Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Send Orders for Reprints to reprints@benthamscience.

net 281

Current Alzheimer Research, 2019, 16, 281-292


RESEARCH ARTICLE
ISSN: 1567-2050
eISSN: 1875-5828

The Role of MAPK's Signaling in Mediating ApoE4-Driven Pathology Impact


Factor
Current: 3.289
5-Year: 3.595

In Vivo
BENTHAM
SCIENCE

Shiran Salomon-Zimria, Amit Korena, Ariel Angelb, Tali Ben-Zurb, Daniel Offenb and
Daniel M. Michaelsona,*
a
Department of Neurobiology, Sagol School of Neuroscience, The George S. Wise Faculty of Life Sciences, Tel Aviv
University, Tel Aviv, Israel; bSackler School of Medicine, Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv,
Israel

Abstract: Background: Alzheimer's Disease (AD) is associated with impairments in key brain Mito-
gen-Activated Protein Kinase (MAPK) signaling cascades including the p38, c-Jun N-terminal kinase
(JNK), ERK and Akt pathways. Apolipoprotein E4 (ApoE4) is the most prevalent genetic risk factor of
AD.
Objectives: To investigate the extent to which the MAPK signaling pathway plays a role in mediating
the pathological effects of apoE4 and can be reversed by experimental manipulations.
Methods: Measurements of total level and activation of MAPK signaling pathway factors, obtained util-
Current Alzheimer Research

ARTICLE HISTORY izing immunoblot assay of hippocampal tissues from naïve and viral-treated apoE3 and apoE4 targeted
replacement mice.
Received: October 09, 2018
Revised: December 18, 2018 Results: ApoE4 mice showed robust activation of the stress related p38 and JNK pathways and a corre-
Accepted: February 04, 2019
sponding decrease in Akt activity, which is coupled to activation of GSK3β and tau hyper-
phosphorylation. There was no effect on the ERK pathway. We have previously shown that the apoE4-
DOI:
10.2174/1567205016666190228120254 related pathology, namely; accumulation of Aβ, hyper-phosphorylated tau, synaptic impairments and
decreased VEGF levels can be reversed by up-regulation of VEGF level utilizing a VEGF-expressing
adeno-associated virus. Utilizing this approach, we assessed the extent to which the AD-hallmark and
synaptic pathologies of apoE4 are related to the corresponding MAPK signaling effects. This revealed
that the reversal of the apoE4-driven pathology via VEGF treatment was associated with a reversal of
the p38 and Akt related effects.
Conclusion: Taken together, these results suggest that the p38 and Akt pathways play a role in mediat-
ing the AD-related pathological effects of apoE4 in the hippocampus.

Keywords: Alzheimer's disease (AD), apolipoprotein E4 (apoE4), VEGF, MAPK, signaling, adeno-associated virus, hippo-
campus, targeted replacement mice.

1. INTRODUCTION apoE2 (Cys-112, Cys-158), apoE3 (Cys-112, Arg-158), and


apoE4 (Arg-112, Arg-158) of which APOEε4 is the most
Alzheimer’s Disease (AD) is the most common cause of
prevalent genetic risk factor for AD [5, 6]. About half of the
dementia. AD brain pathology is characterized by the occur- AD patients express APOEε4, whereas only about a quarter
rence of brain senile plaques and Neuro-Fibrillary Tangles
of the population expresses this isoform. ApoE4 increases
(NFTs), as well as synaptic and neuronal loss and increased
the risk for AD by ~3- and 15-fold for apoE4 heterozygotes
neuro-inflammation [1-4]. Genetic studies revealed allelic
and homozygotes, respectively, compared with ApoE3. In-
segregation of the apolipoprotein E gene in families and in-
terestingly, ApoE2, which is the least prevalent allele, is as-
dividuals with a higher risk of late onset and sporadic Alz-
sociated with decreased AD risk [5-8]. ApoE4 is associated
heimer’s disease [5-7]. with distinct brain pathologies, including increased neurode-
Apolipoprotein E (APOE for gene, apoE for protein) ex- generation [9], impaired neurite outgrowth, impaired synap-
pressed in the brain as three common isoforms: apoE2, togenesis [10], and impaired neuronal remodeling [11].
apoE3, and apoE4, corresponding to the three APOE alleles: ApoE4 is also associated with increased vascular pathology
APOEε2, APOEε3, APOEε4, which differ from one another [12] and increased brain inflammation [13]. The prevalence
by one amino acid substitutions at positions 112 and 158: of AD in apoE4 carriers is higher in females than in males
[14], Additionally, apoE4-driven pathologies are exacerbated
*Address correspondence to this author at the Department of Neurobiology, in the presence of additional risk factors or stressors, such as
Sagol School of Neuroscience, The George S. Wise Faculty of Life Sci- old age, environmental factors, female sex and other genetic
ences, Tel Aviv University, Ramat Aviv 6997801, Tel Aviv, Israel; modulation. Moreover, Recent findings from our lab suggest
Tel: 972-3-6409624; Fax: 972-6406356; E-mail: dmichael@post.tau.ac.il

1875-5828/19 $58.00+.00 © 2019 Bentham Science Publishers


282 Current Alzheimer Research, 2019, Vol. 16, No. 4 Salomon-Zimri et al.

that female mice have a more pronounced apoE4-induced ERK is activated and JNK is inhibited by apoE4 in primary
pathological phenotype than the corresponding male mice neurons [23], p38 is activated in primary apoE4 microglial
[15]. cultures [32], and apoE4 inhibits the PI3K/Akt pathway in
endothelial cells [33]. ApoE4-driven MAPK signaling was
Because of the critical role of the Mitogen-Activated Pro-
tein Kinase (MAPK) pathways in regulating cellular func- also observed in vivo, where fasting of apoE4- targeted re-
placement mice was found to be associated with decreased
tion, the importance of MAPKs in AD pathogenesis is being
Akt activities, whereas p38 activity increased during aging
increasingly recognized. The MAPK signaling pathways,
[34]. A key unresolved issue is the extent to which the effect
which include the Extracellular Signal-Regulated Kinase
of apoE4 on MAPK signaling plays a role in mediating the
(ERK), c-Jun N-terminal kinase (JNK), and p38 pathways,
pathological effects of apoE4. One way to address this issue
are activated by various types of cellular stress such as oxi-
dative, genotoxic, mitochondrial dysfunction, and osmotic is to determine whether experimental manipulations that
have been found to reverse apoE4 pathological effect, such
stress as well as pro-inflammatory cytokines [16, 17]. All of
as up-regulation of VEGF, can also reverse apoE4- driven
these MAPK pathways are activated in vulnerable neurons in
MAPK signaling pathologies. Utilizing apoE4 and apoE3 TR
patients with AD, suggesting that MAPK pathways are in-
mice we have recently shown that apoE4 is associated cogni-
volved in the pathophysiology and pathogenesis of AD [18].
tive decline [35] and with marked synaptic and neuronal
Specifically, p38, which enables cells to respond to stress-
related stimuli from the extracellular environment, has in- effects in the hippocampus (e.g., reduced levels of synapto-
physin, the presynaptic transporters VGluT and VGaT, and
creased activity in human AD brains [19, 20], and mediates
the apoE receptor apoER2) and that apoE4 is associated with
β-amyloid-driven microglial activation [21] and tau hyper-
elevated levels of brain Aβ and hyper-phosphorylated Tau
phosphorylation [16]. Specifically, p38 plays more than one
[15, 36, 37], as well as with reduced levels of VEGF [38].
role in AD pathophysiology; in microglial p38 MAPK con-
Furthermore, reversal of the apoE4-driven reduction in hip-
tributes to the inflammation of the AD brains, in astrocytes
the p38 MAPK cascade expands the inflammatory response pocampal VEGF levels by intra-hippocampal injection of
VEGF-expressing-lentivirus reversed the apoE4-driven cog-
and modulates the excitotoxicity, while the neuronal p38
nitive deficits and synaptic pathologies [38]. This experi-
MAPK signaling contributes to tau phosphorylation, NFT
mental manipulation is used for investigating the possible
formation and depression of synaptic plasticity [19]. JNK is
relationship between apoE4-driven brain pathology and the
activated in response to cellular stress, and regulates various
MAPK signaling pathway. Accordingly, the present study
processes such as brain development and repair, as well as
neuro-inflammation, cell proliferation, and cell death [22]. investigated the effects of apoE4 on MAPK signaling, and
the extent to which VEGF is involved in these signal trans-
JNK also mediates the neurotoxic signals driven via Aβ and
duction pathways. This was pursued by measuring the levels
contributes to cell death and various processes in AD pathol-
and activation of p38, JNK/c-Jun, ERK, and Akt/GSK-3β in
ogy [23, 24]. Activated JNK and p38 are associated with
the hippocampus of apoE4 mice, in which it was shown to
cells that contain filamentous tau, suggesting that they may
particularly be affected by apoE4. An additional aim was to
also contribute to the hyper-phosphorylation of tau and the
formation of paired helical filament and involved in abnor- investigate the extent to which hippocampal-specific injec-
tion of a VEGF-expressing virus reverses the apoE4-driven
mal processes, ranging from tau phosphorylation to neuronal
MAPK signaling.
death [19, 21, 24]. The PI3K/Akt pathway promotes survival
and growth in response to extracellular signals. In normal
brains, the P13K/Akt pathway inactivates GSK3, and pre- 2. MATERIALS AND METHODS
vents apoptosis of confluent cells. In AD, as well as in 2.1. Mice
ischemia and oxidative stress, Akt is inhibited. This leads to
GSK3β activation and subsequently to tau hyper- ApoE-TR mice, in which the endogenous mouse apoE
phosphorylation and the formation of PHF-tau [25, 26]. was replaced by either human apoE3 or apoE4, were created
GSK3β is a pleiotropic enzyme involved in a variety of cell by gene targeting, as previously described [34]. These mice
activities, and has been postulated as a therapeutic target for were purchased from Taconic (Germantown, NY). In order
AD due to its multiple connections to the pathology of the to minimize possible genetic drift between the apoE4 and
disease [27]. The Extracellular signal–Regulated Kinase apoE3 mice, which were offspring of apoE4 and apoE3 ho-
(ERK) pathway is activated by many different stimuli, in- mozygous mouse colonies generated by Taconic around
cluding growth factors, cytokines, and transforming agents. 2001, the mice were back-crossed by us with Harlan
The ERK cascade is involved in a wide range of processes, C57Bl/6JOlaHsd mice for 10 generations. The present ex-
such as metabolism, motility, inflammation, cell death, and periments were performed with apoE3 and apoE4 homozy-
survival [28]. In AD and corresponding transgenic mouse gous mice on an α synuclein -/- background in which the
models, ERK signaling plays a role in mediating the patho- apoE4-driven synaptic and AD-related phenotype are more
logical effects of oligomeric Aβ42, in the activation of GSK- pronounced [32]. These mice are referred to in the text as
3 [29], and in impaired mitochondrial fission/fusion events apoE3 and apoE4 mice, respectively. The apoE genotype of
[30]. the mice was confirmed by PCR analysis [35-37]. All ex-
periments were performed on 4-month-old male and female
Accumulating evidence suggests that apoE4 is associated mice, and were approved by the Tel Aviv University Animal
with increased cellular stress, specifically oxidative stress, Care Committee. Following the indicated treatments, the
endoplasmic reticulum stress, as well as with impaired im- mice were anesthetized with ketamine and xylazine and were
mune function [31]. In some in-vitro studies, this was shown perfused transcardially with phosphate buffered saline
to be associated with altered MAPK signaling. For example, (PBS). Their brains were then removed and halved, and each
The Role of MAPK's Signaling in Mediating ApoE4-Driven Current Alzheimer Research, 2019, Vol. 16, No. 4 283

hemisphere was further processed for either biochemical or coordinates: ±2.3 mm medial/lateral, −2.1 mm ante-
histological analysis, as outlined below. In the current study, rior/posterior, and −2.2 mm dorsal/ventral from the bregma.
we chose to focus on female mice. This is driven by the lit- The preparation was injected with a speed of 0.5 μL/min
erature based findings that females are more susceptible to over a period of 2 min utilizing a Hamilton 10-μL syringe
AD than males, and that the apoE4 related pathology is more and a 26-gage needle. The needle was kept in place for ~3
pronounced in females than in the corresponding males. Fur- min in order to prevent backflow. The mice were then
thermore, recent findings from our lab showed that apoE4- stitched and returned to their cages.
TR female mice have a more pronounced pathological phe-
notype than the corresponding male mice. Specifically, the 2.4. Immunoblot Analysis
AD-related pathology (e.g. Aβ and tau phosphorylation),
synaptic deficits (e.g. VGluT1,VGaT and SYP), vascular Immunoblot analysis was performed as previously de-
pathology (collagen IV) and cognitive impairments were scribed [38, 39]. The following Abs were used: rabbit anti-
more robust in female mice compared to male mice [15]. The VEGF (1:1000; Calbiochem), rabbit anti-p38 (1:1000, cell
results presented correspond to 4 female cohorts. The first 2 signaling), rabbit anti-pp38 (1:1000, cell signaling), rabbit
female cohorts were used to characterize the effects of apoE4 anti-Akt (1:1000, cell signaling), rabbit anti-pAkt
and apoE3 on MAPK signaling and contained only naïve (1:1000,cell signaling), rabbit anti-ERK1/2 (1:5000, Santa
non-treated mice (10-12 mice/cohort), whereas the other 2 Cruz), mouse anti-pERK1/2 (1:10000, Sigma), rabbit anti-
female cohorts were used to assess the effects of the VEGF- JNK (1:1000, cell signaling), rabbit anti-pJNK (1:1000, cell
expressing adeno-associated-virus (AAV-VEGF) treatment signaling), mouse anti-GSK3β (1:1000, Santa Cruz), mouse
on apoE4 pathology and MAPK signaling. These experi- anti-pGSK3β (1:1000, Santa Cruz), and mouse anti-tubulin
ments contain 6 groups: 2 genotypes (apoE3 or apoE4) x 3 (1:1000, Sigma). Membranes were scanned utilizing the
treatments (naïve mice, mice treated either with a sham GFP- ChemiDoc Touch imaging system (Bio-Rad) and the blot
labeled virus or mice treated with the VEGF-expressing vi- intensity was then quantified using Image Lab Software
rus). These groups are referred to as naïve, sham-treated, and (Bio-Rad). All results were normalized to the naïve-apoE3
VEGF-treated mice. Each group consisted of 5-7 group and utilized beta-tubulin as gel-loading control.
mice/cohort. A control assay with corresponding apoE3 and
apoE4 male mice conduct was conducted for both the naïve 2.5. Quantification and Statistical Analysis
and AAV-VEGF experiments. The results of the naïve mice, which consisted of non-
treated apoE3 and apoE4 mice of each cohort, were normal-
2.2. Adeno-associated Virus Preparation ized relative to apoE3 mice, after which they were analyzed
Human VEGF and GFP coding sequences were cloned utilizing Student's t-test. The experimental design of the viral
into the PAAV2-GFAP-OPTOAI-P2A-EYFP backbone, construct-treated mice consisted of 2 genotypes (apoE3 and
kindly provided by the Diesseroth lab [39]. The human apoE4, and 3 treatments (naïve, VEGF-expressing virus, and
VEGF was amplified from pBluescript plasmid (purchased sham GFP-labelled virus; n= 5-7 mice /group in each co-
from Harvard Institute of Proteomics, Boston, USA) by PCR hort). Each MAPK-related molecule was assessed utilizing a
using restriction enzymes at the ends (VEGF-AGEI-FOR 5' general phosphorylation-independent antibody whose im-
TAGACCGGTATGAACTTTCTGCTGTCTTGG3' VEGF- munoreactivity corresponded to the total level of the mole-
ECORI-REV 5'TCAAGAATTCCAACCGCCTCGGCTTGT cule and by the indicated phosphorylation-dependent mole-
CACAT3'). Both PAAV2-GFAP-OPTOAI-P2A-EYFP and cule whose immunoreactivity relative to the total level was
PLL3.7-GFAP-EGFP plasmids were cut with AGEI and used as a measure of the kinase activity. The results of each
ECORI. OPTOAI-P2A-EYFP sequence replaced with the cohort were normalized relative to apoE3 mice, after which
GFP sequence or the VEGF sequence in order to obtain the they were analyzed utilizing 2-way ANOVA using STATIS-
final expression constructs. As we described in Salomon- TICA software (Version 8.0 StatSoft, Inc., Tulsa, OK, USA).
Zimri et al, 2016, Concentrated AAV2/1 mosaic particles Further examination of the specific effect of treatment utiliz-
were produced by the Tel Aviv University’s vector core fa- ing 1-way ANOVA, followed by post-hoc Bonferroni correc-
cility. Briefly, HEK293 cells were transfected with the tion, was performed to test the specific effect of VEGF
AAV2-GFAP-GFP/VEGF-WPRE vector plasmid along with treatment on the apoE4 mouse group. Beta-tubulin whose
AdenoHelper, Rep-Cap1 and Rep-Cap2 plasmids. Seventy- levels were similar in the different groups was used as load-
two hours post transfection the cells were harvested, lysed ing controls and all the results were normalized relative to
using three freeze-thaw cycles and incubated with Benzonase the apoE3 naïve group standard. All graphs were prepared
for 90 min. The lysate was purified using a heparin-agarose and double analyzed via GraphPed prism 5.3 software.
binding column and subsequently concentrated and desali-
nated using an Amicon filter. 3. RESULTS
The effects of the apoE genotype on the p38 MAPK
2.3. Intracerebral Administration of Viral Vectors pathway were assessed by measuring the effects of apoE4
Four-month-old apoE3 and apoE4-TR mice were anes- and apoE3 on the levels of p38 and phospho/activated p38 in
thetized with a ketamine-xylazine mixture and placed in a the hippocampus of the corresponding targeted replacement
stereotactic apparatus (model 940; David Kopf). Subse- mice. As can be seen in Fig. (1), the levels of p38 were the
quently, 1 μL of the viral preparation was injected bilaterally same in apoE3 and apoE4 mice, whereas the extent of activa-
into the CA3 region of the hippocampus using the following tion of p38, as measured by the ratio of phospho-p38 to total
p38, was significantly higher in the apoE4 than in the apoE3
284 Current Alzheimer Research, 2019, Vol. 16, No. 4 Salomon-Zimri et al.

Fig. (1). The effects of apoE4 on p38 levels and activation. Representative immunoblots of the effects of apoE4 and apoE3 on the total levels
of p38 in the hippocampus and on the corresponding levels of phosphorylated p38. . Hippocampi of 4-month-old female apoE3 and apoE4
TR mice were excised, homogenized, and subjected to p-38 and phosphorylated p-38 immunoblotting , as described in Materials and Meth-
ods. Quantitation of the total hippocampal p38 levels and of activated p38, as presented by the ratio of the phosphorylated p38 to its levels,
are presented in the in the central and right panels. ApoE3 mice are depicted in the white bar and the corresponding apoE4 mice are depicted
in the black bars. Beta-tubulin was used as a loading reference and the results presented (2.3 +/- 0.35 and 1+\- 0.08 for the ratio of apoE4 and
apoE3 mice, respectively; n=10-12 per group) are normalized relative to the apoE3 mice ***p<0.001 by Student’s t-test. Similar results were
obtained with a second cohort of mice.

mice (2.3 +/- 0.35 and 1+\- 0.08 ; ***p<0.0001, respec- this treatment on the total levels and activation of the p38,
tively). The corresponding effects of the apoE genotype on JNK, ERK, and Akt pathways were measured. The resulting
the JNK MAPK pathway were next examined. As can be effects on p38 activation are depicted in Fig. (5). As can be
seen in Fig. (2), the total levels of both JNK and c-Jun in the seen, the AAV-VEGF treatment abolished the apoE4-related
hippocampus of female apoE3 and apoE4 mice were similar, p38 activation and rendered it similar to that of apoE3 mice,
whereas the ratios of phosphorylated/activated JNK were which was not affected by this treatment. Quantification of
significantly higher in the apoE4 mice (1.6 +\-0.18 vs. 1 +\- these results by 2-way ANOVA, revealed significant effects
0.09; *p<0.01, respectively) as were those of the activated c- for group, ***p<0.0001 and *p<0.05 for group and treat-
Jun (1.4 +/- 0.15 vs. 0.9+\- 0.1; **P<0.01, respectively). ment, respectively. Further Bonferroni post-hoc analysis
revealed *p<0.05 for the specific effect of VEGF treatment
We next focused on the effects of apoE4 on the growth
factor-related MAPK pathways utilizing ERK, Akt, and on apoE4 mice. In contrast, the total levels of p38, which
were similar in the two control mouse groups (Fig. 1), were
GSK3β as markers. As shown in Fig. (3), and unlike the re-
not affected by the VEGF treatment (not shown). Measuring
sults that were obtained with the p38 and JNK pathways,
the effects of the AAV-VEGF treatment on c-Jun and JNK
apoE4 did not affect either the levels of ERK or the extent of
phosphorylation revealed that they were higher in naïve
its activation (1.2 +\- 0.2 relative to 0.938 +/-0.16; respec-
apoE4 mice than in the corresponding apoE3 mice and were
tively). Measurements of the Akt-GSK3β pathway, in which
GSK3β is to be a well-established substrate of Akt and not affected by the sham treatment (Fig. 6). However, this
effect was reduced to non-significant levels by the AAV-
whose levels of activation correlate inversely with those of
VEGF treatment (Fig. 6). Quantification of these results re-
Akt [40, 41], revealed that apoE4 down-regulates the activa-
vealed significant effects for the group. **p<0.01 and treat-
tion of Akt relative to the corresponding apoE3 mice (0.6 +\-
ment *p<0.05 for JNK activation measurements, and
0.1 relative to 0.93 +/-0.11; **p<0.01, respectively) and in
*p<0.05 for the effect of the group for c-Jun activation by 2-
turn up-regulated GSK3β levels of activation (1.8+\- 0.2
relative to 1 +/-0/03; ***<0.001, respectively) without af- way ANOVA. The total levels of c-Jun and JNK, which were
the same in the naïve apoE4 and apoE3 mice (Fig. 2), were
fecting the total levels of both Akt and GSK3β (Fig. 4; A and
also not affected by either the AAV-VEGF or sham treat-
B, respectively). Tau phosphorylation is highly dependent on
ments. Further examination of the ERK pathway, which was
GSK3β activity [41], and these results are in accordance with
not affected by the apoE genotype under basal conditions
previous observations that tau is hyper-phosphorylated in
(Fig. 2), revealed that it was also unaffected by neither the
apoE4 mice [15, 36] in a manner similar to that observed in
AD patients. sham control nor the AAV-VEGF treatment (Fig. 7)
The effects of apoE4 on Akt and GSK3β activations were
The possible association of apoE4-driven MAPK signal-
also reversed by the AAV-VEGF treatment. Accordingly,
ing with the synaptic and AD pathological effects of apoE4
the AAV-VEGF treatment abolished the related apoE4-
was examined next. This was performed by intra-
driven decrease in Akt, as depicted in Fig. (4), and rendered
hippocampal injection of VEGF-expressing virus (AAV-
VEGF), which was previously shown to elevate the levels of it similar to that of the corresponding apoE3 mice, which
were not affected by this treatment (Fig. 7). Quantification of
VEGF and reverse the synaptic and AD-related phenotype of
these results revealed significant effects for the group,
apoE4 [38]. The extent to which these protective effects are
*p<0.05, by 2-way ANOVA. A complementary effect was
associated with reversal of the apoE4-driven effects on
observed in GSK3β activation, in which the elevated acti-
MAPK signaling was examined. Accordingly, the effects of
vated GSK3β levels of the naïve apoE4 mice (Fig. 4) were
The Role of MAPK's Signaling in Mediating ApoE4-Driven Current Alzheimer Research, 2019, Vol. 16, No. 4 285

Fig. (2). The effects of apoE4 on JNK and Cc-Jun levels and activation. Hippocampi of 4-month-old female apoE3 and apoE4 TR mice were
excised, homogenized, and subjected to phosphorylated and total JNK(A) and c-Jun (B) immunoblotting , as described in Materials and
Methods. (A) Representative immunoblots of the effects of apoE4 and apoE3 on the total levels and phosphorylation of JNK in the hippo-
campus are presented in the left panel. Quantification of the total hippocampal JNK levels in these mice and of activated JNK levels, as pre-
sented by the ratio of phosphorylated JNK to its total level, are presented in in the central and right panels (1.6 +/-0.17 and 1+\- 0.09 for the
ratio of apoE4 and apoE3 mice, respectively; n=10-12 per group). (B) Representative immunoblots of the effects of apoE4 and apoE3 on the
total levels and phosphorylation of c-Jun in the hippocampus, as presented by the ratio of phosphorylated c-Jun to its total level, are presented
in the panel on the left. Quantitation of the total hippocampal Cc-Jun levels in these mice and of the ratio of the activated c-Jun to the total c-
Jun levels are presented in the central and right panels (1.5+/-0.15 and 1+\- 0.1 for the ratio of apoE4 and apoE3 mice, respectively). ApoE3
mice are depicted in the white bar and the corresponding apoE4 mice are depicted in the black bars. Beta-tubulin was used as a loading refer-
ence and the results presented are normalized relative to the apoE3 **p<0.01 and ***p<0.001 by Student’s t-test. Similar results were ob-
tained with another cohort of such mice.

Fig. (3). The effects of apoE4 on ERK levels and activation. Hippocampi of 4-month-old female apoE3 and apoE4 TR mice were excised,
homogenized, and subjected to phosphorylated and total ERK immunoblotting , as described in Materials and Methods. Representative im-
munoblots of the effects of apoE4 and apoE3 on the total and on the phosphorylation levels of ERK in the hippocampus are presented in the
panel on the left. Quantification of the total hippocampal ERK levels and activation, as presented by the ratio of phosphorylated JNK to its
total level, are presented in the central and right panels. 0.978 +/-0.16 and 1.2+\- 0.18 for the ratio of apoE4 and apoE3 mice, respectively;
n=10-12 per group). ApoE3 mice are depicted in the white bar and the corresponding apoE4 mice are depicted in the black bars. Beta-tubulin
was used as a loading reference and results presented are normalized relative to the apoE3. Similar results were obtained with another cohort
of such mice.
286 Current Alzheimer Research, 2019, Vol. 16, No. 4 Salomon-Zimri et al.

Fig. (4). The effects of apoE4 on the Akt and GSK levels and activation. Hippocampi of 4-month-old female apoE3 and apoE4 TR mice were
excised, homogenized, and subjected to phosphorylated and total Akt (A) and GSK-3β(B) immunoblotting , as described in Materials and
Methods. (A) Representative immunoblots of the effects of apoE4 and apoE3 on the total levels and phosphorylation of Akt in the hippocam-
pus are presented in the left panel. Quantification of the total hippocampal Akt levels in these mice and of the activated Akt levels, as pre-
sented by the ratio of phosphorylated Akt to its total level, are presented in the central and right panels (0.6 +/-0.1 and 0.93+\- 0.11 for the
ratio of apoE4 and apoE3 mice, respectively). (B) Representative immunoblots of the effects of apoE4 and apoE3 on the total levels and
phosphorylation of GSK3β in the hippocampus, as presented by the ratio of phosphorylated GSK3β to its total level, are presented in the
panel on the left. Quantitation of the total hippocampal c-Jun levels in these mice and of the activated GSK3β to the total GSK3β levels are
presented in in the central and right panels (1.8 +/-0.2 and 1+\- 0.03 for the ratio of apoE4 and apoE3 mice, respectively). ApoE3 mice are
depicted in the white bar and the corresponding apoE4 mice are depicted in the black bars. Beta-tubulin was used as a loading reference and
the results presented are normalized relative to the apoE3 **p<0.01 and ***p<0.001 by student’s t-test. Similar results were obtained with
another cohort of such mice.

Fig. (5). The effects of AAV-VEGF treatment on the p38 activation of apoE4 and apoE3 mice. Hippocampi of 4-month-old naïve, sham, and
AAV-VEGF-treated female apoE3 and apoE4 TR mice were excised, homogenized, and subjected to phosphorylated and total p-38 im-
munoblotting , as described in Materials and Methods . Representative immunoblots of total p38 and the corresponding phosphorylated p38
of naïve, sham, and AAV-VEGF-treated apoE4 and apoE3 mice, as presented by the ratio of phosphorylated p38 to its total level, are pre-
sented in the panel on the left. Quantitation of the activated p38 levels is presented in the right panels. ApoE3 mice are depicted in the white
bar and the corresponding apoE4 mice are depicted in the black bars. Beta-tubulin was used as a loading reference and the results presented
are normalized relative to apoE3 and are presented on the left. Quantification of these results revealed significant effects for the group,
***p<0.0001, and *p<0.05 for group x treatment, by 2-way ANOVA; Further Bonnferoni post-hoc analysis showed #p<0.05 for the specific
effect of VEGF treatment on apoE4 mice. n= 5-7 per group.
The Role of MAPK's Signaling in Mediating ApoE4-Driven Current Alzheimer Research, 2019, Vol. 16, No. 4 287

Fig. (6). The effects of AAV-VEGF treatment on JNK and c-Jun activation of apoE4 and apoE3 mice. Hippocampi of 4-month-old naïve,
sham, and AAV-VEGF-treated female apoE3 and apoE4 TR mice were excised, homogenized, and subjected to phosphorylated and total
JNK (A) and (B) c-Jun immunoblotting , as described in Materials and Methods . (A) Representative immunoblots of total JNK and the corre-
sponding phosphorylated JNK of naïve, sham, and AAV-VEGF-treated apoE4 and apoE3 mice in the hippocampus, as presented by the ratio
of phosphorylated JNK to its total level, are presented in the panel on the left. Quantification of these results revealed significant effects for
the group, **p<0.01, and *p<0.05 for treatment, by 2-way ANOVA. (B) Representative hippocampal immunoblots of total c-Jun and the cor-
responding phosphorylated c-Jun of naïve, sham, and AAV-VEGF-treated apoE4 and apoE3 mice in the hippocampus, as presented by the
ratio of phosphorylated c-Jun to its total level, are presented in the panel on the left. Quantification of these results revealed significant effects
for the group, *p<0.05 by 2-way ANOVA. ApoE3 mice are depicted in the white bar and the corresponding apoE4 mice are depicted in the
black bars. Beta-tubulin was used as a loading reference and the results presented are normalized relative to the apoE3 and are presented on
the left. n= 5-7 per group.

Fig. (7). The effects of AAV-VEGF treatment on ERK activation of apoE4 and apoE3 mice. Hippocampi of 4-month-old naïve, sham, and
AAV-VEGF-treated female apoE3 and apoE4 TR mice were excised, homogenized, and subjected to phosphorylated and total ERK im-
munoblotting , as described in Materials and Methods .Representative hippocampal immunoblots of total ERK and the corresponding phos-
phorylated ERK of naïve, sham, and AAV-VEGF-treated apoE4 and apoE3 mice in the hippocampus, as presented by the ratio of phosphory-
lated c-Jun to its total level, are presented in the panel on the left. Quantification of these results showed no significant effect by 2-way
ANOVA. n= 5-7 per group.
288 Current Alzheimer Research, 2019, Vol. 16, No. 4 Salomon-Zimri et al.

reduced following AAV-VEGF treatment and became non- were accompanied by decreased activation of Akt, followed
significant (Fig. 8). Quantification of these results revealed by hyperphosphorylation of GSK3β and tau. In contrast, the
significant effects for the group, *p<0.05, by 2-way ANOVA. ERK pathway was not affected by apoE4. We have previ-
ously shown that apoE4 is associated with reduced levels of
In conclusion, the present results show that apoE4 is as-
hippocampal VEGF, and that reversal of this effect by intra-
sociated with specific activation of the p38 and JNK MAPK
hippocampal injection of an AAV-VEGF virus reverses the
pathways with the corresponding suppression of Akt signal-
synaptic and AD-related pathological effects of apoE4 [38].
ing pathway, and suggest that the p38 and Akt pathways are
associated with the apoE4-driven synaptic pathologies as Applying this therapeutic assay to the apoE4-driven effects
on MAPK activities revealed that apoE4-driven activation of
well as with the Aβ and tau-related pathological effects in
p38 and inhibition of Akt were reversed by AAV-VEGF
vivo.
treatment. The mechanism by which apoE4 induces the
down-regulation of VEGF is not directly addressed in this
4. DISCUSSION study. However, it should be noted that apoE4 down-
This study investigated the effects of apoE4 on the acti- regulates the levels of numerous different receptors includ-
vation patterns of different MAPK pathways, and the possi- ing the NMDA receptor [42], the apoER2 receptor [37], and
ble role of these pathways in mediating the pathological ef- the Insulin receptor [43], and targets them for degradation
fects of apoE4 in vivo. This was pursued by measuring the [43-45]. It is thus possible that the apoE4-related reduction
total levels and extent of activation of distinct MAPK-related in VEGF levels is driven by the reduced levels of VEGF
kinases in the hippocampus of apoE4 and the corresponding receptors [38]. Additionally, the apoE4 induced effect on the
apoE3 TR mice. This revealed significant apoE4-driven acti- JNK pathway became non-significant following the AAV-
vation and enhanced phosphorylation of p38 in the hippo- VEGF manipulation, and the ERK signaling, which was not
campus of the apoE4 mice. A similar activation pattern was affected by apoE4, was also not affected by the AAV-VEGF
seen in JNK and its transcription factor c-Jun. These effects treatment. A schematic summary of these effects is presented

Fig. (8). The effects of AAV-VEGF treatment on Akt and GSK3β activation of apoE4 and apoE3 mice. Hippocampi of 4-month-old naïve,
sham, and AAV-VEGF-treated female apoE3 and apoE4 TR mice were excised, homogenized, and subjected to phosphorylated and total (A)
Akt and (B) GSK-3β immunoblotting , as described in Materials and Methods . (A) Representativeimmunoblots of total Akt and the corre-
sponding phosphorylated Akt of naïve, sham, and AAV-VEGF-treated apoE4 and apoE3 mice in the hippocampus, as presented by the ratio
of phosphorylated Akt to its total level, are presented in the panel on the left. Quantification of these results revealed significant effects for
the group, *p<0.05 by 2-way ANOVA. (B) Representative immunoblots of total GSK3β and the corresponding phosphorylated GSK3β of
naïve, sham, and AAV-VEGF-treated apoE4 and apoE3 mice in the hippocampus, as presented by the ratio of phosphorylated GSK3β to its
total level, are presented in the panel on the left. Quantification of these results revealed significant effects for the group, *p<0.05 by 2-way
ANOVA. ApoE3 mice are depicted in the white bar and the corresponding apoE4 mice are depicted in the black bars. Beta-tubulin was used
as a loading reference and the results presented are normalized relative to the apoE3 and are presented on the left. n= 5-7 per group.
The Role of MAPK's Signaling in Mediating ApoE4-Driven Current Alzheimer Research, 2019, Vol. 16, No. 4 289

in (Fig. 9). Similar results were obtained with the ing, what is the process through which VEGF counteracts the
corresponding control experiment using male mice (not effects of apoE4 on MAPK signaling, and how do the
shown). The present findings that apoE4 is associated with apoE4-driven effects on the MAPK system lead to the down-
decreased activation of Akt and increased activation of c-Jun stream apoE4-driven pathology? The Akt MAPK pathway is
and JNK is in accordance with previous findings obtained regulated and triggered by growth factors [50, 51]. Accord-
with apoE4 x APP double transgenic mice and with apoE- ingly, it is possible that the presently observed down-
expressing cells [46]. regulation of this pathway in apoE4 brains is driven by the
corresponding reduced levels of brain VEGF [38] or includ-
The specific role of the kinases in the apoE4-related pa-
ing osmotic shock, inflammatory cytokines, LPS, UV, endo-
thology is remained to be determined. Though, it has been
plasmic reticulum stress, mitochondrial dysfunction, and
shown that p38 MAPK, could play more than one role in AD
pathophysiology, in microglia p38 MAPK contributes to impaired immune function [16, 17, 46, 52, 53]. Since apoE4
induces numerous pathways of cellular stress including oxi-
brain inflammation in AD, in astrocytes the p38 MAPK cas-
dative and endoplasmic reticulum stress [52], it is possible
cade also modulates the excitotoxicity, whereas the neuronal
that the effects of apoE4 on JNK and p38 are indirect and
p38 MAPK signaling contributes to tau phosphorylation,
result from the apoE4-induced stress.
NFT formation and depression of synaptic plasticity [19, 47].
Nevertheless, the specific effects of the apoE genotype on With regard to the second question, the mechanisms by
p38 signaling have not yet been reported. In vitro studies which VEGF counteracts the effects of apoE4 on MAPK
utilizing primary neurons [48] and astrocytic apoE3 and pathway and the specific cell type involved in the pathology
apoE4 embryonic stem cells [49] revealed that unlike the are not yet known. However, based on the results, we can
present in vivo findings (Fig. 3), ERK signaling is activated hypothesize that VEGF alters apoE4 related signaling by two
by apoE4. This may be due to the experimental differences mechanisms; a direct mechanism by which the effect of
between the in vivo and in vitro systems. VEGF is driven by the down-regulation of the downstream
signaling cascade, and an indirect mechanism by which
The mechanisms underlying the present findings will be
apoE4 and the decreased levels of VEGF induce cellular
addressed by focusing on three questions. Namely, which
stress that results in activation of p38 and JNK pathways.
mechanisms underlie the effects of apoE4 on MAPK signal-
Examination of the direct pathway shows that lower levels of

Fig. (9). Summary of the effects of apoE4 on key MAPK signaling pathways. Schematic presentation of the effect of ApoE4 on MAPK and
may play a role in mediating the downstream pathological effects. The figure depicts the four MAPK pathways in which the kinases exam-
ined depicted in a filled elliptic cycle. Arrows indicate the proteins whose activation is modulated by apoE4; up for activation and down for
inhibition, whereas an asterisk denotes the signaling molecules that may play a role in mediating the apoE4-driven pathology. This summary
is based on the results shown in Figs. (1-8).
290 Current Alzheimer Research, 2019, Vol. 16, No. 4 Salomon-Zimri et al.

VEGF are associated with lower Akt phosphorylation, lead- Increased p38 activation has been reported in human AD
ing to hyper-phosphorylation of GSK-3β which may also brain tissue, as well as in AD-relevant animal models and the
lead in turn to tau phosphorylation. Furthermore, upregu- corresponding cell culture systems [18, 19, 47, 57]. Further-
lation of this pathway by elevating VEGF levels alters all more, p38 activation has been linked to increased production
detected downstream phenotypes, which leads to reversal of of pro-inflammatory cytokines by glia activated with human
tau pathology (as observed in Salomon-Zimri et al. 2016) amyloid-beta (Aβ), as well as other diverse AD-related
[36]. The indirect pathway, on the other hand, shows hyper- events such as neurotoxicity and synaptic dysfunction [19,
activation of the stress related markers, namely p-38 and 58]. It is thus possible that the apoE4-driven increase in p38
JNK/c-Jun in apoE4 mice, may be driven by various types of plays a role in the apoE4-driven synaptic impairments and in
apoE4-induced cellular stress such as oxidative, genotoxic, the accumulation of intra-neuronal Aβ as well as astrocytic
mitochondrial dysfunction and osmotic stress, as well as pro- activation [15, 36]. The finding that the apoE4-driven activa-
inflammatory cytokines and extracellular environment stress tion of JNK pathway is not reversed by VEGF treatment,
[54]. Up-regulation of VEGF by the AAV-VEGF treatment whereas the synaptic and AD-related pathological effects of
which reverses the p38 related pathology and reduces the apoE4 are, suggest that this signaling pathway does not play
apoE4 related effect on the JNK/c-Jun pathway possibly by a major role in the apoE4 phenotypes identified to date. The
the contribution of the up-regulation of VEGF to reduce the extent to which JNK mediates yet to be identified pathologi-
stress related processing. A schematic summary of the di- cal effects of apoE4 remains to be determined.
rect/indirect hypothesis is presented in (Fig. 10).
Overall, the present study suggests a possible role of
The processes through which apoE4 reduces Akt levels MAPK's related signaling in mediating the apoE4 pathology.
and increases p38 activity, mediating the synaptic and AD- The fact that diverse treatments such as ABCA1 agonist,
related pathological effects of apoE, are not fully understood. AAV-VEGF virus, and an anti-apoE4 Ab, all reversed both
The finding that the apoE4-driven inhibition of Akt is asso- the cognitive impairments and synaptic loss, suggest that
ciated with increased activation of GSKβ (Fig. 8) and tau there may be correlation between the two [38, 59, 60]. This
hyperphosphorylation, as obtained previously by Salomon- observation, together with previous in-vitro studies, in which
Zimri et al. [38], is in accordance with findings obtained a causal effect of apoE4 and synapse function has been
with young AD apoE4 carriers at lower Braak stages [55]. shown, suggest that the pathological effect of apoE4 on cog-
These findings suggest that the cytoskeletal and tau pathol- nitive impairment may be related to synaptic loss [61, 62].
ogy observed in the apoE4 mice [36] may be driven via the Since the MAPK pathways were shown to be involved in the
inhibition of Akt. Since Akt signaling has been implicated in pathophysiology and pathogenesis of AD [18, 19, 24] and
numerous other cellular activities such as growth, survival, specifically in synaptic dysfunction [27], we have strong
and migration [56], it is also possible that the Akt-related reason to assume that the MAPKs are involve in the apoE4-
pathological effects of apoE4 are mediated by these systems. related AD-like pathology.

Fig. (10). The direct/indirect hypothesis. A model of the direct/indirect mechanisms underling the role of VEGF in mediating the patho-
logical effects of apoE4.
The Role of MAPK's Signaling in Mediating ApoE4-Driven Current Alzheimer Research, 2019, Vol. 16, No. 4 291

CONCLUSION [3] Masliah E, Crews L, Hansen L. Synaptic remodeling during aging


and in Alzheimer's disease. J Alzheimer's Dis 9(3 Suppl): 91-9
The present study shows that apoE4 is associated with (2006).
the specific activation of the stress-related p38 and JNK [4] Schwartz M, Deczkowska A. Neurological disease as a failure of
brain-immune crosstalk: the multiple faces of neuroinflammation.
MAPK pathways, and the inhibition of the corresponding Trends Immunol 37(10): 668-79 (2016).
growth factor-related Akt pathways in the hippocampus. The [5] Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell
synaptic and AD-related pathological effects of apoE4 are PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele
counteracted by reversal of the p38 and Akt-related signaling and the risk of Alzheimer's disease in late onset families. Science
effects of apoE4, suggesting that these MAPK pathways (New York, NY 261(5123): 921-3 (1993).
[6] Roses AD. Apolipoprotein E alleles as risk factors in Alzheimer's
could serve as possible apoE4-related therapeutic targets. disease. Ann Rev Med 47: 387-400 (1996).
The mechanism underlying the observed effect of apoE4 [7] Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PH,
Pericak-Vance MA, Joo SH, et al. Association of apolipoprotein E
on these growth factors is unknown, although it is interesting allele epsilon 4 with late-onset familial and sporadic Alzheimer's
to note that apoE4 has important effects on numerous recep- disease. Neurology 43(8): 1467-72 (1993).
tors such as VEGF-R2, ApoER2, and NMDA of which [8] Liu DS, Pan XD, Zhang J, Shen H, Collins NC, Cole AM, et al.
apoE4 is a down-regulator, presumably by directing them to APOE4 enhances age-dependent decline in cognitive function by
down-regulating an NMDA receptor pathway in EFAD-Tg mice.
lysosomal degradation. Accordingly, it is possible that the Mol Neurodegen 10: 7 (2015).
effects of apoE4 on these growth factor-related pathways are [9] Schindler S, Gratia P, Mullenberger G, Arendt J. Adenosarcoma
driven by the effect of apoE4 on the receptors, which in turn, and other uterine sarcomas. Bulletin de la Societe des sciences
results in decreased growth factor. medicales du Grand-Duche de Luxembourg 134(2): 27-30 (1997).
[10] Mahley RW, Huang Y. Apolipoprotein e sets the stage: response to
injury triggers neuropathology. Neuron 76(5): 871-85 (2012).
ETHICS APPROVAL AND CONSENT TO PARTICI- [11] Kutner KC, Erlanger DM, Tsai J, Jordan B, Relkin NR. Lower
PATE cognitive performance of older football players possessing
apolipoprotein E epsilon4. Neurosurgery 47(3): 651-7, 57-8 (2000).
These experiments were conducted under the approval of [12] Peila R, Rodriguez BL, Launer LJ, Honolulu-Asia Aging S. Type 2
the institutional Tel-Aviv University Animal Care Commit- diabetes, APOE gene, and the risk for dementia and related
tee (approval number: 04-14-022), and all animal rights were pathologies: the Honolulu-Asia aging study. Diabetes 51(4): 1256-
62 (2002).
strictly kept by ethical guidelines, Israel. [13] Michaelson DM. APOE epsilon4: the most prevalent yet
understudied risk factor for Alzheimer's disease. Alzheimer's
HUMAN AND ANIMAL RIGHTS Dementia: J Alzheimer's Assoc 10(6): 861-8 (2014).
[14] Ungar L, Altmann A, Greicius MD. Apolipoprotein E, gender, and
No humans were used in this study. All animal proce- Alzheimer's disease: an overlooked, but potent and promising
dures were followed in accordance with F16-00009 (A5010- interaction. Brain Imaging Behav 8(2): 262-73 (2014).
01) approval by Office of the Laboratory Animal Welfare [15] Bar R, Boehm-Cagan A, Luz I, Kleper-Wall Y, Michaelson DM.
The effects of apolipoprotein E genotype, alpha-synuclein
(OLAW) and the American National Research Council, deficiency, and sex on brain synaptic and Alzheimer's disease-
USA. related pathology. Alzheimers Dement (Amst) 10: 1-11 (2018).
[16] Kim EK, Choi EJ. Compromised MAPK signaling in human
CONSENT FOR PUBLICATION diseases: an update. Arch Toxicol 89(6): 867-82 (2015).
[17] Yu Q, Du F, Douglas JT, Yu H, Yan SS, Yan SF. Mitochondrial
Not applicable. dysfunction triggers synaptic deficits via activation of p38 map
kinase signaling in differentiated Alzheimer's disease trans-
mitochondrial cybrid cells. J Alzheimer's Dis 59(1): 223-39 (2017).
CONFLICT OF INTEREST [18] Zhu X, Lee HG, Raina AK, Perry G, Smith MA. The role of
mitogen-activated protein kinase pathways in Alzheimer's disease.
The authors declare no conflict of interest, financial or Neuro-Signals 11(5): 270-81 (2002).
otherwise. [19] Munoz L, Ammit AJ. Targeting p38 MAPK pathway for the
treatment of Alzheimer's disease. Neuropharmacology 58(3): 561-8
(2010).
ACKNOWLEDGEMENTS [20] Hensley K, Floyd RA, Zheng NY, Nael R, Robinson KA, Nguyen
We thank Alex Smolar for his technical assistance and X, et al. p38 kinase is activated in the Alzheimer's disease brain. J
Neurochem 72(5): 2053-58 (1999).
Dr. Ori Liraz for his scientific counseling. This research was [21] Cuenda A, Rousseau S. p38 MAP-kinases pathway regulation,
supported in part by grants from the Israel Science Founda- function and role in human diseases. Biochimica et Biophysica
tion (grant No. 794/17), from the Joseph K. and Inez Acta 1773(8): 1358-75 (2007).
Eichenbaum Foundation, from the Harold and Eleanore [22] Mehan S, Meena H, Sharma D, Sankhla R. JNK: a stress-activated
protein kinase therapeutic strategies and involvement in
Foonberg Foundation, and from the Joseph Sagol fellowship Alzheimer's and various neurodegenerative abnormalities. J Mol
program for brain research. Neurosci 43(3): 376-90 (2011).
[23] Hoe HS, Harris DC, Rebeck GW. Multiple pathways of
REFERENCES apolipoprotein E signaling in primary neurons. J Neurochem 93(1):
145-55 (2005).
[1] Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, [24] Okazawa H, Estus S. The JNK/c-Jun cascade and Alzheimer's
Beyreuther K. Amyloid plaque core protein in Alzheimer disease disease. Am J Alzheimer's Dis Other Demen 17(2): 79-88 (2002).
and Down syndrome. Proc Nat Acad Sci USA 82(12): 4245-9 [25] Takashima A. GSK-3 is essential in the pathogenesis of
(1985). Alzheimer's disease. J Alzheimer's Dis 9(3 Suppl): 309-17 (2006).
[2] Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An [26] Zhang Y, Huang NQ, Yan F, Jin H, Zhou SY, Shi JS, et al.
English translation of Alzheimer's 1907 paper, "Uber eine Diabetes mellitus and Alzheimer's disease: GSK-3beta as a
eigenartige Erkankung der Hirnrinde". Clin Anat 8(6): 429-31 potential link. Behav Brain Res 339: 57-65 (2018).
(1995). [27] Morroni F, Sita G, Tarozzi A, Rimondini R, Hrelia P. Early effects
of Abeta1-42 oligomers injection in mice: involvement of
292 Current Alzheimer Research, 2019, Vol. 16, No. 4 Salomon-Zimri et al.

PI3K/Akt/GSK3 and MAPK/ERK1/2 pathways. Behav Brain Res [45] Chen Y, Durakoglugil MS, Xian X, Herz J. ApoE4 reduces
314: 106-15 (2016). glutamate receptor function and synaptic plasticity by selectively
[28] Sun J, Nan G. The extracellular signal-regulated kinase 1/2 impairing ApoE receptor recycling. Proc Nat Acad Sci USA
pathway in neurological diseases: a potential therapeutic target 107(26): 12011-6 (2010).
(Review). Intern J Mol Med 39(6): 1338-46 (2017). [46] Cash JG, Kuhel DG, Basford JE, Jaeschke A, Chatterjee TK,
[29] Kirouac L, Rajic AJ, Cribbs DH, Padmanabhan J. Activation of Weintraub NL, et al. Apolipoprotein E4 impairs macrophage
Ras-ERK signaling and GSK-3 by amyloid precursor protein and efferocytosis and potentiates apoptosis by accelerating endoplasmic
amyloid beta facilitates neurodegeneration in Alzheimer's disease. reticulum stress. J Biol Chem 287(33): 27876-84 (2012).
eNeuro 4(2): (2017). [47] Pei JJ, Braak E, Braak H, Grundke-Iqbal I, Iqbal K, Winblad B, et
[30] Gan X, Wu L, Huang S, Zhong C, Shi H, Li G, et al. Oxidative al. Localization of active forms of C-jun kinase (JNK) and p38
stress-mediated activation of extracellular signal-regulated kinase kinase in Alzheimer's disease brains at different stages of
contributes to mild cognitive impairment-related mitochondrial neurofibrillary degeneration. J Alzheimer's Dis 3(1): 41-48 (2001).
dysfunction. Free Rad Biol Med 75: 230-40 (2014). [48] Hoe HS, Pocivavsek A, Dai H, Chakraborty G, Harris DC, Rebeck
[31] Lau D, Bengtson CP, Buchthal B, Bading H. BDNF reduces toxic GW. Effects of apoE on neuronal signaling and APP processing in
extrasynaptic nmda receptor signaling via synaptic NMDA rodent brain. Brain Res 1112(1): 70-9 (2006).
receptors and nuclear-calcium-induced transcription of [49] Lee KI, Su CC, Yang CY, Hung DZ, Lin CT, Lu TH, et al.
inhba/activin A. Cell reports 12(8): 1353-66 (2015). Etoposide induces pancreatic beta-cells cytotoxicity via the
[32] Maezawa I, Maeda N, Montine TJ, Montine KS. Apolipoprotein E- JNK/ERK/GSK-3 signaling-mediated mitochondria-dependent
specific innate immune response in astrocytes from targeted apoptosis pathway. Toxicol In vitro: an international journal
replacement mice. J Neuroinflammation 3: 10 (2006). published in association with BIBRA 36: 142-52 (2016).
[33] DeKroon R, Robinette JB, Hjelmeland AB, Wiggins E, Blackwell [50] Song G, Ouyang G, Bao S. The activation of Akt/PKB signaling
M, Mihovilovic M, et al. APOE4-VLDL inhibits the HDL- pathway and cell survival. J Cell Mol Med 9(1): 59-71 (2005).
activated phosphatidylinositol 3-kinase/Akt Pathway via the [51] Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, et
phosphoinositol phosphatase SHIP2. Circ Res 99(8): 829-36 al. Impaired insulin and insulin-like growth factor expression and
(2006). signaling mechanisms in Alzheimer's disease--is this type 3
[34] Ong QR, Chan ES, Lim ML, Cole GM, Wong BS. Reduced diabetes? J Alzheimer's Dis 7(1): 63-80 (2005).
phosphorylation of brain insulin receptor substrate and Akt proteins [52] Dose J, Huebbe P, Nebel A, Rimbach G. APOE genotype and
in apolipoprotein-E4 targeted replacement mice. Sci Rep 4: 3754 stress response - a mini review. Lipids Health Dis 15: 121 (2016).
(2014). [53] Dorey E, Bamji-Mirza M, Najem D, Li Y, Liu H, Callaghan D, et
[35] Salomon-Zimri S, Boehm-Cagan A, Liraz O, Michaelson DM. al. Apolipoprotein E isoforms differentially regulate alzheimer's
Hippocampus-related cognitive impairments in young apoE4 disease and amyloid-beta-induced inflammatory response in vivo
targeted replacement mice. Neuro-degenerative Dis 13(2-3): 86-92 and in vitro. J Alzheimer's Dis 57(4): 1265-79 (2017).
(2014). [54] Kusaka G, Ishikawa M, Nanda A, Granger DN, Zhang JH.
[36] Liraz O, Boehm-Cagan A, Michaelson DM. ApoE4 induces Signaling pathways for early brain injury after subarachnoid
Abeta42, tau, and neuronal pathology in the hippocampus of young hemorrhage. J Cerebral Blood Flow Metabol 24(8): 916-25 (2004).
targeted replacement apoE4 mice. Mol Neurodegen 8: 16 (2013). [55] Simpson JE, Ince PG, Shaw PJ, Heath PR, Raman R, Garwood CJ,
[37] Gilat-Frenkel M, Boehm-Cagan A, Liraz O, Xian X, Herz J, et al. Microarray analysis of the astrocyte transcriptome in the
Michaelson DM. Involvement of the Apoer2 and Lrp1 receptors in aging brain: relationship to Alzheimer's pathology and APOE
mediating the pathological effects of ApoE4 in vivo. Curr genotype. Neurobiol Aging 32(10): 1795-807 (2011).
Alzheimer Res 11(6): 549-57 (2014). [56] Majewska E, Szeliga M. AKT/GSK3beta Signaling in
[38] Salomon-Zimri S, Glat MJ, Barhum Y, Luz I, Boehm-Cagan A, Glioblastoma. Neurochem Res 42(3): 918-24 (2017).
Liraz O, et al. Reversal of ApoE4-driven brain pathology by [57] Sun A, Liu M, Nguyen XV, Bing G. P38 MAP kinase is activated
vascular endothelial growth factor treatment. J Alzheimer's Dis at early stages in Alzheimer's disease brain. Exp Neurol 183(2):
53(4): 1443-58 (2016). 394-405 (2003).
[39] Liu Y, Deisseroth A. Tumor vascular targeting therapy with viral [58] Lee JK, Kim NJ. Recent advances in the inhibition of p38 MAPK
vectors. Blood 107(8): 3027-33 (2006). as a potential strategy for the treatment of Alzheimer's disease.
[40] Hermida MA, Dinesh Kumar J, Leslie NR. GSK3 and its Molecules 22(8): (2017).
interactions with the PI3K/AKT/mTOR signalling network. Adv [59] Boehm-Cagan A, Bar R, Liraz O, Bielicki JK, Johansson JO,
Biol Regul 65: 5-15 (2017). Michaelson DM. ABCA1 Agonist reverses the ApoE4-driven
[41] Hooper C, Killick R, Lovestone S. The GSK3 hypothesis of cognitive and brain pathologies. J Alzheimer's Dis 54(3): 1219-33
Alzheimer's disease. J Neurochem 104(6): 1433-9 (2008). (2016).
[42] Lane-Donovan C, Herz J. ApoE, ApoE receptors, and the synapse [60] Luz I, Liraz O, Michaelson DM. An Anti-apoE4 specific
in Alzheimer's disease. Trends Endocrinol Metabol 28(4): 273-84 monoclonal antibody counteracts the pathological effects of apoE4
(2017). in vivo. Curr Alzheimer Res 13(8): 918-29 (2016).
[43] Zhao N, Liu CC, Van Ingelgom AJ, Martens YA, Linares C, [61] Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in
Knight JA, et al. Apolipoprotein E4 impairs neuronal insulin Alzheimer's disease. Neuron 63(3): 287-303 (2009).
signaling by trapping insulin receptor in the endosomes. Neuron [62] Sun GZ, He YC, Ma XK, Li ST, Chen DJ, Gao M, et al.
96(1): 115-29 e5 (2017). Hippocampal synaptic and neural network deficits in young mice
[44] Zhao N, Liu CC, Qiao W, Bu G. Apolipoprotein E, receptors, and carrying the human APOE4 gene. CNS Neurosci Therap 23(9):
modulation of Alzheimer's disease. Biol Psychiat 83(4): 347-57 748-58 (2017).
(2018).

You might also like