 (9 m)
Kainic acid induced neurodegeneration
Synaptophysin and MAP-2:
E3 > E4 = KO (CX, 5 m)
[25]

ApoE-TR E2 ≥ E3 > E4 Spine density and length:
E3 > E4 (1, 3 and 12 m)
Water maze/Active Y maze avoidance:
E3 > E4 (15 m, F)
Water maze:
M > F (6 and 13 m)
Dendritic length and node number:
E3 > E4 (1 m)
Anxiety elevated platform:
E4 > E3 (6–8 m, F)
EPSP interval:
E4/E4 > E2/E4 > E3/E3 (1 m)
[18, 2835]
LTP:
E3 > E4 (2–4 m)
ICH:
E4 > E3 (15 m+)
Vascular amyloid:
E4 > E3 (15 m+)

Several approaches have been used to make Tg mouse models to assess the function of apoE. ApoE−/− Tg-mice were initially used to help understand the role of apoE in the brain [15, 16] although the homology between mouse (m-) and h-apoE is 70%, and mice express only a single isoform, comparable to apoE4 at residues 112 and 158 [54]. ApoE−/− mice have been used as the background for several h-APOE-Tg mouse lines. Heterologous promoters have been used to drive the expression of h-apoE in glia or neurons. Examples include glial fibrillar acidic protein (GFAP; glial) [22, 5557], transferrin (neuronal) [58], platelet-derived growth factor (PDGF; neuronal) [59], neuron-specific enolase (NSE; neuronal) [15], and thymocyte differentiation antigen 1 (Thy1; neuronal) [59]. However, these models have limitations inherent in the use of a heterologous promoter and specific to apoE: (1) the expression of protein by a heterologous promoter is not regulated as it would be by the endogenous promoter; (2) the inserted copy number of the transgene cannot be regulated by a traditional Tg mouse approach; (3) while the cell-specific expression of apoE in the brain is controversial [6063], the majority of evidence suggests that glia, not neurons, are the primary cell type to express apoE [13, 6467]; (4) by using the m-apoE−/− background and inducing apoE expression via CNS-specific promoters, lack of peripheral apoE becomes a variable of potential importance when interpreting results from these mice; (5) evidence suggests that in both humans and apoE-TR mice, apoE4 levels are significantly lower than apoE2 or apoE3 [6873]. Knock-in or targeted-replacement (TR) mice were developed that express h-apoE under the control of the endogenous mouse promoter and provide an alternative to heterologous expression of h-apoE. In apoE-TR mice, the coding domain for each of the h-APOE isoforms replaces the coding domain for m-APOE. Thus, in apoE-TR mice, glial cells express h-apoE in a native conformation at physiologically regulated levels, and in the same temporal and spatial pattern as endogenous m-apoE. Thus, the interpretation of apoE isoform-specific results is determined by the nature of the apoE-Tg mouse model.

As discussed, a number of apoE-Tg mouse models have been developed with apoE expression under the control of different promoters [74]. The general phenotypes of apoE−/− mice and three examples of the most widely studied apoE-Tg mice (GFAP-apoE, NSE-apoE, and apoE-TR) are briefly discussed (Table 1).

2.1. ApoE −/−

Compared to wild type, apoE−/− mice have decreased excitatory transmission, spine density, and dendritic length [1618]. These changes may underlie behavioral deficits, as apoE−/− mice are cognitively impaired [15, 19, 20]. However, lack of peripheral apoE can have profound effects on plasma lipid homeostasis, potentially leading to a number of confounding variables, including metabolic disease and increased risk for cardiovascular disease, compromising the ability to compensate for oxidative stress or inflammation, deficits that can effect the vasculature of the brain. In addition, the relevance of these mice is unclear as there are no apoE−/− humans. ApoE−/− provides the background for a number of the h-apoE-Tg mice.

2.2. GFAP-apoE Mice

GFAP is an intermediate filament protein, expressed at high levels by glia in the CNS. To target apoE expression to glia, mice expressing h-apoE under the control of the GFAP promoter were crossed with apoE−/− mice [17, 22, 23]. At the cellular level, GFAP-apoE4 mice show increased CA1 cellular atrophy and decreased spine density compared to GFAP-apoE3 mice [75]. In addition, compared to GFAP-apoE3 mice, GFAP-apoE4 mice demonstrate impaired cognition and increased anxiety; common symptoms in AD patients [17, 23, 24]. Interestingly, cognitive impairment is evident at an earlier age in female compared to male GFAP-apoE4 mice [23, 24]. A limitation of this model is that the effect of apoE isoform on neuroinflammatory responses cannot be interpreted, as apoE is expressed under the control of a promoter that is induced by neuroinflammation.

2.3. Neuronal apoE Expression

Neuronal apoE expression remains controversial, although expression has been identified under conditions of stress [25]. To investigate the role of neuronal expression, apoE-Tg mice expressing h-apoE under the control of the NSE promoter were crossed with apoE−/− mice [2527]. Kainic acid was used to induce apoE expression in these mice and resulted in the protection of NSE-apoE3 mice from age- and kainic acid-induced presynaptic and dendritic degeneration compared to NSE-apoE4 mice [25]. In addition, female NSE-apoE4 mice demonstrate impaired cognition compared to female NSE-apoE3 mice [25]. In contrast, male NSE-apoE4 mice do not exhibit cognitive deficits [15]. Thus, the effect of apoE4 on cognition may be modulated by gender. This is similar to the early development of cognitive deficits in GFAP-apoE4.

2.4. ApoE-TR

To investigate the function(s) of apoE in a more physiologically relevant apoE-Tg model, apoE-TR mice were developed to express each h-apoE isoform under the control of the endogenous m-APOE promoter [2835]. Although apoE protein levels in brain homogenates of these mice were initially described as being similar for each h-apoE isoform [32], subsequent analysis demonstrated that apoE4 levels are significantly lower in plasma, CSF, and brain homogenates than apoE2 and apoE3 [28, 30], similar to the levels of apoE isoforms in humans [6873]. Compared to apoE3-TR mice, apoE4-TR mice demonstrate decreased spine density and dendrite length, reduced excitatory transmission, and long-term potentiation (Table 1). Again, similar to the cognitive deficits observed in female GFAP-apoE4 mice and NSE-apoE4 mice, female apoE4-TR mice are cognitively impaired compared to female apoE3-TR mice [31, 32]. However, male apoE3-TR and apoE4-TR mice are not significantly different. Interestingly, both apoE3- and apoE4-TR females are cognitively impaired when compared to apoE-isoform-matched males.

3. ApoE in Aβ Transgenic Models (Table 2)

Table 2.

APOE in Aβ transgenic models. Genetic deletion of mouse apoE (apoE−/−) from Aβ-Tg mice delays amyloid deposition, which is further delayed by the introduction of h-APOE into Aβ-Tg mice. *LaDu Lab unpublished observations.

Aβ pathology (age of onset)
Transgenic line 2–4 months 4–8 months 8–10 months 11–13 months ≥13 months Total Aβ levels
PDAPP+/− [3639] Diffuse Aβ staining:
hippocampus
cortex
ThioS (moderate):
hippocampus
cortex
ThioS (heavy):
hippocampus
cortex
ThioS (heavier):
hippocampus
cortex
PDAPP+/− × apoE+/− [40] ThioS (low):
hippocampus
cortex
ThioS (moderate):
hippocampus
cortex
PDAPP+/− × apoE−/−[41] Diffuse Aβ staining:
hippocampus
Thios (low):
hippocampus
cortex
6, 12, 15, 18, and 21 months:
PDAPP+/− > PDAPP+/−/apoE−/−
PDAPP+/− × GFAP-apoE+/− [42] Thios (moderate):
hippocampus
E4 > E3 > E2
6, 12, 15, 18, and 21 months:
E4 > E3
PDAPP+/+ × apoE-TR [43] ThioS low:
hippocampus
E4 > E3 = E2
cortex
E4 > E3 > E2
3 and 12 months:
E4 > E3

Tg2576 [42, 4446] Thios (low):
hippocampus
cortex
ThioS (moderate):
hippocampus cortex
ThioS (heavy):
hippocampus cortex
CAA-fibrillar (low):
leptomeningeal and cortical vessels
CAA-fibrillar (heavy):
leptomeningeal and cortical vessels
CAA-fibrillar (heavy):
leptomeningeal and cortical vessels
Tg2576 × apoE−/− [42] Diffuse Aβ staining:
hippocampus
cortex
12 months Tg2576 > Tg2576/apoE−/−
Tg2576 × apoE-TR[47] CAA-fibrillar (moderate):
leptomeningeal vessels
E4 > E3

J9 mice [48] ThioS (moderate):
cortex
hippocampus
J9 X NSE-apoE [26] ThioS:
hippocampus
E4 > E3

5xFAD [49] ThioS (moderate):
hippocampus
cortex
ThioS (heavy):
hippocampus
cortex
*5xFAD × apoE-TR [5053] ThioS (low):
hippocampus
cortex

3.1. Aβ-Tg

Although amyloid plaques are a hallmark of AD, the mechanisms underlying Aβ-induced toxicity remain unknown. To help determine the effects of Aβ and amyloid deposition on neurotoxicity in vivo, Aβ-Tg mouse models have been produced that express human FAD mutations. These models include but are not limited to: PDAPP (APPV717F), Tg2576 (APPK670N, M671L), J9 (PS1M146V,L286V, APPV717F), as well as 5xFAD (APPK670N, M671L,I716V,V717I, PS1M146V,L286V) (reviewed in [2]). These Aβ-Tg mice express either APP mutations alone (PDAPP, Tg2576) or in combination with PS mutations (J9, 5xFAD). Plaque development generally begins at 6 months of age in these mice (with the exception of 5xFAD), and the onset of cognitive deficits is dependent on the model (e.g., PDAPP at 6 months, Tg2576 at 9 months). Because the APPK670N, M671L mutation is linked primarily to CAA, the Tg2576 mice also develop Aβ deposition around blood vessels, particularly leptomeningeal and cortical vessels. However, one limitation of Aβ-Tg models is the relative lack of neuronal loss as observed in AD (reviewed [76]) although there are some exceptions (e.g., 5xFAD [49], APPSLPS1KI, and TBA2 mice [76]).

3.2. ApoE −/−/Aβ-Tg

Initially, the effect of apoE on Aβ deposition was investigated using apoE−/− crossed with Aβ-Tg mice (Table 2). Specifically, apoE−/− mice were crossed with PDAPP [7779] or Tg2576 mice [80]. In both models, the absence of apoE significantly delayed ThioS-positive plaque deposition by 6 months and decreased Aβ levels in the hippocampus and cortex, as measured biochemically from brain homogenates and by Aβ immunoreactivity [42, 77] (Table 2). APOE affects plaque deposition in a gene-dose-dependent manner, as plaque levels were intermediate in apoE+/−/PDAPP compared with apoE+/+/PDAPP and apoE−/−/PDAPP mice [78]. In addition, the Aβ40 CAA present in the Tg2576 mice was absent in the apoE−/−/Tg2576 mice [80].

3.3. GFAP-apoE/Aβ-Tg

The initial studies demonstrating that the lack of m-APOE delayed plaque deposition in Aβ-Tg mice led to the question of what would be the effect of introducing h-APOE into apoE−/−/Aβ-Tg mice (Table 2). To initially address this issue, GFAP-apoE mice was crossed with apoE−/−/PDAPP mice [41, 42]. Surprisingly, the introduction of h-APOE did not result in the expected reduction in the age of onset of Aβ pathology, rather the presence of h-APOE further delayed Aβ deposition. Amyloid accumulation is delayed from 6 to 12 months in apoE+/−/PDAPP mice, and to 15 months in GFAP-apoE+/−/PDAPP mice (Table 2). Once plaque pathology returned, the greatest plaque burden was found in GFAP-apoE4/PDAPP mice, compared with GFAP-apoE2/PDAPP and GFAP-apoE3/PDAPP mice. One potential confounding factor in these mice, as well as the NSE-apoE mice described below, is the equal expression of the h-APOE isoforms, in contrast to human data where inheritance of an APOE4 allele results in lower apoE levels [68, 69, 73].

3.4. Neuronal apoE Expression/Aβ-Tg

To determine the effect of neuronal h-APOE expression on Aβ pathology, NSE-apoE were crossed with J9 mice (PS1M146V,L286V, APPV717F) [81]. Similar to GFAP-apoE/PDAPP-Tg mice, the introduction of h-APOE to J9 mice delayed plaque pathology from 8 to 19 months, with deposition greater in the NSE-apoE4/J9 than NSE-apoE3/J9 [26].

3.5. ApoE-TR/Aβ-Tg

The physiologic advantages of using apoE-TR mice to study the function(s) of apoE in vivo led to the generation of apoE-TR/PDAPP [30] and apoE-TR/Tg2576 mice [47] (Table 2). The resulting data confirmed that h-APOE delayed plaque deposition. In apoE-TR/PDAPP mice, plaque deposition was delayed from 6 months to 18 months of age. An apoE isoform-specific effect on Aβ pathology was also observed, with ThioS staining, Aβ immunoreactivity, and Aβ levels in brain homogenates highest with apoE4 [30]. In Tg2576 mice, plaque deposition initiates at 9 months of age, while in apoE-TR/Tg2576 mice there is minimal plaque staining at 15 months. Interestingly, at 15 months, the isoform effect in these mice is primarily on CAA (E4 > E3), as amyloid deposition in the parenchyma is minimal [47].

3.6. Addressing h-APOE-Induced Delay in Aβ Pathology

The major drawback to the apoE/Aβ-Tg crosses described thus far is the significant h-APOE-induced delay in Aβ pathology. For example, in apoE-TR/PDAPP mice, plaque deposition is not identified until mice are ≥18 months of age (Table 2). This substantial delay precludes timely analyses of apoE isoform-specific effects on early aspects of Aβ pathology. One approach to address this temporal delay is to introduce an additional insult, such as traumatic brain injury (TBI) [82], kainic acid [25], nitric-oxide-synthase-2- (NOS2-) knock-out [83], or by blocking Aβ degradation via neprilysin inhibition [84]. Although no amyloid deposition is present in 12-month-old GFAP-apoE/PDAPP mice, TBI at 9 months leads to amyloid deposition at 12 months [82], which is greater with apoE4 compared to apoE3. Kainic acid decreases synaptophysin and MAP-2 staining in apoE−/− and NSE-apoE Tg mice, with the effect more pronounced with apoE4 than apoE3. Nitric oxide, produced by inducible NOS (encoded for by the NOS2 gene), is an important signaling and redox factor that plays a key role in neuroinflammation and neurodegeneration [83]. NOS2−/− mice have been crossed with multiple Aβ-Tg mouse models, and results demonstrate increased tau phosphorylation and neuronal loss in NOS2−/−/Aβ-Tg mice [83]. In the Tg2576/NOS2−/− mice, Aβ deposition is higher compared to Tg2576 mice. Inhibition of neprilysin, an enzyme that degrades extracellular Aβ, with thiorphan, induces fibrillization and deposition of Aβ and in wild-type mice. Thiorphan treatment of apoE-TR mice leads to aggregation of mouse Aβ 1 week after treatment, with higher Aβ deposition in apoE4-TR compared to apoE3-TR mice [84, 85]. Thus, thiorphan treatment of h-apoE-Aβ-Tg mice represents a potential method of accelerating human Aβ deposition.

An alternative method to address the h-APOE-induced temporal delay in Aβ accumulation is to cross apoE-TR mice with Aβ-Tg mice that have rapid-onset Aβ pathology, such as 5xFAD [49], 3xTR [86, 87], APPSLPS1KI [88], APPPS1 [89], or TgCRND [90]. For example, because 5xFAD mice develop plaques by 2 months, the prediction is that apoE-TR/5xFAD-Tg mice develop plaques around 6 months and 12 months before other human-apoE/Aβ-Tg mice (LaDu lab, unpublished observations and [5053]). The approaches described herein will lead to more tractable apoE/Aβ-Tg-models to assess the apoE isoform-specific effects on Aβ pathology.

4. Concluding Remarks

ApoE is the greatest risk factor for AD. ApoE−/− mice exhibit neuronal and cognitive deficits. Human apoE-Tg mouse models demonstrate that, compared to apoE3, apoE4 increases markers of neurodegeneration and cognitive impairment. Initially, to determine the effect of apoE on Aβ pathology, Aβ-Tg mice were crossed with apoE−/− mice, resulting in a significant delay in plaque deposition. Surprisingly, the introduction of h-APOE to several apoE−/−/Aβ-Tg mouse models further delayed plaque deposition. This temporal delay restricts the usefulness of the current apoE/Aβ-Tg mice for investigating the process of Aβ accumulation and the resulting neurotoxicity. To accelerate Aβ deposition, current apoE/Aβ-Tg models could be treated with an additional insult such as TBI, crossed with other Tg models of neurodegeneration (NOS2−/−), or treated with drugs that decrease Aβ degradation. Alternatively, the development of Aβ pathology could be accelerated by crossing Aβ-Tg models with a rapid onset of Aβ pathology, such as 5xFAD mice with apoE-TR mice. Transgenic models such as these provide tractable models for identifying biomarkers and the efficient initial validation of therapeutic targets.

Acknowledgments

Current LaDu lab funding includes NIH (NIA) P01AG03012801, Alzheimer's Association ZEN-08-99900, UIC CCTS UL1RR029879. The authors would like to acknowledge Steve Estus, Goujun Bu, William Rebeck, and Edwin Weeber for ongoing collaborations.

References

  • 1.Hardy J. The amyloid hypothesis for Alzheimer's disease: a critical reappraisal. Journal of Neurochemistry. 2009;110(4):1129–1134. doi: 10.1111/j.1471-4159.2009.06181.x. [DOI] [PubMed] [Google Scholar]
  • 2.Wisniewski T, Sigurdsson EM. Murine models of Alzheimer’s disease and their use in developing immunotherapies. Biochimica et Biophysica Acta. 2010;1802(10):847–859. doi: 10.1016/j.bbadis.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Takeda M, Martínez R, Kudo T, et al. Apolipoprotein e and central nervous system disorders: reviews of clinical findings. Psychiatry and Clinical Neurosciences. 2010;64(6):592–607. doi: 10.1111/j.1440-1819.2010.02148.x. [DOI] [PubMed] [Google Scholar]
  • 4.Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer's disease and other neurological disorders. The Lancet Neurology. 2011;10(3):241–252. doi: 10.1016/S1474-4422(10)70325-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bu G. Apolipoprotein e and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nature Reviews Neuroscience. 2009;10(5):333–344. doi: 10.1038/nrn2620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hardy J. Alzheimer’s disease: the amyloid cascade hypothesis—an update and reappraisal. Journal of Alzheimer’s Disease. 2006;9(3, supplement):151–153. doi: 10.3233/jad-2006-9s317. [DOI] [PubMed] [Google Scholar]
  • 7.Adlard PA, Vickers JC. Morphologically distinct plaque types differentially affect dendritic structure and organisation in the early and late states of Alzheimer’s disease. Acta Neuropathologica. 2002;103(4):377–383. doi: 10.1007/s00401-001-0476-6. [DOI] [PubMed] [Google Scholar]
  • 8.van Groen T, Liu L, Ikonen S, Kadish I. Diffuse amyloid deposition, but not plaque number, is reduced in amyloid precursor protein/presenilin 1 double-transgenic mice by pathway lesions. Neuroscience. 2003;119(4):1185–1197. doi: 10.1016/s0306-4522(03)00215-x. [DOI] [PubMed] [Google Scholar]
  • 9.Thal DR, Capetillo-Zarate E, Del Tredici K, Braak H. The development of amyloid beta protein deposits in the aged brain. Science of Aging Knowledge Environment. 2006;2006(6):p. re1. doi: 10.1126/sageke.2006.6.re1. [DOI] [PubMed] [Google Scholar]
  • 10.Fiala JC. Mechanisms of amyloid plaque pathogenesis. Acta Neuropathologica. 2007;114(6):551–571. doi: 10.1007/s00401-007-0284-8. [DOI] [PubMed] [Google Scholar]
  • 11.Thal DR, Griffin WST, Braak H. Parenchymal and vascular Aβ-deposition and its effects on the degeneration of neurons and cognition in Alzheimer’s disease. Journal of Cellular and Molecular Medicine. 2008;12(5):1848–1862. doi: 10.1111/j.1582-4934.2008.00411.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dickson TC, Vickers JC. The morphological phenotype of β-amyloid plaques and associated neuritic changes in Alzheimer’s disease. Neuroscience. 2001;105(1):99–107. doi: 10.1016/s0306-4522(01)00169-5. [DOI] [PubMed] [Google Scholar]
  • 13.Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y. Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. Journal of Neuroscience. 2006;26(19):4985–4994. doi: 10.1523/JNEUROSCI.5476-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer’s disease. Neuron. 2009;63(3):287–303. doi: 10.1016/j.neuron.2009.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Raber J, Wong D, Buttini M, et al. Isoform-specific effects of human apolipoprotein E on brain function revealed in ApoE knockout mice: increased susceptibility of females. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(18):10914–10919. doi: 10.1073/pnas.95.18.10914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gordon I, Grauer E, Genis I, Sehayek E, Michaelson DM. Memory deficits and cholinergic impairments in apolipoprotein E-deficient mice. Neuroscience Letters. 1995;199(1):1–4. doi: 10.1016/0304-3940(95)12006-p. [DOI] [PubMed] [Google Scholar]
  • 17.Ji Y, Gong Y, Gan W, Beach T, Holtzman DM, Wisniewski T. Apolipoprotein E isoform-specific regulation of dendritic spine morphology in apolipoprotein E transgenic mice and Alzheimer’s disease patients. Neuroscience. 2003;122(2):305–315. doi: 10.1016/j.neuroscience.2003.08.007. [DOI] [PubMed] [Google Scholar]
  • 18.Klein RC, Mace BE, Moore SD, Sullivan PM. Progressive loss of synaptic integrity in human apolipoprotein E4 targeted replacement mice and attenuation by apolipoprotein E2. Neuroscience. 2010;171(4):1265–1272. doi: 10.1016/j.neuroscience.2010.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Piedrahita JA, Zhang SH, Hagaman JR, Oliver PM, Maeda N. Generation of mice carrying a mutant apolipoprotein E gene inactivated by gene targeting in embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(10):4471–4475. doi: 10.1073/pnas.89.10.4471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Veinbergs I, Mante M, Jung MW, Van Uden E, Masliah E. Synaptotagmin and synaptic transmission alterations in apolipoprotein E- deficient mice. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 1999;23(3):519–531. doi: 10.1016/s0278-5846(99)00013-5. [DOI] [PubMed] [Google Scholar]
  • 21.Robertson J, Curley J, Kaye J, Quinn J, Pfankuch T, Raber J. apoE isoforms and measures of anxiety in probable AD patients and apoE−/− mice. Neurobiology of Aging. 2005;26(5):637–643. doi: 10.1016/j.neurobiolaging.2004.06.003. [DOI] [PubMed] [Google Scholar]
  • 22.Sun Y, Wu S, Bu G, et al. Glial fibrillary acidic protein-apolipoprotein E (apoE) transgenic mice: astrocyte-specific expression and differing biological effects of astrocyte- secreted apoE3 and apoE4 lipoproteins. Journal of Neuroscience. 1998;18(9):3261–3272. doi: 10.1523/JNEUROSCI.18-09-03261.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hartman RE, Wozniak DF, Nardi A, Olney JW, Sartorius L, Holtzman DM. Behavioral phenotyping of GFAP-ApoE3 and -ApoE4 transgenic mice: ApoE4 mice show profound working memory impairments in the absence of Alzheimer’s-like neuropathology. Experimental Neurology. 2001;170(2):326–344. doi: 10.1006/exnr.2001.7715. [DOI] [PubMed] [Google Scholar]
  • 24.van Meer P, Acevedo S, Raber J. Impairments in spatial memory retention of GFAP-apoE4 female mice. Behavioural Brain Research. 2007;176(2):372–375. doi: 10.1016/j.bbr.2006.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Buttini M, Orth M, Bellosta S, et al. Expression of human apolipoprotein E3 or E4 in the brains of apoE−/− mice: isoform-specific effects on neurodegeneration. Journal of Neuroscience. 1999;19(12):4867–4880. doi: 10.1523/JNEUROSCI.19-12-04867.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Buttini M, Yu GQ, Shockley K, et al. Modulation of Alzheimer-like synaptic and cholinergic deficits in transgenic mice by human apolipoprotein E depends on isoform, aging, and overexpression of amyloid β peptides but not on plaque formation. Journal of Neuroscience. 2002;22(24):10539–10548. doi: 10.1523/JNEUROSCI.22-24-10539.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Harris FM, Brecht WJ, Xu Q, Mahley RW, Huang Y. Increased tau phosphorylation in apolipoprotein E4 transgenic mice is associated with activation of extracellular signal-regulated kinase. Modulation by zinc. The Journal of Biological Chemistry. 2004;279(43):44795–44801. doi: 10.1074/jbc.M408127200. [DOI] [PubMed] [Google Scholar]
  • 28.Sullivan PM, Han B, Liu F, et al. Reduced levels of human apoE4 protein in an animal model of cognitive impairment. Neurobiology of Aging. 2011;32(5):791–801. doi: 10.1016/j.neurobiolaging.2009.05.011. [DOI] [PubMed] [Google Scholar]
  • 29.Sullivan PM, Mace BE, Estrada JC, Schmechel DE, Alberts MJ. Human Apolipoprotein E4 Targeted Replacement Mice Show Increased Prevalence of Intracerebral Hemorrhage Associated with Vascular Amyloid Deposition. Journal of Stroke and Cerebrovascular Diseases. 2008;17(5):303–311. doi: 10.1016/j.jstrokecerebrovasdis.2008.03.011. [DOI] [PubMed] [Google Scholar]
  • 30.Bales KR, Liu F, Wu S, et al. Human APOE isoform-dependent effects on brain β-amyloid levels in PDAPP transgenic mice. Journal of Neuroscience. 2009;29(21):6771–6779. doi: 10.1523/JNEUROSCI.0887-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bour A, Grootendorst J, Vogel E, et al. Middle-aged human apoE4 targeted-replacement mice show retention deficits on a wide range of spatial memory tasks. Behavioural Brain Research. 2008;193(2):174–182. doi: 10.1016/j.bbr.2008.05.008. [DOI] [PubMed] [Google Scholar]
  • 32.Grootendorst J, Bour A, Vogel E, et al. Human apoE targeted replacement mouse lines: H-apoE4 and h-apoE3 mice differ on spatial memory performance and avoidance behavior. Behavioural Brain Research. 2005;159(1):1–14. doi: 10.1016/j.bbr.2004.09.019. [DOI] [PubMed] [Google Scholar]
  • 33.Dumanis SB, Tesoriero JA, Babus LW, et al. ApoE4 decreases spine density and dendritic complexity in cortical neurons in vivo. Journal of Neuroscience. 2009;29(48):15317–15322. doi: 10.1523/JNEUROSCI.4026-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Trommer BL, Shah C, Yun SH, et al. ApoE isoform affects LTP in human targeted replacement mice. NeuroReport. 2004;15(17):2655–2658. doi: 10.1097/00001756-200412030-00020. [DOI] [PubMed] [Google Scholar]
  • 35.Siegel JA, Haley GE, Raber J. Apolipoprotein E isoform-dependent effects on anxiety and cognition in female TR mice. doi: 10.1016/j.neurobiolaging.2010.03.002. Neurobiology of Aging. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Games D, Adams D, Alessandrini R, et al. Alzheimer-type neuropathology in transgenic mice overexpressing V717F β-amyloid precursor protein. Nature. 1995;373(6514):523–527. doi: 10.1038/373523a0. [DOI] [PubMed] [Google Scholar]
  • 37.Johnson-Wood K, Lee M, Motter R, et al. Amyloid precursor protein processing and Aβ42 deposition in a transgenic mouse model of Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(4):1550–1555. doi: 10.1073/pnas.94.4.1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Irizarry MC, Soriano F, McNamara M, et al. Aβ deposition is associated with neuropil changes, but not with overt neuronal loss in the human amyloid precursor protein V717F (PDAPP) transgenic mouse. Journal of Neuroscience. 1997;17(18):7053–7059. doi: 10.1523/JNEUROSCI.17-18-07053.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dodart JC, Mathis C, Saura J, Bales KR, Paul SM, Ungerer A. Neuroanatomical abnormalities in behaviorally characterized APP(V717F) transgenic mice. Neurobiology of Disease. 2000;7(2):71–85. doi: 10.1006/nbdi.1999.0278. [DOI] [PubMed] [Google Scholar]
  • 40.Bales KR, Verina T, Cummins DJ, et al. Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(26):15233–15238. doi: 10.1073/pnas.96.26.15233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fagan AM, Watson M, Parsadanian M, Bales KR, Paul SM, Holtzman DM. Human and murine apoE markedly alters Aβ metabolism before and after plaque formation in a mouse model of Alzheimer’s Disease. Neurobiology of Disease. 2002;9(3):305–318. doi: 10.1006/nbdi.2002.0483. [DOI] [PubMed] [Google Scholar]
  • 42.Holtzman DM, Bales KR, Tenkova T, et al. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(6):2892–2897. doi: 10.1073/pnas.050004797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bales KR, Liu F, Wu S, et al. Human APOE isoform-dependent effects on brain β-amyloid levels in PDAPP transgenic mice. Journal of Neuroscience. 2009;29(21):6771–6779. doi: 10.1523/JNEUROSCI.0887-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hsiao K, Chapman P, Nilsen S, et al. Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice. Science. 1996;274(5284):99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
  • 45.Irizarry MC, McNamara M, Fedorchak K, Hsiao K, Hyman BT. APP(Sw) transgenic mice develop age-related Aβ deposits and neuropil abnormalities, but no neuronal loss in CA1. Journal of Neuropathology and Experimental Neurology. 1997;56(9):965–973. doi: 10.1097/00005072-199709000-00002. [DOI] [PubMed] [Google Scholar]
  • 46.Takahashi RH, Milner TA, Li F, et al. Intraneuronal Alzheimer Aβ42 accumulates in multivesicular bodies and is associated with synaptic pathology. American Journal of Pathology. 2002;161(5):1869–1879. doi: 10.1016/s0002-9440(10)64463-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fryer JD, Simmons K, Parsadanian M, et al. Human apolipoprotein E4 alters the amyloid-β 40:42 ratio and promotes the formation of cerebral amyloid angiopathy in an amyloid precursor protein transgenic model. Journal of Neuroscience. 2005;25(11):2803–2810. doi: 10.1523/JNEUROSCI.5170-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hsia AY, Masliah E, Mcconlogue L, et al. Plaque-independent disruption of neural circuits in Alzheimer’s disease mouse models. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(6):3228–3233. doi: 10.1073/pnas.96.6.3228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Oakley H, Cole SL, Logan S, et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. Journal of Neuroscience. 2006;26(40):10129–10140. doi: 10.1523/JNEUROSCI.1202-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Youmans K, Laxton K, Jungbauer LM, Yu C, LaDu M. The Effects of Human ApoE on Amyloid-beta Pathology in a Novel Transgenic Mouse Model. San Diego, Calif, USA: Society for Neuroscience; 2010. [Google Scholar]
  • 51.Youmans K, Tai L, Laxton K, et al. ApoE isoform-specific effects on Aβ pathology in EFAD mice. In: Proceedings of the 2nd annual symposium of ApoE, ApoE Receptors, and Neurodegeneration; 2011; Chicago, Ill, USA. [Google Scholar]
  • 52.Youmans KL. The effects of human apoE on amyloid-beta (42) pathology and synaptic loss in a novel transgenic mouse model. In: Proceedings of the American Society for Neurochemistry Meeting; 2010; Santa Fe, NM, USA. Transactions of the American Society for Neurochemistry. [Google Scholar]
  • 53.Yu C, Youmans KL, Jungbauer LM, et al. Selective increases in soluble ApoE4 and Aβ42 in EFAD mouse brains. In: Proceedings of the ApoE, ApoE Receptors and Neurodegeneration; 2011; Chicago, Ill, USA. [Google Scholar]
  • 54.Hatters DM, Peters-Libeu CA, Weisgraber KH. Apolipoprotein E structure: insights into function. Trends in Biochemical Sciences. 2006;31(8):445–454. doi: 10.1016/j.tibs.2006.06.008. [DOI] [PubMed] [Google Scholar]
  • 55.Tesseur I, Van Dorpe J, Bruynseels K, et al. Prominent axonopathy and disruption of axonal transport in transgenic mice expressing human apolipoprotein E4 in neurons of brain and spinal cord. American Journal of Pathology. 2000;157(5):1495–1510. doi: 10.1016/S0002-9440(10)64788-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Smith JD, Sikes J, Levin JA. Human apolipoprotein E allele-specific brain expressing transgenic mice. Neurobiology of Aging. 1998;19(5):407–413. doi: 10.1016/s0197-4580(98)00076-1. [DOI] [PubMed] [Google Scholar]
  • 57.Huber G, März W, Martin JR, et al. Characterization of transgenic mice expressing Apolipoprotein E4(C112R) and Apolipoprotein E4(L28P; C112R) Neuroscience. 2000;101(1):211–218. doi: 10.1016/s0306-4522(00)00341-9. [DOI] [PubMed] [Google Scholar]
  • 58.Bowman BH, Jansen L, Yang F, et al. Discovery of a brain promoter from the human transferrin gene and its utilization for development of transgenic mice that express human apolipoprotein E alleles. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(26):12115–12119. doi: 10.1073/pnas.92.26.12115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tesseur I, Van Dorpe J, Spittaels K, Van den Haute C, Moechars D, Van Leuven F. Expression of human apolipoprotein E4 in neurons causes hyperphosphorylation of protein tau in the brains of transgenic mice. American Journal of Pathology. 2000;156(3):951–964. doi: 10.1016/S0002-9440(10)64963-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Han SH, Einstein G, Weisgraber KH, et al. Apolipoprotein E is localized to the cytoplasm of human cortical neurons: a light and electron microscopic study. Journal of Neuropathology and Experimental Neurology. 1994;53(5):535–544. doi: 10.1097/00005072-199409000-00013. [DOI] [PubMed] [Google Scholar]
  • 61.Han SH, Hulette C, Saunders AM, et al. Apolipoprotein E is present in hippocampal neurons without neurofibrillary tangles in Alzheimer’s disease and in age-matched controls. Experimental Neurology. 1994;128(1):13–26. doi: 10.1006/exnr.1994.1108. [DOI] [PubMed] [Google Scholar]
  • 62.Buttini M, Masliah E, Yu GQ, et al. Cellular source of apolipoprotein E4 determines neuronal susceptibility to excitotoxic injury in transgenic mice. American Journal of Pathology. 2010;177(2):563–569. doi: 10.2353/ajpath.2010.090973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chen HK, Ji ZS, Dodson SE, et al. Apolipoprotein E4 domain interaction mediates detrimental effects on mitochondria and is a potential therapeutic target for alzheimer disease. The Journal of Biological Chemistry. 2011;286(7):5215–5221. doi: 10.1074/jbc.M110.151084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mufson EJ, Benzing WC, Cole GM, et al. Apolipoprotein E-immunoreactivity in aged rhesus monkey cortex: colocalization with amyloid plaques. Neurobiology of Aging. 1994;15(5):621–628. doi: 10.1016/0197-4580(94)00064-6. [DOI] [PubMed] [Google Scholar]
  • 65.Fujita SC, Sakuta K, Tsuchiya R, Hamanaka H. Apolipoprotein E is found in astrocytes but not in microglia in the normal mouse brain. Neuroscience Research. 1999;35(2):123–133. doi: 10.1016/s0168-0102(99)00075-9. [DOI] [PubMed] [Google Scholar]
  • 66.Stone DJ, Rozovsky I, Morgan TE, Anderson CP, Hajian H, Finch CE. Astrocytes and microglia respond to estrogen with increased apoE mRNA in vivo and in vitro. Experimental Neurology. 1997;143(2):313–318. doi: 10.1006/exnr.1996.6360. [DOI] [PubMed] [Google Scholar]
  • 67.Grehan S, Tse E, Taylor JM. Two distal downstream enhancers direct expression of the human apolipoprotein E gene to astrocytes in the brain. Journal of Neuroscience. 2001;21(3):812–822. doi: 10.1523/JNEUROSCI.21-03-00812.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Poirier J. Apolipoprotein E, cholesterol transport and synthesis in sporadic Alzheimer’s disease. Neurobiology of Aging. 2005;26(3):355–361. doi: 10.1016/j.neurobiolaging.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 69.Poirier J. Apolipoprotein E represents a potent gene-based therapeutic target for the treatment of sporadic Alzheimer’s disease. Alzheimer’s and Dementia. 2008;4(1, supplement):S91–S97. doi: 10.1016/j.jalz.2007.11.012. [DOI] [PubMed] [Google Scholar]
  • 70.Ramaswamy G, Xu Q, Huang Y, Weisgraber KH. Effect of domain interaction on apolipoprotein E levels in mouse brain. Journal of Neuroscience. 2005;25(46):10658–10663. doi: 10.1523/JNEUROSCI.1922-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Raffaï RL, Dong LM, Farese RV, Weisgraber KH. Introduction of human apolipoprotein E4 "domain interaction" into mouse apolipoprotein E. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(20):11587–11591. doi: 10.1073/pnas.201279298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bertrand P, Poirier J, Oda T, Finch CE, Pasinetti GM. Association of apolipoprotein E genotype with brain levels of apolipoprotein E and apolipoprotein J (clusterin) in Alzheimer disease. Molecular Brain Research. 1995;33(1):174–178. doi: 10.1016/0169-328x(95)00097-c. [DOI] [PubMed] [Google Scholar]
  • 73.Glöckner F, Meske V, Ohm TG. Genotype-related differences of hippocampal apolipoprotein E levels only in early stages of neuropathological changes in Alzheimer’s disease. Neuroscience. 2002;114(4):1103–1114. doi: 10.1016/s0306-4522(02)00178-1. [DOI] [PubMed] [Google Scholar]
  • 74.Sun Y, Wu S, Bu G, et al. Glial fibrillary acidic protein-apolipoprotein E (apoE) transgenic mice: astrocyte-specific expression and differing biological effects of astrocyte- secreted apoE3 and apoE4 lipoproteins. Journal of Neuroscience. 1998;18(9):3261–3272. doi: 10.1523/JNEUROSCI.18-09-03261.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tang KF, Cai L, Zhou JN. Observation of the density and size of cells in hippocampus and vascular lesion in thalamus of GFAP-apoE transgenic mice. Neuroscience Bulletin. 2009;25(4):167–178. doi: 10.1007/s12264-009-0324-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wirths O, Bayer TA. Neuron loss in transgenic mouse models of Alzheimer’s disease. International Journal of Alzheimer’s Disease. 2010;2010:6 pages. doi: 10.4061/2010/723782. Article ID 723782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dodart JC, Bales KR, Johnstone EM, Little SP, Paul SM. Apolipoprotein E alters the processing of the β-amyloid precursor protein in APPV717F transgenic mice. Brain Research. 2002;955(1-2):191–199. doi: 10.1016/s0006-8993(02)03437-6. [DOI] [PubMed] [Google Scholar]
  • 78.Bales KR, Verina T, Cummins DJ, et al. Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(26):15233–15238. doi: 10.1073/pnas.96.26.15233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bales KR, Verina T, Dodel RC, et al. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nature genetics. 1997;17(3):263–264. doi: 10.1038/ng1197-263. [DOI] [PubMed] [Google Scholar]
  • 80.Holtzman DM, Fagan AM, Mackey B, et al. Apolipoprotein E facilitates neuritic and cerebrovascular plaque formation in an Alzheimer’s disease model. Annals of Neurology. 2000;47(6):739–747. [PubMed] [Google Scholar]
  • 81.Raber J, Wong D, Yu GQ, et al. Apolipoprotein E and cognitive performance. Nature. 2000;404(6776):352–354. doi: 10.1038/35006165. [DOI] [PubMed] [Google Scholar]
  • 82.Hartman RE, Laurer H, Longhi L, et al. Apolipoprotein E4 influences amyloid deposition but not cell loss after traumatic brain injury in a mouse model of Alzheimer’s disease. Journal of Neuroscience. 2002;22(23):10083–10087. doi: 10.1523/JNEUROSCI.22-23-10083.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Colton CA, Wilcock DM, Wink DA, Davis J, Van Nostrand WE, Vitek MP. The effects of NOS2 gene deletion on mice expressing mutated human AβPP. Journal of Alzheimer’s Disease. 2008;15(4):571–587. doi: 10.3233/jad-2008-15405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Belinson H, Lev D, Masliah E, Michaelson DM. Activation of the amyloid cascade in apolipoprotein E4 transgenic mice induces lysosomal activation and neurodegeneration resulting in marked cognitive deficits. Journal of Neuroscience. 2008;28(18):4690–4701. doi: 10.1523/JNEUROSCI.5633-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Dolev I, Michaelson DM. A nontransgenic mouse model shows inducible amyloid-β (Aβ) peptide deposition and elucidates the role of apolipoprotein E in the amyloid cascade. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(38):13909–13914. doi: 10.1073/pnas.0404458101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Oddo S, Caccamo A, Shepherd JD, et al. Triple-transgenic model of Alzheimer’s Disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron. 2003;39(3):409–421. doi: 10.1016/s0896-6273(03)00434-3. [DOI] [PubMed] [Google Scholar]
  • 87.Oddo S, Caccamo A, Smith IF, Green KN, LaFerla FM. A dynamic relationship between intracellular and extracellular pools of Aβ . American Journal of Pathology. 2006;168(1):184–194. doi: 10.2353/ajpath.2006.050593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Casas C, Sergeant N, Itier JM, et al. Massive CA1/2 neuronal loss with intraneuronal and N-terminal truncated Aβ42 accumulation in a novel Alzheimer transgenic model. American Journal of Pathology. 2004;165(4):1289–1300. doi: 10.1016/s0002-9440(10)63388-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Radde R, Bolmont T, Kaeser SA, et al. Aβ42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Reports. 2006;7(9):940–946. doi: 10.1038/sj.embor.7400784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Chishti MA, Yang DS, Janus C, et al. Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid precursor protein 695. The Journal of Biological Chemistry. 2001;276(24):21562–21570. doi: 10.1074/jbc.M100710200. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Alzheimer's Disease are provided here courtesy of Wiley

RESOURCES