Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

J Mol Neurosci

DOI 10.1007/s12031-015-0665-8

Liver X Receptor Agonist Modifies the DNA Methylation Profile


of Synapse and Neurogenesis-Related Genes in the Triple
Transgenic Mouse Model of Alzheimer’s Disease
A. G. Sandoval-Hernández 1 & H. G. Hernández 4 & A. Restrepo 1 & J. I. Muñoz 2 &
G. F. Bayon 3 & A. F. Fernández 3 & M. F. Fraga 3 & G. P. Cardona-Gómez 2 & H. Arboleda 4 &
Gonzalo H. Arboleda 1,5

Received: 19 March 2015 / Accepted: 8 October 2015


# Springer Science+Business Media New York 2015

Abstract The liver X receptor agonist, GW3965, improves These changes in methylation probes include 29 gene loci.
cognition in Alzheimer’s disease (AD) mouse models. Here, Importantly, changes in methylation status were mainly from
we determined if short-term GW3965 treatment induces chang- synapse-related genes (SYP, SYN1, and DLG3) and
es in the DNA methylation state of the hippocampus, which are neurogenesis-associated genes (HMGB3 and RBBP7). Thus,
associated with cognitive improvement. Twenty-four-month- our results indicate that liver X receptors (LXR) agonist treat-
old triple-transgenic AD (3xTg-AD) mice were treated with ment induces rapid changes in DNA methylation, particularly
GW3965 (50 mg/kg/day for 6 days). DNA methylation state in loci associated with genes involved in neurogenesis and syn-
was examined by modified bisulfite conversion and hybridiza- aptic function. Our results suggest a new potential mechanism
tion on Illumina Infinium Methylation BeadChip 450 k arrays. to explain the beneficial effect of GW3965.
The Morris water maze was used for behavioral analysis. Our
results show in addition to improvement in cognition methyla- Keywords Alzheimer’s disease . Liver X receptors . Triple
tion changes in 39 of 13,715 interrogated probes in treated transgenic mice . GW3965
3xTg-AD mice compared with untreated 3xTg-AD mice.

Introduction
A. G. Sandoval-Hernández and H. G. Hernández contributed equally to
this work. Alzheimer’s disease (AD) is an age-dependent neurodegenera-
tive disorder and a major public health problem. The hippocam-
Electronic supplementary material The online version of this article
(doi:10.1007/s12031-015-0665-8) contains supplementary material, pus is involved in learning and memory and is the earliest
which is available to authorized users. affected site in AD (Braak et al. 1993). The dentate gyrus of
the hippocampus plays an essential role in normal hippocampal
* Gonzalo H. Arboleda function throughout life, and is one of the main areas for adult
gharboledab@unal.edu.co neurogenesis, which is affected in AD (Ming and Song 2011).
DNA methylation is an important epigenetic modification
1
Grupo de Muerte Celular, Instituto de Genética, Universidad of the genome that occurs on cytosine residues at carbon 5 of
Nacional de Colombia, Bogotá, Colombia the pyrimidine ring of simple sequences termed CpG dinucle-
2
Área de Neurobiología Celular y Molecular, Grupo de Neurociencias otides and subsequently controls gene expression. DNA meth-
de Antioquia, Universidad de Antioquia, Medellín, Colombia ylation at CpG dinucleotides and 2 kb upstream and down-
3
Cancer Epigenetics Laboratory, Institute of Oncology of Asturias stream from CpG dinucleotides is strongly associated with
(IUOPA), Hospital Universitario Central de Asturias (HUCA), stable transcriptional repression (Jones 2012). Thus, DNA
Universidad de Oviedo, Oviedo, Spain methylation controls expression of many genes and is in-
4
Grupo de Neurociencias, Universidad Nacional, Bogotá, Colombia volved in important cellular processes including embryonic
5
Departamento de Patología, Facultad de Medicina, Universidad development, chromatin structure, and X chromosome inacti-
Nacional de Colombia, Bogotá, Colombia vation. Recently, a growing number of reports have shown
J Mol Neurosci

aberrant changes in DNA methylation patterns in many hu- Materials and Methods
man diseases (Coppieters et al. 2014). In the human AD brain,
two independent analyses of genomic DNA methylation (the Animals Triple-transgenic AD (3xTg-AD) mice (Oddo et al.
Bmethylome^) showed variations in the methylation pattern 2003) and wild-type (WT) mice from the house colony from
compared with controls, which correlated with the pathologi- the species pathogen-free animal facility of Sede de
cal stage of the disease (De Jager et al. 2014; Masliah et al. Investigación Universitaria at the University of Antioquia
2013). In addition, similar changes in the methylation pattern (Medellín-Colombia) were used. Animals were kept in a 12-
were also observed in the brain of AD mouse models h dark/light cycle and received food and water ad libitum. A
(Hernandez et al. 2014; Sanchez-Mut et al. 2013; Barrachina total of 24 3xTg-AD mice (24-month-old) and 7 WT mice
and Ferrer 2009). (24-month-old) were used. Female animals were randomly
Liver X receptors (LXR) belong to the nuclear receptor distributed within the groups. Animals were treated orally
superfamily that regulates DNA transcription (Wojcicka everyday with the LXR agonist GW3965 (50 mg/kg) or with
et al. 2007). Transcriptional activity of LXR is regulated by vehicle (dimethyl sulfoxide; 20 mg/mL) for six consecutive
binding to its natural ligands, most of which are oxygenated days, in groups of five animals in regular cages. Animals
metabolites of cholesterol (i.e., oxysterols) (Mitro et al. 2007), were handled following the Colombian (Law 84 of 1989
and by interaction with additional co-regulatory proteins. and Resolution 8430 of 1993) and international regulations
Some of these co-regulatory proteins directly regulate epige- and standards on animal welfare and conformed to the An-
netic changes, including the SWItch/Sucrose Non-Fermentable imal Research: Reporting In Vivo Experiments (ARRIVE)
chromatin-remodeling factors that induce changes in DNA guidelines (Kilkenny et al. 2010).
methylation and promote transcriptional activation (Kim et al.
2012; O’Malley et al. 2008; Rosenfeld et al. 2006; Banine et al. Morris Water Maze Hippocampal-dependent spatial memo-
2005). However, nuclear receptor function, and in particular ry was examined using the water maze paradigm (Morris et al.
their control over DNA methylation during brain development, 1982). The apparatus consisted of a white pool (100 cm di-
in adulthood, and their impact on human diseases remain poor- ameter and 54 cm high). The tank was filled to a depth of
ly understood (Otaegui-Arrazola et al. 2010). LXR can be 35 cm with opaque water with non-toxic white gouache paint,
pharmacologically modulated by synthetic agonists such as which remained at a constant temperature (22 ± 2 °C). The
GW3965, a selective agonist for LXRα (EC50 = 190 nM) platform (10 cm diameter) was located 1.5 cm below the
and LXRβ (EC50 = 30 nM) (Collins et al. 2002) that can water surface during spatial learning and 1 cm above the
induce LXR activation in the brain because of its ability to water surface during the visible session. Extra-maze visual
cross the blood–brain barrier (Xu et al. 2013; Donkin et al. cues around the room remained in fixed positions through-
2010; Fitz et al. 2010). out the experiment.
Recently, LXR activation was shown to improve cognitive
alterations based on its ability to regulate gene expression and, Initial Spatial Training The aim was for the mice to acquire a
in particular, apolipoprotein E (ApoE) and ATP-binding cas- learning task, in which they were trained to find the platform,
sette transporter (ABCA1), in mouse models of AD (Donkin within two daily sessions for 5 days. Each session consisted of
et al. 2010; Jiang et al. 2008). Moreover, LXR knockout mice four successive trials, and each trial began with the mouse
on an amyloidogenic mouse model background develop a placed pseudo-randomly in one of four starting positions. Be-
severe amyloid-β (Aβ) pathology (Zelcer et al. 2007), sug- fore the initial trial, the mice were trained to stay for 30 s on
gesting that LXR is an important mediator of Aβ metabolism the platform. Forty-eight hours after the learning phase, the
and a plausible therapy for AD. animals were tested for retention during a 90-s probe trial
To identify novel potential mechanisms that play a role in without the platform. During the probe trial, the latency to
cognitive improvement following LXR treatment in a murine reach the exact platform location was determined. To control
model of AD, we examined the effect of short-term treat- for any difference between experimental groups in visual–
ment of GW3965 in the triple transgenic mouse model of motor abilities or motivation, latencies to reach the platform
AD (3xTg-AD), using a microarray platform to compare were determined using a visible platform (four trials) at the
DNA methylation changes in the hippocampus of treated end of the retention phase. Behavior was recorded with an
and untreated mice. We found that cognitive improvement automated system (Viewpoint, Lyon, France).
conferred by the LXR agonist was associated with broad
changes in the DNA methylation pattern of 39 (of 13,715) DNA Extraction and DNA Methylation Microarrays Mice
interrogated probes in treated compared with untreated were anesthetized with ketamine/xylazine, the brains ex-
3xTg-AD mice. Interestingly, the most important changes tracted, manually dissected, and immediately frozen in liq-
were observed in synapse (SYP, SYN1, and CASK)- and uid nitrogen for DNA isolation using phenol-chloroform,
neurogenesis (HMGB3 and RBBP7)-related genes. as previously described (Masliah et al. 2013). To identify
J Mol Neurosci

global DNA methylation changes, the Illumina Infinium genotype group for the number of animals per group, as
450 k DNA Methylation assay (Illumina, San Diego, CA, indicated in the figure legends. For group comparisons,
USA) was performed in three 3xTg-AD mice that received statistical analysis was performed by analysis of variance
GW3965 and three mice that received vehicle. The 450 k with Bonferroni post test. Thioflavin-S (Thio-S) staining
protocol was performed at the Genome Quebec Facilities was assessed as described previously (Ly et al. 2011). In
(Montreal, Canada), according to the manufacturer’s pro- brief, slides were rinsed consecutively in ethanol (70 then
tocols and as previously described (Wong et al. 2013). In 80 % for 1 min each). Next, slides were rinsed with dis-
brief, DNA conversion was performed using the EZ DNA tilled water twice and covered with glycerol jelly. Images
Methylation Kit (Zymo Research Corp., Orange, CA, were captured by confocal fluorescence microscopy (×20
USA) and then subsequently hybridized to an Illumina magnification) (Nikon Eclipse C1 plus; Nikon Instruments
Infinium 450 k DNA Methylation BeadChip and scanned Inc., Melville, NY, USA).
(iScan, Illumina). Finally, the previously described adapta-
tions for data analysis of the mouse genome were used Western Blotting Following pharmacological treatment, all
(Wong et al. 2013). These adaptations are based on human animals received behavioral training and then the brains were
genome similarity and enable reliable evaluation of 13,715 extracted and manually dissected. Hippocampal tissue was
probes in a mouse using the Illumina Infinium 450 k DNA homogenized in 300 μL ice-cold RIPA buffer (Pierce Biotech-
Methylation assay (Wong et al. 2013). nology) containing complete protease inhibitor cocktail tab-
lets and phosphatase inhibitor cocktail tablets (Complete,
Histology of Aβ Plaques and Neurofibrillary Tangles Mice PhosSTOP; Roche Applied Science, Mannheim, Germany)
were anesthetized with ketamine/xylazine and perfused using a PRO Scientific homogenizer (Oxford, CT, USA) at
transcardially with 4 % paraformaldehyde in phosphate- full speed for 20 s. Homogenates were sonicated at 30 %
buffered saline, then the brains were removed and transferred output for 10 s, then centrifuged (12,500 g) at 4 °C for
into 30 % sucrose. Brains were cut into 50-μm coronal sec- 45 min in a microcentrifuge (Thermo Scientific). The protein
tions using a vibratome (Leica VT 1000S, Leica, Nussloch, concentration of supernatant fractions was determined using
Germany) and stored at −20 °C in anti-freezing medium until the BCA protein assay kit (Thermo Scientific) and 20 μg of
further processing. For diaminobenzidine (DAB) staining, protein samples run on polyacrylamide gels at 100 V. After
sections were pretreated for 20 min at room temperature in electrophoresis, proteins were transferred to polyvinylidene
0.1 M phosphate buffer (PB) (pH 7.4) with methanol (PB/ fluoride (PVDF) membranes (Amersham Hybond™-P; GE
methanol 1:1) and 1 % hydrogen peroxide (to block endoge- Healthcare, Buckinghamshire, UK) and incubated in a
nous peroxidase). Afterwards, sections were rinsed and incu- blocking buffer (5 % powder milk in TBS-Tween 20) for 1 h
bated for 1 h in 0.1 M PB with 1 % BSA and 0.3 % Triton at room temperature until probed. PVDF membranes were
X-100. Sections were incubated with primary antibodies in the then incubated with 5 mL anti-synapsin-I antibody, (1:1000,
same buffer overnight at 4 °C. Primary antibodies that rec- ab64581; Abcam, Cambridge, UK) in a blocking buffer over-
ognized phosphorylated tau at S202/205 in mice and night at 4 °C, washed three times in TBS-Tween (5 min/wash),
humans (Biernat et al. 1992) (AT8, 1:500; Thermo Scien- followed by incubation with a peroxidase conjugated second-
tific, Rockford, IL, USA) and 6E10 of human APP/Aβ ary antibody for 1 h (1:2000) and then washed three times
(SIG-39320; Signet/Covance, Dedham, MA, USA) were with TBS-Tween. Bound antibody was detected using the
used. Subsequently, sections were incubated with a bio- ECL system (Thermo Scientific).
tinylated mouse secondary antibody and then ABC–HRP
complex (Pierce Biotechnology; Rockford, IL, USA) for
2 h. Detection using DAB was performed. Sections were Results
dehydrated and covered with mounting solution and ob-
served using a zoom microscope (Axio-Zoom-V16, Zeiss, GW3965 Treatment Restores Cognitive Deficits
Munich, Germany). Images were acquired using the Zeiss in 3xTg-AD Mice
Axio Cam HRc digital color camera (1388 × 1040 pixels)
with ZEN pro software (Zeiss). For each captured image, We initially examined the effect of LXR agonist treatment on
semi-quantitative analysis of appropriate areas was per- cognition using the Morris water maze (WMW) task to deter-
formed by segmentation analysis using ImageJ software mine if short-term LXR activation contributes to improvement
(National Institutes of Health, Bethseda, MD, USA). Areas of spatial learning and memory in 3xTg-AD mice. In the
identified as positive for 6E10 or AT8 immunoreactivity learning task (Fig. 1a), WT mice performed significantly bet-
after thresholding were individually inspected to confirm ter than untreated 3xTg-AD mice on 5-day trials (P < 0.001).
the presence of plaques or hyperphosphorylated tau. The Treating 3xTg-AD mice with GW3965 resulted in no signif-
mean area value was averaged per treatment in each icant difference with WT. In the retention task, which was
J Mol Neurosci

Fig. 1 LXR agonist restores memory and cognition in 3xTg-AD mice. two-way analysis of variance (ANOVA). ***P < 0.001. b Retention tasks
Spatial learning and memory were examined using the MWM after 6 days by 4× analysis: time spent in the area near the platform (in sec). Signif-
of treatment with the LXR agonist GW3965. a Learning task: treated icant differences were detected by ANOVA followed by Bonferroni mul-
3xTg-AD mice took significantly less time to learn the location of the tiple comparison test
hidden platform. Statistically significant differences were detected by

analyzed in a 4× proximity design (Fig. 1b), WT animals spent identify differentially methylated probes. Beta mean bar
significantly more time near the platform than untreated 3xTg- plots did not show group differences in DNA methylation
AD (P < 0.001). Further, GW3965-treated 3xTg-AD mice patterns for the probes evaluated (Fig. 3c).
spent significantly more time near the platform than untreated To identify differentially methylated probes, we used the
3xTg-AD (P < 0.01) and were not significantly different from multiple testing correction, Benjamini–Hochberg false dis-
WT mice. These results indicate that in 3xTg-AD mice, covery rate, as well as a delta beta filter of 0.05 (Table 1).
GW3965 improves hippocampal-dependent behavior tested We detected 47 differentially methylated probes
by the MWM. (P < 0.0003) with P values corresponding to the false discov-
ery Benjamini–Hochberg correction (P < 0.1). We observed a
GW3965 Treatment Does Not Modify the Pathological decrease in DNA methylation for a specific subset of probes,
Hallmarks of AD as shown in the density plot (Fig. 3d). In addition, we exclud-
ed results with beta value differences less than 10 % (delta
To determine if LXR activation induces changes in amyloid beta filter of 0.1). Thus, overall, we identified 39 probes, cor-
plaque load and phosphorylated tau, and also if these changes responding to 29 genes, which were differentially methylated
are associated with the observed cognitive improvement, we (Table S1). Classification and function of each differentially
performed immunohistochemistry using DAB detection. No methylated gene were performed by an extensive PubMed
significant differences in positive immunoreactivity for anti- literature search. The differentially methylated genes were
Aβ (6E10) or anti-PHF1 (AT8) were observed between treat- found to be particularly related to synaptic function, prolifer-
ed and untreated 3xTg-AD mice (Fig. 2). ation, and neurogenesis (Tables 1 and 2).

DNA-Methylation Data Analysis GW3965 Treatment Restores Expression of Specific


Synapse-Related Genes
Hippocampal DNA was analyzed using Illumina Infinium
450 k DNA Methylation microarrays adjusted for the mouse We found that compared with untreated 3xTg-AD mice,
genome. A previous report found that only a subset of 13,715 GW3965 treatment of 3xTg-AD mice promotes significant
probes bind identically to genetic sequences within the mouse hypo-methylation of synapse-related genes (Table 1). These
genome (Wong et al. 2013). We used the minfi Bioconductor genes include synapsin-1, synaptophysin, and synapse-
R package (Aryee et al. 2014) to import and normalize raw associated protein 102 (SAP-102), expression of which has
data intensities. DNA methylation density plots in mouse previously been shown to be decreased in human AD brain
samples (Fig. 3a) showed the same distribution as in hu- (Proctor et al. 2010; Qin et al. 2004). Thus, we next deter-
man samples (Fig. 3b), as previously reported (Wong et al. mined if the DNA methylation changes observed following
2013). To obtain reliable intensities, we excluded probes GW3965 treatment of 3xTg-AD mice play a regulatory role in
with detection values of P > 0.001. After subset-quantile expression of two specific synaptic proteins: synapsin-1
within array normalization (SWAN) (Aryee et al. 2014), (shows decreased DNA-methylation) and postsynaptic densi-
robust Bayesian analysis was performed using the Limma ty protein 95 (PSD95) (no change in DNA methylation).
Bioconductor R package (Teschendorff et al. 2013) to GW3965 treatment significantly restored expression of
J Mol Neurosci

Fig. 2 GW3965 treatment does not produce detectable changes in anti-Aβ- or PHF1-positive areas in the hippocampus or cortex of treated
histopathological AD hallmarks. Amyloid plaque deposition and hyper- 3xTg-AD mice. No significant change in c Aβ-positive area or d neuro-
phosphorylated tau in brains of GW3965-treated 3xTg-AD mice exam- fibrillary tangles, was observed in the hippocampus of treated 3xTg-AD
ined by immunohistochemistry. Representative micrographs showing the mice. Data are expressed as mean ± SEM. Statistical analysis was per-
hippocampus. a Aβ immunohistochemistry was examined using anti- formed by one-way analysis of variance followed by Tukey’s multiple
body anti-Aβ (6E10). b Hyper-phosphorylated tau was examined using comparison test. Female mice, n = 3 per 3xTg-AD group
antibody anti-PHF1 (AT8). No significant differences were observed in

synapsin-1 protein in 3xTg-AD mice compared with untreated changes that occur and that are potentially related to the
3xTg-AD mice (P > 0.01, n = 4) (Fig. 4a, c) but did not change mechanism of action of this agonist.
PSD95 expression (Fig. 4a, b). First, we found that short-term GW3965 treatment
(50 mg/kg/day for 6 days) improved performance on learning
and retention tasks in treated 3xTg-AD compared with un-
treated 3xTg-AD mice. Similar behavioral results have been
Discussion previously reported using the same treatment in a contextual
fear-conditioning paradigm in the double transgenic AD
LXR activation is a promising therapeutic target for AD, as mouse model (Tg2576), which is a purely amyloidogenic
demonstrated by its ability to improve cognitive function in model (Jiang et al. 2008). Second, functional studies have
murine models of AD. Here, we investigated the potential shown that LXR agonist treatment contributes to Aβ clear-
mechanisms associated with the beneficial effects exerted by ance after 4 months of treatment (Jiang et al. 2008). However,
LXR agonist treatment in 24-month-old 3xTg-AD mice, a we did not find significant changes in the pathological hall-
murine model of AD. We used the 3xTg-AD animals at this marks of AD expressed in the 3xTg-AD mice model, most
particularly old age, as they are likely to suffer from a probably owing to the short-term treatment, and more time
detrimental aging process that mediates gene silencing as may be required to replicate the significant amyloid burden
a consequence of the human transgenes. We treated this reduction observed in the previous study (Jiang et al. 2008).
model short term to identify the first DNA methylation Moreover, transgene differences in the 3xTg-AD mice
J Mol Neurosci

Fig. 3 Quality control. The beta


density plot after probe selection
in our assay (a) is similar to that
performed on the human sample
included (b). The average
methylation ratio is similar with
or without treatment (c).
Therefore, changes in
methylation derived from the
treatment appear to be specific for
genomic loci (rather than a
general effect). The volcano plot
illustrates that the epigenetic
differences are mainly due to
decreased DNA methylation in a
specific set of probes (d, left side)

Table 1 Differentially
methylated synapses genes Gene Probe code P value Related function References
Name

Dlg3 23429746 0.00007 Regulation of NMDA signals pathway; (Han et al. 2011; Qu et al.
role in synaptic plasticity; mutations in 2009; Proctor et al.
this gene are associated with X-linked 2010)
mental retardation; is significantly de-
creased human AD patients
Syn1 00779763 0.00014 Localization at cytoplasmic surface of (Qin et al. 2004)
synaptic vesicles; role in synaptogenesis
and the modulation of neurotransmitter
release; is significantly decreased in
humans AD patients
Syp 10818284 0.00011 Integral membrane protein present in small (Proctor et al. 2010; Sze
synaptic vesicles; is significantly et al. 1997)
decreased in humans AD patients
Gpc4 07312966 0.00002 Localized in pre- and postsynaptic mem- (de Wit et al. 2013;
branes of excitatory synapses; belongs to Decktor et al. 1990;
GPC family associated with Aβ plaques van Horssen et al.
2001)
Tspan7 cg04345928 0.00008 Role in the control of neurite outgrowth; (Wakabayashi et al.
regulatory protein of γ-secretase com- 2009)
plex and activity
Cask 19638040 0.00020 Scaffold protein, is located at synapses in (Moog et al. 1993)
the brain
Il1rapl1 13077484 0.00022 Mediates excitatory synapse formation; (Ramos-Brossier et al.
mutation cause intellectual disabilities 2015; Yasumura et al.
2014)
J Mol Neurosci

Table 2 Differentially
methylated neurogenesis and Gene Probe P value Related function References
proliferation genes Name codes

Sox-3 21773962 0.00006 Involve in embryonic and primary (Rogers et al. 2013;
neurogenesis and mental retardation Laumonnier et al.
2002; Archer et al.
2011)
Hmgb3 12284142 0.00007 Regulates hematopoietic stem cell self- (Nemeth et al. 2006)
25685741 0.00009 renewal and differentiation
Zfx 06019215 0.00020 Control the self-renewal of human (Harel et al. 2012)
embryonic stem cells
pp90RSK2 17794813 0.00025 Codifies for serine/threonine kinases that (Anjum and Blenis
regulates mitogen-activated kinase 2008; Carriere et al.
(MAPK); controlling growth, 2008)
proliferation and migration
Zic3 06915321 0.00022 Pluripotency regulator of embryonic stem (Declercq et al. 2013;
13613682 0.00009 cells; induce conversion of human Kumar et al. 2012;
fibroblast to neuronal progenitor cells Lim et al. 2010)

compared with the Tg2576 mice, and also the strong pa- for cortical DNA methylation of CpG dinucleotides of cal-
thology of the very old 3xTg-AD mice used here, may cineurin, a memory-suppressing gene (Miller et al. 2010). In
have also contributed. the context of AD, we found changes in DNA methylation of
Third, the hippocampus is essential for cognitive function synapse-related genes associated with LXR activation and
and recent evidence indicates that learning and long-term specifically within DNA regions that codify for proteins lo-
memory depend on histone modifications and dynamic calized at synapses, either at the presynaptic terminal, e.g.,
changes in DNA methylation (for a review see Day and synapsin-1 and synaptophysin, or the postsynaptic terminal,
Sweatt 2011). These changes in DNA methylation can be e.g., SAP-102. These genes are reduced in the postmortem
transitory (for hours) or long-lasting (persist for weeks). The brain from AD patients (Proctor et al. 2010; Qin et al. 2004;
first evidence for transitory DNA methylation changes was Sze et al. 1997) and show decreased methylation following
detected on CpG dinucleotides of the Reelin gene after GW3965 treatment in the present analysis. Because changes
contextual fear conditioning (Miller and Sweatt 2007). In- in gene expression are frequently reported to be necessary
terestingly, Reelin and ApoE exert their effects by direct for synaptic plasticity, which is one of the main mecha-
interaction with ApoE receptors (ApoER2 and VLDLR) nisms associated with learning and memory (Flavell and
and LXR agonists upregulate ApoE protein expression. Greenberg 2008), it is interesting that LXR activation by
Similarly, persistent epigenetic changes have been described GW3965 treatment rapidly causes increased transcriptional

Fig. 4 GW3965 increases protein expression of synapsin-1 but not blot results in hippocampal lysates. Samples from each mouse were nor-
PSD95. Hippocampal levels of synapsin-1 and PSD95 were examined malized to actin and expressed as fold change. Data are expressed as
by Western blot analysis. a Representative Western blot images of mean ± SEM. Statistical analysis was performed by one-way analysis
synapsin-1 (Syn1), PSD95, and β-actin in the hippocampus of three of variance and Student’s t test comparison between groups, n = 4 per
different groups of animals: WT, GW3965-treated 3xTg-AD, and untreat- group. **P < 0.01 compared with WT; &&P < 0.01 compared with un-
ed 3xTg-AD mice. b Densitometric quantification of PSD95 Western blot treated 3XTg-AD
results in hippocampal lysates. c Densitometric analysis of Syn1 Western
J Mol Neurosci

activity of synapse-related proteins and appears to counter- improvement. However, the relationship mediated by LXR
act the epigenetic modifications mediated by the patholog- and methylation changes at the ABCA1 loci are not known.
ical condition. Fourth, we also found changes in DNA Unfortunately, the ApoE1 and Abca1 genes are not present in
methylation of genes that regulate proliferation and are the gene subset of the Illumina 450 k Infinium DNA Methyl-
most likely associated with regulating neuronal precursors ation microarray and therefore require a specific analysis ap-
in the subgranular zone of the hippocampus (Levison and proach (Wong et al. 2013).
Goldman 1993; Paterson et al. 1973), a primary niche for In conclusion, we have demonstrated changes in the DNA-
neuronal stem cells (NSC) and the site of adult neurogenesis. methylation pattern mediated by short-term treatment with an
Dysfunction in hippocampal neurogenesis is associated with LXR agonist (GW3965) in the 3xTg-AD mouse model.
AD in different transgenic animal models, namely those ex- Previous reports have shown that LXR agonists produce
pressing mutant presenilin-1 and mutant amyloid precursor cognitive improvements with short-term treatment (Jiang et al.
protein (APP) and in the 3xTg-AD (Rodriguez et al. 2008; 2008) and also in 21-month-old animals (Vanmierlo et al.
Donovan et al. 2006; Chevallier et al. 2005; Haughey et al. 2011) in a mechanism that is independent of Aβ plaques.
2002). It has been suggested that control of NSC proliferation Our results suggest an alternative mechanism of action to ex-
either by extrinsic factors (Wnt3, Wnt10b, Wnt2, BDNF, and plain the cognitive changes associated with short-term LXR
Reelin) or intrinsic factors (Sox2, Sox4, Sox11, Ngn2, agonist treatment in AD mice models, namely by mediating
NeuroD1, and NeuroD2) is dynamically regulated by epige- specific DNA hypo-methylation and re-expression of genes
netic modification of DNA or histones (Covic et al. 2010). In related to synaptic function in the hippocampus, one of the
agreement with this, LXR promotes neurogenesis in vitro in main affected regions in AD. We also observed changes in
human embryonic stem cells, and in vivo in the ventral mid- neurogenesis-related genes, and both pathways are widely
brain during development (Theofilopoulos et al. 2013; Ma described to have a role in the physiopathology and pro-
et al. 2009; Sacchetti et al. 2009), supporting our findings gression of AD.
of changes in neurogenesis-related genes. Further analysis
on the impact of LXR agonists on NSC gene regulation Acknowledgments We thank Dr. Jon Collins (GlaxoSmithKline,
described in the present model are needed. Stevenage, UK) for providing GW683965A. This study was funded by
Interestingly, we also found changes in DNA methylation Colciencias (Contract Nos. 401-2011 and 498-2012).
in additional genes such as FMR2P, PTCHD1, RPS6KA3, and
SLC6A8 (Table S1), which are associated with X-linked men- Compliance with Ethical Standards Animals were handled following
tal retardation and/or autism (Chaudhry et al. 2014; Bensaid the Colombian (Law 84 of 1989 and Resolution 8430 of 1993) and
et al. 2009; Rosenberg et al. 2004; Merienne et al. 1999). international regulations and standards on animal welfare and conformed
to the Animal Research: Reporting In Vivo Experiments (ARRIVE)
SLC6A8 is involved in creatine transport into cells and was guidelines (Kilkenny et al. 2010).
recently demonstrated to be required for energy control in the
brain and associated with impaired metabolism in different Conflict of Interest The authors declare that they have no competing
neuronal disorders including AD (Burklen et al. 2006; Gallant interests.
et al. 2006). However, the role of most of these genes on
synaptic function and development remains unclear, as well
as their role in AD physiopathology.
Previous research has shown cognitive improvement by
LXR agonist treatment with increased ABCA1 and ApoE
References
protein expression (Donkin et al. 2010; Jiang et al. 2008).
Anjum R, Blenis J (2008) The RSK family of kinases: emerging roles in
Both these genes are subjected to epigenetic regulation, and cellular signalling. Nat Rev Mol Cell Biol 9:747–758
ABCA1 epigenotypes are associated with familial hypercho- Archer TC, Jin J, Casey ES (2011) Interaction of Sox1, Sox2, Sox3 and
lesterolemia, aging, and coronary artery disease (Guay et al. Oct4 during primary neurogenesis. Dev Biol 350:429–440
2014; Guay et al. 2012). Regarding ApoE, a CpG dinucleotide Aryee MJ, Jaffe AE, Corrada-Bravo H, et al. (2014) Minfi: a flexible and
was found within the 3′-coding region of the gene, which was comprehensive Bioconductor package for the analysis of Infinium
DNA methylation microarrays. Bioinformatics 30:1363–1369
associated with expression levels at the APOE locus. In addi-
Banine F, Bartlett C, Gunawardena R, et al. (2005) SWI/SNF chromatin-
tion, the CpG dinucleotide is highly methylated in the human remodeling factors induce changes in DNA methylation to promote
postmortem brain (Yu et al. 2013). More recently, methylation transcriptional activation. Cancer Res 65:3542–3547
of an APOE CpG dinucleotide was shown and associated to Barrachina M, Ferrer I (2009) DNA methylation of Alzheimer disease
the pathological process of AD (Chibnik et al. 2015). LXR and tauopathy-related genes in postmortem brain. J Neuropathol
Exp Neurol 68:880–891
agonists are well known to increase ABCA1 gene expression Bensaid M, Melko M, Bechara EG, Davidovic L, Berretta A, Catania
in murine double transgenic models of AD, which promotes MV, Gecz J, Lalli E, Bardoni B (2009) FRAXE-associated mental
increasing proteolytic pathways of Aβ and mediates cognitive retardation protein (FMR2) is an RNA-binding protein with high
J Mol Neurosci

affinity for G-quartet RNA forming structure. Nucleic Acids Res 37: receptor agonist GW3965 on object recognition memory and amy-
1269–1279 loid burden in amyloid precursor protein/presenilin 1 mice. J Biol
Biernat J, Mandelkow EM, Schroter C, Lichtenberg-Kraag B, Steiner B, Chem 285:34,144–334154
Berling B, Meyer H, Mercken M, Vandermeeren A, Goedert M Donovan MH, Yazdani U, Norris RD, Games D, German DC, Eisch AJ
(1992) The switch of tau protein to an Alzheimer-like state includes (2006) Decreased adult hippocampal neurogenesis in the PDAPP
the phosphorylation of two serine-proline motifs upstream of the mouse model of Alzheimer’s disease. J Comp Neurol 495:70–83
microtubule binding region. EMBO J 11:1593–1597 Fitz NF, Cronican A, Pham T, Fogg A, Fauq AH, Chapman R, Lefterov I,
Braak H, Braak E, Bohl J (1993) Staging of Alzheimer-related cortical Koldamova R (2010) Liver X receptor agonist treatment ameliorates
destruction. Eur Neurol 33:403–408 amyloid pathology and memory deficits caused by high-fat diet in
Burklen TS, Schlattner U, Homayouni R, Gough K, Rak M, Szeghalmi APP23 mice. J Neurosci 30:6862–6872
A, Wallimann T (2006) The creatine kinase/creatine connection to Flavell SW, Greenberg ME (2008) Signaling mechanisms linking neuro-
Alzheimer’s disease: CK-inactivation, APP-CK complexes and fo- nal activity to gene expression and plasticity of the nervous system.
cal creatine deposits. J Biomed Biotechnol 2006:35936 Annu Rev Neurosci 31:563–590
Carriere A, Ray H, Blenis J, Roux PP (2008) The RSK factors of activat- Gallant M, Rak M, Szeghalmi A, Del Bigio MR, Westaway D, Yang J,
ing the Ras/MAPK signaling cascade. Front Biosci 13:4258–4275 Julian R, Gough KM (2006) Focally elevated creatine detected in
Chaudhry A, Noor A, Degagne B, Baker K, Bok LA, Brady AF, Chitayat amyloid precursor protein (APP) transgenic mice and Alzheimer
D, Chung BH, Cytrynbaum C, Dyment D, Filges I, Helm B, disease brain tissue. J Biol Chem 281:5–8
Hutchison HT, Jeng LJ, Laumonnier F, Marshall CR, Menzel M, Guay SP, Brisson D, Munger J, Lamarche B, Gaudet D, Bouchard L
Parkash S, Parker MJ, Raymond LF, Rideout AL, Roberts W, Rupps (2012) ABCA1 gene promoter DNA methylation is associated with
R, Schanze I, Schrander-Stumpel CT, Speevak MD, Stavropoulos HDL particle profile and coronary artery disease in familial hyper-
DJ, Stevens SJ, Thomas ER, Toutain A, Vergano S, Weksberg R, cholesterolemia. Epigenetics 7:464–472
Scherer SW, Vincent JB, Carter MT (2014) Phenotypic spectrum Guay SP, Legare C, Houde AA, Mathieu P, Bosse Y, Bouchard L (2014)
associated with PTCHD1 deletions and truncating mutations in- Acetylsalicylic acid, aging and coronary artery disease are associat-
cludes intellectual disability and autism spectrum disorder. Clin ed with ABCA1 DNA methylation in men. Clin Epigenetics 6:14
Genet Han N, Shi Z, Zhang K, Gao X, Zheng Z, Gong P, Guo Y, Huang S,
Chevallier NL, Soriano S, Kang DE, Masliah E, Hu G, Koo EH (2005) Zhang F (2011) Polymorphisms in the DLG3 gene is not associated
Perturbed neurogenesis in the adult hippocampus associated with with non-syndromic mental retardation in the Chinese Han popula-
presenilin-1 A246E mutation. Am J Pathol 167:151–159 tion of Qin-Ba mountain. Cell Mol Neurobiol 31:695–700
Chibnik LB, Yu L, Eaton ML, Srivastava G, Schneider JA, Kellis M, Harel S, Tu EY, Weisberg S, Esquilin M, Chambers SM, Liu B, Carson
Bennett DA, De Jager PL (2015) Alzheimer’s loci: epigenetic asso- CT, Studer L, Reizis B, Tomishima MJ (2012) ZFX controls the self-
ciations and interaction with genetic factors. Ann Clin Transl Neurol renewal of human embryonic stem cells. PLoS One 7, e42302
2:636–647 Haughey NJ, Liu D, Nath A, Borchard AC, Mattson MP (2002)
Collins JL, Fivush AM, Watson MA, Galardi CM, Lewis MC, Moore L, Disruption of neurogenesis in the subventricular zone of adult mice,
Parks DJ, Wilson JG, Tippin TK, Binz JG, Plunket KD, Morgan DG, and in human cortical neuronal precursor cells in culture, by amyloid
Beaudet EJ, Whitney KD, Kliewer SA, Willson TM (2002) beta-peptide: implications for the pathogenesis of Alzheimer’s dis-
Identification of a nonsteroidal liver X receptor agonist through par- ease. NeuroMolecular Med 1:125–135
allel array synthesis of tertiary amines. J Med Chem 45:1963–1966 Hernandez HG, Mahecha MF, Mejia A, Arboleda H, Forero DA (2014)
Coppieters N, Dieriks BV, Lill C, Faull RL, Curtis MA, Dragunow M Global long interspersed nuclear element 1 DNA methylation in a
(2014) Global changes in DNA methylation and Colombian sample of patients with late-onset Alzheimer’s disease.
hydroxymethylation in Alzheimer’s disease human brain. Am J Alzheimers Dis Other Demen 29:50–53
Neurobiol Aging 35:1334–1344 Jiang Q, Lee CY, Mandrekar S, Wilkinson B, Cramer P, Zelcer N, Mann
Covic M, Karaca E, Lie DC (2010) Epigenetic regulation of neurogenesis K, Lamb B, Willson TM, Collins JL, Richardson JC, Smith JD,
in the adult hippocampus. Heredity (Edinb) 105:122–134 Comery TA, Riddell D, Holtzman DM, Tontonoz P, Landreth GE
Day JJ, Sweatt JD (2011) Epigenetic mechanisms in cognition. Neuron (2008) ApoE promotes the proteolytic degradation of Abeta. Neuron
70:813–829 58:681–693
De Jager PL, Srivastava G, Lunnon K, Burgess J, Schalkwyk LC, Yu L, Jones PA (2012) Functions of DNA methylation: islands, start sites, gene
Eaton ML, Keenan BT, Ernst J, McCabe C, Tang A, Raj T, Replogle bodies and beyond. Nat Rev Genet 13:484–492
J, Brodeur W, Gabriel S, Chai HS, Younkin C, Younkin SG, Zou F, Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG (2010)
Szyf M, Epstein CB, Schneider JA, Bernstein BE, Meissner A, Improving bioscience research reporting: the ARRIVE guidelines
Ertekin-Taner N, Chibnik LB, Kellis M, Mill J, Bennett DA for reporting animal research. PLoS Biol 8:e1000412
(2014) Alzheimer’s disease: early alterations in brain DNA methyl- Kim MS, Kondo T, Takada I, Youn MY, Yamamoto Y, Takahashi S,
ation at ANK1, BIN1, RHBDF2 and other loci. Nat Neurosci 17: Matsumoto T, Fujiyama S, Shirode Y, Yamaoka I, Kitagawa H,
1156–1163 Takeyama K, Shibuya H, Ohtake F, Kato S (2012) Retraction:
de Wit J, O'Sullivan ML, Savas JN, Condomitti G, Caccese MC, DNA demethylation in hormone-induced transcriptional derepres-
Vennekens KM, Yates JR III, Ghosh A (2013) Unbiased discovery sion. Nature 486:280
of glypican as a receptor for LRRTM4 in regulating excitatory syn- Kumar A, Declercq J, Eggermont K, Agirre X, Prosper F, Verfaillie CM
apse development. Neuron 79:696–711 (2012) Zic3 induces conversion of human fibroblasts to stable neural
Decktor DL, Allen ML, Robinson M (1990) Esophageal motility, heart- progenitor-like cells. J Mol Cell Biol 4:252–255
burn, and gastroesophageal reflux: variations in clinical presentation Laumonnier F, Ronce N, Hamel BC, Thomas P, Lespinasse J, Raynaud
of esophageal dysphagia. Dysphagia 5:211–215 M, Paringaux C, Van BH, Kalscheuer V, Fryns JP, Chelly J, Moraine
Declercq J, Sheshadri P, Verfaillie CM, Kumar A (2013) Zic3 enhances C, Briault S (2002) Transcription factor SOX3 is involved in X-
the generation of mouse induced pluripotent stem cells. Stem Cells linked mental retardation with growth hormone deficiency. Am J
Dev 22:2017–2025 Hum Genet 71:1450–1455
Donkin JJ, Stukas S, Hirsch-Reinshagen V, Namjoshi D, Wilkinson A, Levison SW, Goldman JE (1993) Both oligodendrocytes and astrocytes
May S, Chan J, Fan J, Collins J, Wellington CL (2010) ATP-binding develop from progenitors in the subventricular zone of postnatal rat
cassette transporter A1 mediates the beneficial effects of the liver X forebrain. Neuron 10:201–212
J Mol Neurosci

Lim LS, Hong FH, Kunarso G, Stanton LW (2010) The pluripotency IL1RAPL1 mutations associated with intellectual disability impair
regulator Zic3 is a direct activator of the Nanog promoter in ESCs. synaptogenesis. Hum Mol Genet 24:1106–1118
Stem Cells 28:1961–1969 Rodriguez JJ, Jones VC, Tabuchi M, Allan SM, Knight EM, LaFerla FM,
Ly PT, Cai F, Song W (2011) Detection of neuritic plaques in Alzheimer’s Oddo S, Verkhratsky A (2008) Impaired adult neurogenesis in the
disease mouse model. J Vis Exp dentate gyrus of a triple transgenic mouse model of Alzheimer’s
Ma DK, Ming GL, Song H (2009) Oxysterols drive dopaminergic disease. PLoS One 3:e2935
neurogenesis from stem cells. Cell Stem Cell 5:343–344 Rogers N, Cheah PS, Szarek E, Banerjee K, Schwartz J, Thomas P (2013)
Masliah E, Dumaop W, Galasko D, Desplats P (2013) Distinctive patterns Expression of the murine transcription factor SOX3 during embry-
of DNA methylation associated with Parkinson disease: identifica- onic and adult neurogenesis. Gene Expr Patterns 13:240–248
tion of concordant epigenetic changes in brain and peripheral blood Rosenberg EH, Almeida LS, Kleefstra T, deGrauw RS, Yntema HG, Bahi
leukocytes. Epigenetics 8:1030–1038 N, Moraine C, Ropers HH, Fryns JP, Degrauw TJ, Jakobs C,
Merienne K, Jacquot S, Pannetier S, Zeniou M, Bankier A, Gecz J, Salomons GS (2004) High prevalence of SLC6A8 deficiency in
Mandel JL, Mulley J, Sassone-Corsi P, Hanauer A (1999) A mis- X-linked mental retardation. Am J Hum Genet 75:97–105
sense mutation in RPS6KA3 (RSK2) responsible for non-specific Rosenfeld MG, Lunyak VV, Glass CK (2006) Sensors and signals: a
mental retardation. Nat Genet 22:13–14 coactivator/corepressor/epigenetic code for integrating signal-de-
Miller CA, Gavin CF, White JA, Parrish RR, Honasoge A, Yancey CR, pendent programs of transcriptional response. Genes Dev 20:
Rivera IM, Rubio MD, Rumbaugh G, Sweatt JD (2010) Cortical 1405–1428
DNA methylation maintains remote memory. Nat Neurosci 13: Sacchetti P, Sousa KM, Hall AC, Liste I, Steffensen KR, Theofilopoulos
664–666 S, Parish CL, Hazenberg C, Richter LA, Hovatta O, Gustafsson JA,
Miller CA, Sweatt JD (2007) Covalent modification of DNA regulates Arenas E (2009) Liver X receptors and oxysterols promote ventral
memory formation. Neuron 53:857–869 midbrain neurogenesis in vivo and in human embryonic stem cells.
Ming GL, Song H (2011) Adult neurogenesis in the mammalian brain: Cell Stem Cell 5:409–419
significant answers and significant questions. Neuron 70:687–702 Sanchez-Mut JV, Aso E, Panayotis N, Lott I, Dierssen M, Rabano A,
Mitro N, Mak PA, Vargas L, Godio C, Hampton E, Molteni V, Kreusch A, Urdinguio RG, Fernandez AF, Astudillo A, Martin-Subero JI,
Saez E (2007) The nuclear receptor LXR is a glucose sensor. Nature Balint B, Fraga MF, Gomez A, Gurnot C, Roux JC, Avila J,
445:219–223 Hensch TK, Ferrer I, Esteller M (2013) DNA methylation map of
mouse and human brain identifies target genes in Alzheimer’s dis-
Moog U, Uyanik G, Kutsche K (2013) CASK-related disorders. In:
ease. Brain 136:3018–3027
Pagon RA, Adam MP, Ardinger HH, et al (eds) GeneReviews®
Sze CI, Troncoso JC, Kawas C, Mouton P, Price DL, Martin LJ (1997)
[Internet]. University of Washington, Seattle, 1993-2015. http://
Loss of the presynaptic vesicle protein synaptophysin in hippocam-
www.ncbi.nlm.nih.gov/books/NBK169825/
pus correlates with cognitive decline in Alzheimer disease. J
Morris RG, Garrud P, Rawlins JN, O’Keefe J (1982) Place navigation
Neuropathol Exp Neurol 56:933–944
impaired in rats with hippocampal lesions. Nature 297:681–683
Teschendorff AE, Marabita F, Lechner M, Bartlett T, Tegner J, Gomez-
O’Malley BW, Qin J, Lanz RB (2008) Cracking the coregulator codes. Cabrero D, Beck S (2013) A beta-mixture quantile normalization
Curr Opin Cell Biol 20:310–315 method for correcting probe design bias in Illumina Infinium 450 k
Nemeth MJ, Kirby MR, Bodine DM (2006) Hmgb3 regulates the balance DNA methylation data. Bioinformatics 29:189–196
between hematopoietic stem cell selfrenewal and differentiation. Theofilopoulos S, Wang Y, Kitambi SS, Sacchetti P, Sousa KM, Bodin K,
Proc Natl Acad Sci U S A 103:13783–13788 Kirk J, Salto C, Gustafsson M, Toledo EM, Karu K, Gustafsson JA,
Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Steffensen KR, Ernfors P, Sjovall J, Griffiths WJ, Arenas E (2013)
Kayed R, Metherate R, Mattson MP, Akbari Y, LaFerla FM Brain endogenous liver X receptor ligands selectively promote mid-
(2003) Triple-transgenic model of Alzheimer’s disease with brain neurogenesis. Nat Chem Biol 9:126–133
plaques and tangles: intracellular Abeta and synaptic dysfunc- van Horssen J, Otte-Holler I, David G, Maat-Schieman ML, van den
tion. Neuron 39:409–421 Heuvel LP, Wesseling P, de Waal RM, Verbeek MM (2001)
Otaegui-Arrazola A, Menendez-Carreno M, Ansorena D, Astiasaran I Heparan sulfate proteoglycan expression in cerebrovascular amyloid
(2010) Oxysterols: a world to explore. Food Chem Toxicol 48: beta deposits in Alzheimer's disease and hereditary cerebral hemor-
3289–3303 rhage with amyloidosis (Dutch) brains. Acta Neuropathol 102:604–
Paterson JA, Privat A, Ling EA, Leblond CP (1973) Investigation of glial 614
cells in semithin sections. 3. Transformation of subependymal cells Vanmierlo T, Rutten K, Dederen J, Bloks VW, van der Varkzee LC,
into glial cells, as shown by radioautography after 3 H-thymidine Kuipers F, Kiliaan A, Blokland A, Sijbrands EJ, Steinbusch H,
injection into the lateral ventricle of the brain of young rats. J Comp Prickaerts J, Lutjohann D, Mulder M (2011) Liver X receptor acti-
Neurol 149:83–102 vation restores memory in aged AD mice without reducing amyloid.
Proctor DT, Coulson EJ, Dodd PR (2010) Reduction in post-synaptic Neurobiol Aging 32:1262–1272
scaffolding PSD-95 and SAP-102 protein levels in the Alzheimer Wakabayashi T, Craessaerts K, Bammens L, Bentahir M, Borgions F,
inferior temporal cortex is correlated with disease pathology. J Herdewijn P, Staes A, Timmerman E, Vandekerckhove J,
Alzheimers Dis 21:795–811 Rubinstein E, Boucheix C, Gevaert K, De SB (2009) Analysis of
Qin S, Hu XY, Xu H, Zhou JN (2004) Regional alteration of synapsin I in the gamma-secretase interactome and validation of its association
the hippocampal formation of Alzheimer’s disease patients. Acta with tetraspanin-enriched microdomains. Nat Cell Biol 11:1340–
Neuropathol 107:209–215 1346
Qu M, Aronica E, Boer K, Fallmar D, Kumlien E, Nister M, Wester K, Wojcicka G, Jamroz-Wisniewska A, Horoszewicz K, Beltowski J (2007)
Ponten F, Smits A (2009) DLG3/SAP102 protein expression in Liver X receptors (LXRs). Part I: structure, function, regulation of
malformations of cortical development: a study of human epileptic activity, and role in lipid metabolism. Postepy Hig Med Dosw
cortex by tissue microarray. Epilepsy Res 84:33–41 (Online) 61:736–759
Ramos-Brossier M, Montani C, Lebrun N, Gritti L, Martin C, Wong NC, Ng J, Hall NE, Lunke S, Salmanidis M, Brumatti G, Ekert PG,
Seminatore-Nole C, Toussaint A, Moreno S, Poirier K, Dorseuil Craig JM, Saffery R (2013) Exploring the utility of human DNA
O, Chelly J, Hackett A, Gecz J, Bieth E, Faudet A, Heron D, methylation arrays for profiling mouse genomic DNA. Genomics
Frank KR, Loeys B, Humeau Y, Sala C, Billuart P (2015) Novel 102:38–46
J Mol Neurosci

Xu P, Li D, Tang X, Bao X, Huang J, Tang Y, Yang Y, Xu H, Fan X Yu CE, Cudaback E, Foraker J, Thomson Z, Leong L, Lutz F, Gill JA,
(2013) LXR agonists: new potential therapeutic drug for neurode- Saxton A, Kraemer B, Navas P, Keene CD, Montine T, Bekris LM
generative diseases. Mol Neurobiol 48:715–728 (2013) Epigenetic signature and enhancer activity of the human
Yasumura M, Yoshida T, Yamazaki M, Abe M, Natsume R, Kanno K, APOE gene. Hum Mol Genet 22:5036–5047
Uemura T, Takao K, Sakimura K, Kikusui T, Miyakawa T, Mishina Zelcer N, Khanlou N, Clare R, Jiang Q, Reed-Geaghan EG, Landreth GE,
M (2014) IL1RAPL1 knockout mice show spine density decrease, Vinters HV, Tontonoz P (2007) Attenuation of neuroinflammation
learning deficiency, hyperactivity and reduced anxiety-like behav- and Alzheimer’s disease pathology by liver x receptors. Proc Natl
iours. Sci Rep 4:6613 Acad Sci U S A 104:10601–10606

You might also like