ArticlePDF Available

Abstract and Figures

Down syndrome (DS) results from one extra copy of human chromosome 21 and leads to several alterations including intellectual disabilities and locomotor defects. The transchromosomic Tc1 mouse model carrying an extra freely-segregating copy of human chromosome 21 was developed to better characterize the relation between genotype and phenotype in DS. The Tc1 mouse exhibits several locomotor and cognitive deficits related to DS. In this report we analyzed the contribution of the genetic dosage of 13 conserved mouse genes located between Abcg1 and U2af1, in the telomeric part of Hsa21. We used the Ms2Yah model carrying a deletion of the corresponding interval in the mouse genome to rescue gene dosage in the Tc1/Ms2Yah compound mice to determine how the different behavioral phenotypes are affected. We detected subtle changes with the Tc1/Ms2Yah mice performing better than the Tc1 individuals in the reversal paradigm of the Morris water maze. We also found that Tc1/Ms2Yah compound mutants performed better in the rotarod than the Tc1 mice. This data support the impact of genes from the Abcg1-U2af1 region as modifiers of Tc1-dependent memory and locomotor phenotypes. Our results emphasize the complex interactions between triplicated genes inducing DS features.
Content may be subject to copyright.
RESEARCH ARTICLE
Dosage of the Abcg1-U2af1 Region Modifies
Locomotor and Cognitive Deficits Observed
in the Tc1 Mouse Model of Down Syndrome
Damien Marechal
1,2,3,4
, Patricia Lopes Pereira
5
, Arnaud Duchon
1,2,3,4
, Yann Herault
1,2,3,4,6
*
1Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 1 rue Laurent Fries, 67404 Illkirch,
France, 2Centre National de la Recherche Scientifique, UMR7104, Illkirch, France, 3Institut National de la
Santé et de la Recherche Médicale, U964, Illkirch, France, 4Université de Strasbourg, Illkirch, France,
5Transgenese et Archivage Animaux Modèles, TAAM, CNRS, UPS44, 3B rue de la Férollerie 45071
Orléans, France, 6Institut Clinique de la Souris, ICS, 1 rue Laurent Fries, 67404 Illkirch, France
*herault@igbmc.fr
Abstract
Down syndrome (DS) results from one extra copy of human chromosome 21 and leads to
several alterations including intellectual disabilities and locomotor defects. The
transchromosomic Tc1 mouse model carrying an extra freely-segregating copy of human
chromosome 21 was developed to better characterize the relation between genotype and
phenotype in DS. The Tc1 mouse exhibits several locomotor and cognitive deficits related
to DS. In this report we analyzed the contribution of the genetic dosage of 13 conserved
mouse genes located between Abcg1 and U2af1, in the telomeric part of Hsa21. We used
the Ms2Yah model carrying a deletion of the corresponding interval in the mouse genome
to rescue gene dosage in the Tc1/Ms2Yah compound mice to determine how the different
behavioral phenotypes are affected. We detected subtle changes with the Tc1/Ms2Yah
mice performing better than the Tc1 individuals in the reversal paradigm of the Morris water
maze. We also found that Tc1/Ms2Yah compound mutants performed better in the rotarod
than the Tc1 mice. This data support the impact of genes from the Abcg1-U2af1 region as
modifiers of Tc1-dependent memory and locomotor phenotypes. Our results emphasize the
complex interactions between triplicated genes inducing DS features.
Introduction
Down syndrome (DS; OMIN #190685) is a multigenic disorder, resulting from three copies of
human chromosome 21 (Hsa21) [1]. This condition is a paradigm of human aneuploid
disorders with a direct consequence of gene dosage [24] and a general perturbation of whole
transcriptome [5]. DS represents one-third of cases of intellectual disabilities and cognitive
impairment in school-aged children [68] and is associated with a wide range of dysmorpholo-
gies, such as characteristic faces, skeletal anomalies and brain alterations at the prefrontal cor-
tex, hippocampus and cerebellum levels [9,10]. Clinical features of DS also include
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 1/13
OPEN ACCESS
Citation: Marechal D, Pereira PL, Duchon A, Herault
Y (2015) Dosage of the Abcg1-U2af1 Region
Modifies Locomotor and Cognitive Deficits Observed
in the Tc1 Mouse Model of Down Syndrome. PLoS
ONE 10(2): e0115302. doi:10.1371/journal.
pone.0115302
Academic Editor: Barbara Bardoni, CNRS
UMR7275, FRANCE
Received: June 25, 2014
Accepted: November 24, 2014
Published: February 23, 2015
Copyright: © 2015 Marechal et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper.
Funding: The National Centre for Scientific
Research is a public research agency in France, The
European commission funded the AnEUploidy project
(LSHG-CT-2006-037627) and the Foundation Jerome
Lejeune granted the authors some funding.
Nevertheless the funders had no role in study design,
data collection and analysis, decision to publish, or
preparation of the manuscript.
developmental delay, metabolic defects, other symptoms and associated diseases but their
overall expressivity and penetrance are highly variable.
Mouse models have been developed in order to better understand the relationship between
phenotype and genotype in DS. The long arm of this chromosome (21q) was completely se-
quenced since 2001 [11], and recent transcription comparisonsstudies report that it contains
696 genes, including at least 235 protein-coding genes and 142 pseudogenes, with a large subset
of genes which have a mouse homolog located on regions of synteny carried by mouse chromo-
somes 16, 17 or 10 [12].
Several models carrying additional copies of regions homologous to Hsa21 were generated
and used to decipher the contribution of segments to DS phenotypes [3,10]. Locomotor and
learning deficits were found associated with trisomy of several segments located on Mmu16:
Ts65Dn [13], Ts1Cje [14]; on Mmu17 Ts(17)1Yey [15] or Ts1Yah [16] and on Mmu10 Ts
(10)1Yey [15] and in a single gene model for Dyrk1a [1720]. A different model was generated
in 2005: the Tc1 transchromosomic mouse line carrying an almost complete copy of Hsa21
with human genes expressed in various tissues [21]. Gribble and al. [22] deciphered the
sequence of the Hsa21 present in Tc1 cells, and they identified one deletion, six duplications
and 25 de novo structural rearrangements presumably due to the gamma irradiation used
during the process of creating the mouse line. Nevertheless, the Tc1 mouse line is the unique
humanized model for DS, and displays phenotypes affecting short term memory impairment,
the hippocampal function [21] and locomotor activities [23,24].
Further analysis started by combining different models to sort out the contribution of subre-
gions to specific DS phenotypes. The Ts65Dn mouse was crossed to the Ms1Rhr to demon-
strate that the Down syndrome critical region previously identified in humans was necessary
but not sufficient to induce DS cognitive phenotypes [2527]. The experiment was carried out
again for the App-Runx1 deletion crossed in Ts65Dn mice, which rescued post-natal lethality
and certain cardiac phenotypes [28]. Similarly, monosomy for the region Cstb-Prmt2 on chro-
mosome Mmu10, named Ms4Yah, was combined with the Tc1 transchromosome to show that
the 50 genes orthologous to the Hsa21 region are not involved in Tc1-induced phenotypes
[29].
We then further explored the contribution of the Abcg1-U2af1 region, located on mouse
chromosome 17, which contains 14 conserved genes, namely Abcg1,Tff3,Tff2,Tff1,Tmprss3,
Ubash3a,Rsph1,Slc37a1,Pde9a,Wdr4,Ndufv3,Pknox1,Cbs,U2af1, and two additional tran-
scribed units (loc 102631757 and AK019514). The trisomy of Abcg1-U2af1 displayed im-
pairment in working memory, in novel object recognition and overexpression of the conserved
genes, except Abcg1 which was inactivated during genetic engineering and U2af1, which is lo-
cated outside the interval [16]. All the genes from Abcg1-U2af1 genetic interval are trisomic in
the Tc1 mouse model except the Ndufv3 gene which is rearranged [22]. The corresponding
monosomy Ms2Yah [30] carries a deletion of the 12 conserved genes, plus the last exons of
Abcg1, and showed fear conditioning and social recognition defects [30]. To determine whether
the region could play a role in several DS phenotypes observed in the Tc1 mouse model,
Ms2Yah and Tc1 mouse models were examined for impairments in Open-Field, Morris water
maze and rotarod.
Materials and Methods
Ethics statement
All animals were treated in compliance with the animal welfare policies of the French Ministry
of Agriculture. Yann Herault was granted permission by the French Ministry of Agriculture
(law 87 848) under accreditation 67369. Behavioral experiments were planned in order to
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 2/13
Competing Interests: Yann Herault is a PLOS ONE
Editorial Board member. This does not alter the
authorsadherence to PLOS ONE Editorial policies
and criteria.
evaluate cognition and motor conditions in these mice as described previously in [29], submit-
ted to the local animal care, use and ethic committee of the IGBMC (ComEth), and approved
under accreditation number (2012069) to comply with the new regulation in France. Mice
were kept under specific pathogen free conditions with free access to food and water for all the
tests. The light cycle was controlled as 12 h light and 12 h dark (lights on at 7AM). The Morris
water maze (MWM) was conducted between 9:00AM and 1:00 PM. All the other tests were
done between 9:00AM and 4:00 PM.
After weaning, male mice were gathered from ten litters and kept as littermates in the same
cage with no isolated individual. The different apparatuses used were placed in a dimly lit test-
ing room (approximatively 20 lux). To produce experimental groups, only animals from litters
containing a minimum of two male pups were selected. Groups of animals were established for
all genotypes on the N2B6C3B genetic background (see below): wt (n = 12), Ms2Yah (n = 11),
Tc1 (n = 11), Tc1/Ms2Yah (n = 8). Animals were transferred to the experimental room 30 min
before each experimental test. The tests were administered in the following order: open field
(week 37), learning and reversal in MWM (weeks 5356), working memory in MWM (week
6263), and rotarod (week 72). No invasive procedure was used and the method of euthanasia
was CO
2
inhalation.
Mouse lines, breeding and genotyping
The Ms2Yah, official name Del(17Abcg1-Cbs)2Yah, mice were generated on 129/Ola ES cells
as described previously [16] and backcrossed on the C57BL/6J genetic background at least to
N10 in this study [30]. The Tc1 transchromosomic line has been described previously [21];
These mice were kept on an F1 B6C3B background; the C3B are sighted C3H/HeH, a congenic
line for the BALB/c allele at the Pde6b gene in C3H/HeH [31]. The two lines were crossed to
generate double mutant and control cohorts on a mixed genetic background B6xB6C3B
(N2B6C3B) under Specific Pathogen Free conditions.
For the identification of the Ms2Yah allele and the Tc1 chromosome, genomic DNA was iso-
lated from tail biopsies using the NaCl precipitation technique. The Ms2Yah allele was identified
using non quantitative PCR with 2 pairs of primers: one control mapping the end of the wild
type (wt) allele form of the U2af1 locus (wt Forward 5-CCAGCTGAAGATGGGTGTGTCTGC-
3 / wt Reverse 5-AGCCTTCCCTGGGGACCTGAAA-3) leading to the amplification of a PCR
product of 468 bp, and one transgenic pair mapping the junction between the U2af1 insert [16]
and the HPRT 3vector (Tg Forward 5-CCAGCTGAAGATGGGTGTGTCTGC-3 / Tg Reverse
5-AACGACCGAGCGCAGCGA-3) amplifying a product of 272 bp. The Hsa21 present in Tc1
mice was identified by PCR using primers D21S55F (5_-GGTTTGAGGGAACACAAAGCT-
TAACTCCCA-3_) and D21S55R (5_-ACAGAGCTACAGCCTCTGACACTATGAACT-3_)
that are specific for the Hsa21 and control primers specific for the mouse genome (MyoF:
50-TTACGTCCATCGTGGACAGCAT-30, MyoR: 50-TGGGCTGGGTGTTAGTCTTAT-30)
with specific PCR products of 208 bp and 245 bp, respectively.
Open field
This test measures rodent behavioral responses such as locomotor activity, hyperactivity and
exploratory behaviors within a closed space. The test was carried out in a 55 cm diameter
round white box and mouse activity was recorded with a video tracking system (Ethovision,
Noldus, France) during a single 30 min session. After each mouse trial, the arena was thor-
oughly cleaned with 50% ethanol, followed by one cleaning with water, and then dried with
paper towels to minimize olfactory cues. We quantified the speed and distance traveled during
3 phases (0 to 10 min; 10 to 20 min and 20 to 30 min). We also measured the percentage of
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 3/13
time spent in each arena zone (peripheral, intermediate, central) in the same phases of
the session.
Morris water maze spatial memory
Learning protocol. The water maze was a circular pool (150-cm diameter, 60-cm height) filled
to a depth of 40 cm with water maintained at 20°C22°C, made opaque using a white aqueous
emulsion (Acusol OP 301 opacifier) and split into 4 quadrants: South-East (SE), North-West
(NW), North-East (NE), South-West (SW). The escape platform, made of rough plastic, was
submerged 1 cm below the waters surface.
This experiment was performed to study reference memory through a spatial search strategy
that involved finding a hidden platform (6 cm diameter) in a pool. In the reversal mode mice
had to learn a new platform position. All the procedures were adapted from Morice et al., 2008
[24] and Duchon et al., 2011 [29]. The spatial memory session consisted of a 6-day (S1 to S6)
learning phase with two 90-second trials per day. Each trial started with mice facing the interior
wall of the pool and ended when they climbed onto the platform located on the SE quadrant or
after a maximum searching time of 90 sec. The starting position was changed pseudo-random-
ly between trials. Mice were left undisturbed in their home cage for 90-min inter-trial intervals.
On the seventh day, mice were given the 60-sec probe test in which the platform had been re-
moved. The distance traveled in each quadrant (NW, NE, SW, SE) was recorded to quantify
the time spent in the target quadrant.
Reversal Learning protocol. After the first probe trial, all mice were given a reversal test, in
which the hidden platform was moved to a new position (NW,). Mice were trained for 5 days
(Reversal Session RS1 to RS5) following the same training procedure and then tested for the
second probe trial on the sixth day (13 days after the beginning of the total test). To test mice
for long term memory, they were left undisturbed for 20 days before being given a third
probe trial.
Rotarod
The Rotarod test was performed to estimate rodent locomotor coordination. The apparatus
(Bioseb, France) is made of a rotating bar 5 cm in diameter (hard plastic materiel covered by
grey rubber foam) on which mice are placed facing the direction of rotation. The first phase
was a learning period composed of one training session with 4 trials per day for 3 days. For
each trial mice were placed on a rotating rod, starting from 0 and accelerating to 40 rpm in
5 minutes. We recorded the time spent on the rod and the speed before the fall. The second
step of the task occurred on the 4
th
day. It consisted of 7 trials of 2 minutes, each trial being per-
formed at one selected speed (4, 10, 16, 22, 28, 34 and 40 rpm). This procedure was repeated
twice and the time spent on the rod was recorded for each trial.
Statistical analyses
ANOVA was performed to analyze differences between the 4 genotype groups using dedicated
commercial Software (Sigmaplot): we applied a two way ANOVA for the Open-Field, Morris
Water Maze Probe trialsand rotarod test phase. we used a two Way repeated measures
ANOVA for Learning/Reversal protocols and the learning Rotarod phase. However, for all the
ANOVA, the post hoc analysis was done using Tukeys method. Data are presented as mean ±
s.e.m.
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 4/13
Results
The exploratory pattern in the open field is not altered in Tc1/Ms2Yah
mice
As described previously the Tc1 mouse displayed a hyperactive exploratory phenotypes in the
B6129 [23] but not in the B6C3B background [29]. In order to evaluate exploratory behavior,
locomotion deficits and increased anxiety related behavior, we used the open field and we mea-
sured horizontal activity during three consecutive 10-minutes intervals (Fig. 1). Habituation
was observed for all the genotypes (Two-way ANOVA time intervalsF(2,111) = 12.524 p <
0.001; Tukeys post hoc test 010 min vs 2030 minq = 7.001, p <0.001) and exploration in
the center versus the periphery was similar with no anxiety pattern in the Tc1 group or in Tc1/
Ms2Yah (data not shown). The total distances travelled are globally the same after thirty min-
utes. However, during the first ten minutes, the Tc1/Ms2Yah group are more active compared
to control and monosomic individuals (Fig. 1; parameter distance travelled, Two way
ANOVA time intervals 010 min, F(2,111) = 12.524 p <0.001; Tuckeys post hoc method
wt vs Tc1/Ms2Yahq = 4.663, p = 0.007; Ms2Yah vs Tc1/Ms2Yahq = 5.108, p = 0.003 and
Tc1 vs Tc1/Ms2Yahq = 3.968, p = 0.03). This slight change in the initial exploratory phase in
the Tc1/Ms2Yah double-mutants suggested that the Abcg1-U2af1 region might be involved in
controlling exploratory behavior. Nevertheless, this behavior was not found in the Tc1 geno-
type mice, unraveling a hyperactive phenotype in the Tc1 mutant mice on this
N2B6C3B background.
Memory impairment in the Morris watermaze of Tc1 mice depends on
the number of copies of the Abcg1-U2af1 region
To evaluate the impact of the Abcg1-U2af1 region on learning potential and memory, we per-
formed a Morris water maze test. During the first phase for place learning (Fig. 2A), all four ge-
notypes found the platform with similar efficiency and memorized where the platform was
located (parameter distance travelled, two way repeated measures ANOVA learning day,
F(1,38) = 64.517 p <0.001). The distance traveled was reduced between the first and the last
day of learning for all genotypes (Two-way repeated measures ANOVA, Tukeys post hoc test
S1 vs S6, wild type q = 5.643, p <0.001; Ms2Yah q = 6.973, p <0.001; Tc1 q = 7.037, p <
0.001, Tc1/Ms2Yah q = 3.513, p <0.001). In the probe test (Fig. 2A right panel, 24 hours after
the learning session, all mice traveled further in the target quadrant than in the rest of the
arena (parameter % distance travelledtwo way ANOVA quadrant, F(1,160) = 133.77 p <
0.001)) and no genotype effect was detected (Tukeys post hoc method target quadrant vs non
target quadrants, wild type q = 9.322, p <0.001; Ms2Yah q = 9.301, p <0.001; Tc1 q = 9.002,
p<0.001, Tc1/Ms2Yah q = 5.678, p <0.001). The spatial learning and memory of the Tc1 and
Tc1/Ms2Yah mice was not affected compared to wild type mice.
During the second phase, the reversal phase, all genotypes showed significant global learn-
ing between RS1 and RS5 (Fig. 2B; Two way repeated measures ANOVA trials,genotype,
F(1,38) = 33.018, p <0.001), but the performance of the Tc1 group are reduced compared to
wild type and Ms2Yah (Tukeys post hoc method, Tc1 RS1 vs RS5, ns). This phenotype was res-
cued in Tc1/Ms2Yah compound mice (Tukeys post hoc method: q = 3.322, p = 0.024). One
day after the last reversal session, a probe trial was performed (Fig. 2B right panel) and we
found that all mice remembered the correct position of the platform with greater distance
travelled in the target quadrant (parameter % distance travelledtwo way ANOVA
quadrant, F(1,160) = 88.878 p <0.001, Tukeys post hoc method target quadrant vs non
target quadrants, wild type q = 9.383, p <0.001; Ms2Yah q = 6.830, p <0.001; Tc1 q = 5.840,
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 5/13
p<0.001, Tc1/Ms2Yah q = 5.088, p <0.001). None of the groups displayed memory deficits
since they described the same performance during the 2 probe trials. However, Tc1 mice need-
ed more time to learn the new task.
Deficits in motor coordination of the Tc1 mice are partially rescued by the
loss of Abcg1-U2af1 trisomy
Mutant mice were screened for motor skill using the rotarod test, as described previously [29].
Two patterns were observed during the training phase: wild type and Ms2Yah mice displayed
the same capacities to stay on the rod, whereas both Tc1 and Tc1/Ms2Yah mice spent less time
on the rod than the wild type control (Fig. 3A; repeated ANOVA genotypeTukeys post hoc
analysis; F(3.64) = 25.932. p <0.001; wild type vs Tc1: q = 11.180. p <0.001; wild type vs Tc1/
Ms2Yah: q = 9.133. p <0.001) and Ms2Yah littermates (Ms2Yah vs Tc1. q = 7.066. p <0.001.
Ms2Yah vs Tc1/Ms2Yah. q = 5.297. p = 0.004) over the 3 days during the learning phase. More-
over, while the wild type improved their performance during the training period (ANOVA
days, Tukeys post hoc method: F(2.64) = 11.477, p <0.001; wild type day3 vs day1, q =
6.014, p <0.001), none of the other groups, Ms2Yah, Tc1 and Tc1/Ms2Yah, progressed at the
end of the training.
During the test phase (Fig. 3B), we determined the time spent on the rod at a given constant
speed, in two sessions per trial and with increasing speed between each trial (4, 10, 16, 22, 28,
34 and 40 rpm) for a maximum of 2 min. As with the training session, a significant difference
was observed in performance between the wild type and Ms2Yah versus the Tc1 and Tc1/
Ms2Yah genotypes while the speed increased from 4 rpm to 34 rpm (Two way ANOVA
speedand genotype; Tukeys post hoc method: F(18,224) = 3.308, p <0.001; wt vs Ms2Yah
q = 3.493, p = 0.06; wt vs Tc1 q = 15.000, p <0.001; wt vs Tc1/Ms2Yah q = 8.389, p <0.001;
Ms2Yah vs Tc1 q = 11.599, p <0.001; Ms2Yah vs Tc1/Ms2Yah q = 5.219, p <0.001). An inter-
mediate behavior was detected in Tc1/Ms2Yah mice with better exercise performance than Tc1
but lower than wild type mice (at 10 rpm, wt vs Tc1/Ms2Yah q = 8.923, p<0.001; Ms2Yah
Figure 1. Open field locomotor activity of mice. The distance travelled (m) during the exploration of the
open-field by mice with different genotypes are shown Tc1/Ms2Yah during the consecutive 010, 1020, 20
30 min intervals. The activity of Tc1/Ms2Yah was increased mainly during the first 10 minutes (parameter
distance travelled, Two-way ANOVA 010 min, F(2,111) = 12.524 p <0.001; Tuckeys post hoc method
wt vs Tc1/Ms2Yahq = 4.663, p = 0.007; Ms2Yah vs Tc1/Ms2Yahq = 5.108, p = 0.003 and Tc1 vs Tc1/
Ms2Yahq = 3.968, p = 0.03Tc1/Ms2YahTc1/Ms2Yah). Values are means + s.e.m.
doi:10.1371/journal.pone.0115302.g001
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 6/13
vs Tc1/Ms2Yah ns; Tc1 vs Tc1/Ms2Yah q = 4.766, p = 0.004 - at 16 rpm, wt vs Tc1/Ms2Yah
q = 10.514, p<0.001; Ms2Yah vs Tc1/Ms2Yah q = 5.502, p<0.001). In this experiment,
Ms2Yah mice showed normal coordination, but could not increase their performance on the
rod during the training days; the Abcg1-U2af1 region might play a role in locomotor abilities
because, although training appeared equivalent to that of wild type mice, a longer test session
revealed that double mutants Tc1/Ms2 could stay longer on the rod than Tc1 mice.
Discussion
The present study highlights the contribution of the Abcg1-U2af1 genetic interval to DS-related
features in Tc1 mouse models. We found that reducing the genetic dosage of this region in the
Figure 2. Learning and Reversal reference memory performance of mice in the Morris water maze. (A) The distance traveled on left panel (m) and the
percentage of distance traveled in the target quadrant (SE) during the probe trial (S7) on the right(with data from every individual shown), are shown during
learning in the Morris water maze. Mice from the four genotypes learned where the platform is located (SE) with a reduction of the distance traveled to find the
platform over the 6 learning sessions (S1 to S6). In the right panel, the horizontal line indicates the distance travelled using a random search strategy 25%.
No difference was observed for the different genotypes. (B) In the consecutive reversal session we observed similar learning capacities when the platform
position was located in the NW quadrant, and after five reversal learning days (RS1 to RS5) shown on the left. During the following probe trial (right panel,
RS6), individuals from all the genotypes spent more time in the new target quadrant (NW). Values represent mean + s.e.m.
doi:10.1371/journal.pone.0115302.g002
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 7/13
Tc1 mouse models rescued subtle impairments in reversal learning, working memory and did
so partially in the rotarod test, but had no impact on hyperactivity or spatial learning.
Robustness of Tc1 induced deficits and the influence of the genetic
background
In this third study focusing on the behavior phenotype of the Tc1 DS mouse model, we
performed the analysis in a new mixed genetic background, i.e. N2B6C3B. Originally, the Tc1
model was studied in the B6129S8 [23,24,32] and more recently transferred in the B6C3B
genetic background [28,29,33]. We generated the Ms2Yah, carrying the deletion of the
Figure 3. Locomotor performance through accelerating and continuous speed rotarod tasks. (A) We examined locomotor coordination through three
days of learning an acceleration protocol (from 4 to 40 rpm over 5 min). Results are expressed as the time (minutes) that mice remained on the rod before
falling (left panel), and the velocity at the time of falling (right panel). Although Ms2Yah did not express any deficit, mice could not improve their performance
over the 3 days (figure A, left panel: § p = 0.376). Conversely, Tc1 mice were unable to stay on the rod longer, or improve their time over the 3 days (figure A,
left panel: *** p<0.001 between wt and Tc1 and ### p<0.001 between wt and Tc1/Ms2Yah groups). (C) The graph displays the time (min) that mice stayed
on the rotarod during the test phase when the speed was set at 4, 10, 16, 22, 28, 34 and then 40 rpm, for a maximum of 2 minutes. The performance of the
Tc1 group was poor from the beginning of the task (Two-way ANOVA wt vs Tc1on speed 4rpm*p<0.05, 10/16 rpm *** p<0.001) and 22/28 rpm **
(p<0.01). Interestingly, the performance of Tc1/Ms2Yah mice was in-between that of Tc1 and controlsand the Ms2Yah did not show major differences
compared to wt. Values represent means + S.E.M.
doi:10.1371/journal.pone.0115302.g003
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 8/13
Abcg1-U2af1 region, on the C57BL/6J background. Thus when we crossed B6.Ms2Yah with the
Tc1 transchromosomic line kept in the B6C3B genetic background, the Tc1/Ms2Yah com-
pound mice and the relative control groups, wild-type, Ms2Yah and Tc1, were on the
N2B6C3B background. Tc1 mice are known to be mosaic, with some cells losing the Hsa21
chromosome [21]; nevertheless, we recapitulated most of the Tc1 phenotypes (Table 1) in the
MWM tests (for spatial learning), and in the rotarod tests (learning phase and challenge tests).
The hyperactive phenotypes observed in the 129S8 [23], was not found in the B6C3B back-
ground [29] but was replicated again in the N2B6C3B background (this study). We hypothesize
a contribution of the B6 genetic background which displays a higher level of locomotor activity
than 129 and C3H [3436]. Thus, even if most of the Tc1-induced phenotypes are robust, a
few are also affected by the genetic background. The set of disomic genes of the different genet-
ic backgrounds modified the Tc1-induced phenotypes. This study proposes that the variability
in the expressivity and penetrance of the features found in DS people depends on genetic inter-
actions between the trisomic genes and the whole genome.
The Abcg1-U2af1 region contributes to learning and memory deficits in
the Tc1 DS mouse models
Here we found that reducing the number of copies of the mouse genes located in the Abcg1-
U2af1 region, restores some deficits of the Tc1 model in the reversal phase of MWM. The Tc1/
Ms2Yah mice learned the location of the platform with similar efficiency to controls while the
Tc1 mice needed more sessions to do so. Although all the genotypes finally learned where the
platform was located in the probe test, the decrease in the performance of the Tc1 group could be
associated with a lack of cognitive flexibility, since learning memory was not altered. This particu-
lar phenotype is observed in Down syndrome people [37] and our results suggest that an increase
in one or more genes of the Abcg1-U2af1 region contributes to decreased behavioral flexibility.
Tc1 mice have severe deficits in motor skills in different motor coordination tasks such as
rotarod, static rod and footprint tests [23]. Rotarod performance analysis in our study confirmed
the deficit in the locomotor activity of Tc1 mice, which occurred in the training days and in the
test phase. After consecutive days of training, mice usually enhance their performance by staying
on the rod longer each day [38,39]. In our experiment, Tc1 and Tc1/Ms2Yah did not improve
their performance in the learning phase. The learning mechanisms of locomotor function were
altered, and decreasing the number of copies of the Abcg1-U2af1 region could not rescue them.
During the test phase with different increasing rotarod speeds, Tc1 mice displayed strong
impairment compared to controls. The Tc1/Ms2Yah showed better performance by staying
Table 1. Robustness of the Tc1 induced phenotypes observed in different genetic backgrounds and interference with different rescues.
Mouse Models Tc1
*
Tc1
§
Tc1/ Ms4Yah
§
Tc1
§
Tc1/ Ms2Yah
§
Ms2Yah
§
Background B6129 B6C3B B6C3B N2F1 N2F1 N2F1
Hyperactivity (open eld) + = = + + =
MWM spatial learning and memory = = = = = =
MWM reversal learning and memory = = = - = =
Rotarod learning - - - - - =
Rotarod test - - - - ±=
(+), (=), ()or(±) respectively indicate a reported effect with an improvement, similar or with an impairment compared to the control littermates. The (±)
corresponds to a partial rescue compared to the transchromosomic Tc1 model. Data adapted from [23,24]
*
,[29]
§
and this work.
doi:10.1371/journal.pone.0115302.t001
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 9/13
longer and at higher speed on the rod than the Tc1 group. We exclude a strong contribution of
the genetic background to this phenotype since the C3H fell earlier than the B6 in a similar
protocol and the learning phase was not compensated [6]. Thus, even if the Ms2Yah region is
not implicated directly in motor learning, at least the over-expression of one or more genes lo-
cated in the interval definitely modifies locomotor activity.
Searching for candidates in the Abcg1-U2af1 genetic interval
Identifying genes in the Abcg1-U2af1 region modulating Tc1-induced locomotor phenotypes
would certainly improve our understanding of DS and stimulate further therapeutic ap-
proaches. As a consequence of the results described here, candidates should be conserved be-
tween humans and mice and expressed during the development of the adult brain. Many
regions of the brain are involved in motor learning and performance, such as the cerebellum,
basal ganglia, and the motor cortex. Thus expression of genes in the brain might help to dis-
criminate candidates for the different phenotypes described here. According to the Allen brain
atlas, Abcg1,Tff1,Ubash3a,Pde9a,Ndufv3,Pknox1 and Cbs were found expressed in the adult
cerebellum and in the isocortex and are thus candidates. Of particular interest are Pde9a and
Cbs, which are both expressed in the central nervous system. Pde9a codes the phosphodiester-
ase 9a and transforms cAMP and cGMP into their respective monophosphate forms and its in-
hibition can stimulate neuronal plasticity [40,41]. A delay in neuronal transmission may
partially explain the change in learning tasks. Otherwise, the Cbs gene encodes cystathionine-
beta-synthase whose deficiency causes homocystinuria (OMIN236200), a metabolic disorder
with intellectual disabilities. Cbs is a strong candidate for phenotypes described by both mono-
somic and trisomic models in the hippocampus [16,30]. In addition, a mouse model overex-
pressing human CBS displayed an increase magnitude of the long term potentiation in vitro
similar to the Ts1Yah electrophysiological phenotypes observed in vivo [42].
Only a few cases of DS have been reported with partial trisomy overlapping the most telo-
meric part of human chromosome 21 [43,44] and so far none have been described with a triso-
my limited to this segment. Conversely several cases with monosomy of the telomeric end have
been described with mild phenotypes [4548]. Similarly, analysis of trisomy and monosomy
models for Cstb-Prmt2 showed no phenotypes in open field, in the Morris water Maze, either
alone or in combination with the Tc1 transchromosome [15,29,49]. Only in fear conditioning
task, the Df(10)Yey/- displayed a deficit in contextual memory [49].
In this report, we confirmed that rescuing the number of copies of Abcg1-U2af1 modulates
Tc1-induced phenotypes only slightly, although the region was sufficient alone to induce cer-
tain learning and memory deficits [15,16], and as such this genetic interval certainly contrib-
utes, along with other regions of Hsa21, to the variability of DS features.
Acknowledgments
We thank the members of the laboratory and the AnEUploidy consortium for their helpful
comments (www.aneuploidy.org). We are grateful to the employees of ICS and TAAM for tak-
ing care of the animals. The mutant mice are available for distribution through the European
Mouse Mutant Archive(www.emmanet.org).
Author Contributions
Conceived and designed the experiments: DM AD YH. Performed the experiments: DM PLP.
Analyzed the data: DM YH. Contributed reagents/materials/analysis tools: PLP AD. Wrote the
paper: DM AD YH.
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 10 / 13
References
1. Antonarakis SE, Lyle R, Dermitzakis ET, Reymond A, Deutsch S (2004) Chromosome 21 and Down
syndrome: From genomics to pathophysiology. Nature Reviews Genetics 5: 725738. PMID:
15510164
2. Herault Y, Duchon A, Velot E, Maréchal D, Brault V (2012) The in vivo Down syndrome genomic library
in mouse. Prog Brain Res 197: 169197. doi: 10.1016/B978-0-444-54299-1.00009-1 PMID: 22541293
3. Dierssen M, Herault Y, Estivill X (2009) Aneuploidy: from a physiological mechanism of variance to
Down syndrome. Physiol Rev 89: 887920. doi: 10.1152/physrev.00032.2007 PMID: 19584316
4. Korenberg JR, Chen XN, Schipper R, Sun Z, Gonsky R, et al. (1994) Down syndrome phenotypes: the
consequences of chromosomal imbalance. Proc Natl Acad Sci U S A 91: 49975001. PMID: 8197171
5. Letourneau A, Santoni FA, Bonilla X, Sailani MR, Gonzalez D, et al. (2014) Domains of genome-wide
gene expression dysregulation in Downs syndrome. Nature 508: 345350. doi: 10.1038/nature13200
PMID: 24740065
6. Nadel L (2003) Downs syndrome: a genetic disorder in biobehavioral perspective. Genes Brain and
Behavior 2: 156166. PMID: 12931789
7. Gibson D, Groeneweg G, Jerry P, Harris A (1988) AGE AND PATTERN OF INTELLECTUAL DECLINE
AMONG DOWN SYNDROME AND OTHER MENTALLY-RETARDED ADULTS. International Journal
of Rehabilitation Research 11: 4755. PMID: 2974839
8. Antonarakis SE, Epstein CJ (2006) The challenge of Down syndrome. Trends in Molecular Medicine
12: 473479. PMID: 16935027
9. Dierssen M, Herault Y, Estivill X (2009) Aneuploidy: from a physiological mechanism of variance to
down syndrome. Physiol Rev 89: 887920. doi: 10.1152/physrev.00032.2007 PMID: 19584316
10. Herault Y, Duchon A, Velot E, Marechal D, Brault V (2012) The in vivo Down syndrome genomic library
in mouse. Down Syndrome: from Understanding the Neurobiology to Therapy 197: 169197. doi: 10.
1523/JNEUROSCI.3728-10.2010 PMID: 21068296
11. Hattori M, Fujiyama A, Taylor TD, Watanabe H, Yada T, et al. (2000) The DNA sequence of human
chromosome 21. Nature 405: 311319. PMID: 10830953
12. Sturgeon X, Gardiner KJ (2011) Transcript catalogs of human chromosome 21 and orthologous chim-
panzee and mouse regions. Mammalian Genome 22: 261271. doi: 10.1007/s00335-011-9321-y
PMID: 21400203
13. Reeves RH, Irving NG, Moran TH, Wohn A, Kitt C, et al. (1995) A MOUSE MODEL FOR DOWN-SYN-
DROME EXHIBITS LEARNING AND BEHAVIOR DEFICITS. Nature Genetics 11: 177184. PMID:
7550346
14. Sago H, Carlson EJ, Smith DJ, Kilbridge J, Rubin EM, et al. (1998) Ts1Cje, a partial trisomy 16 mouse
model for Down syndrome, exhibits learning and behavioral abnormalities. Proceedings of the National
Academy of Sciences of the United States of America 95: 62566261. PMID: 9600952
15. Yu T, Liu CH, Belichenko P, Clapcote SJ, Li SM, et al. (2010) Effects of individual segmental trisomies
of human chromosome 21 syntenic regions on hippocampal long-term potentiation and cognitive be-
haviors in mice. Brain Research 1366: 162171. doi: 10.1016/j.brainres.2010.09.107 PMID: 20932954
16. Lopes Pereira P, Magnol L, Sahún I, Brault V, Duchon A, et al. (2009) A new mouse model for the triso-
my of the Abcg1-U2af1 region reveals the complexity of the combinatorial genetic code of down syn-
drome. Hum Mol Genet 18: 47564769. doi: 10.1093/hmg/ddp438 PMID: 19783846
17. Fotaki V, de Lagran MM, Estivill X, Arbones M, Diersson M (2004) Haploinsufficiency of Dyrk1A in mice
leads to specific alterations in the development and regulation of motor activity. Behavioral Neurosci-
ence 118: 815821. PMID: 15301607
18. de Lagran MM, Altafaj X, Gallego X, Marti E, Estivill X, et al. (2004) Motor phenotypic alterations in
TgDyrk1a transgenic mice implicate DYRK1A in Down syndrome motor dysfunction. Neurobiology of
Disease 15: 132142. PMID: 14751778
19. Dierssen M, Altafaj X, Guimera J, Arbones M, Estivill X, et al. (1999) Transgenic mice overexpressing
the rat minibrain gene (Dyrk1a): implications for Down syndrome. Cytogenetics and Cell Genetics 86:
1112.
20. Altafaj X, Dierssen M, Baamonde C, Marti E, Visa J, et al. (2001) Neurodevelopmental delay, motor ab-
normalities and cognitive deficits in transgenic mice overexpressing Dyrk1A (minibrain), a murine
model of Downs syndrome. Human Molecular Genetics 10: 19151923. PMID: 11555628
21. ODoherty A, Ruf S, Mulligan C, Hildreth V, Errington ML, et al. (2005) An aneuploid mouse strain carry-
ing human chromosome 21 with Down syndrome phenotypes. Science 309: 20332037. PMID:
16179473
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 11 / 13
22. Gribble SM, Wiseman FK, Clayton S, Prigmore E, Langley E, et al. (2013) Massively Parallel Sequenc-
ing Reveals the Complex Structure of an Irradiated Human Chromosome on a Mouse Background in
the Tc1 Model of Down Syndrome. Plos One 8. doi: 10.1371/journal.pone.0082806 PMID: 24482673
23. Galante M, Jani H, Vanes L, Daniel H, Fisher EMC, et al. (2009) Impairments in motor coordination
without major changes in cerebellar plasticity in the Tc1 mouse model of Down syndrome. Human Mo-
lecular Genetics 18: 14491463. doi: 10.1093/hmg/ddp055 PMID: 19181682
24. Morice E, Andreae LC, Cooke SF, Vanes L, Fisher EMC, et al. (2008) Preservation of long-term memo-
ry and synaptic plasticity despite short-term impairments in the Tc1 mouse model of Down syndrome.
Learn Mem 15: 492500. doi: 10.1101/lm.969608 PMID: 18626093
25. Olson LE, Richtsmeier JT, Leszl J, Reeves RH (2004) A chromosome 21 critical region does not cause
specific down syndrome phenotypes. Science 306: 687690. PMID: 15499018
26. Olson LE, Roper RJ, Sengstaken CL, Peterson EA, Aquino V, et al. (2007) Trisomy for the Down syn-
dromecritical regionis necessary but not sufficient for brain phenotypes of trisomic mice. Human Mo-
lecular Genetics 16: 774782. PMID: 17339268
27. Belichenko NP, Belichenko PV, Kleschevnikov AM, Salehi A, Reeves RH, et al. (2009) The Down Syn-
drome Critical RegionIs Sufficient in the Mouse Model to Confer Behavioral, Neurophysiological, and
Synaptic Phenotypes Characteristic of Down Syndrome. Journal of Neuroscience 29: 59385948. doi:
10.1523/JNEUROSCI.1547-09.2009 PMID: 19420260
28. Raveau M, Lignon JM, Nalesso V, Duchon A, Groner Y, et al. (2012) The App-Runx1 Region Is Critical
for Birth Defects and Electrocardiographic Dysfunctions Observed in a Down Syndrome Mouse Model.
Plos Genetics 8. doi: 10.1371/journal.pgen.1003200 PMID: 23300473
29. Duchon A, Pothion S, Brault V, Sharp AJ, Tybulewicz VLJ, et al. (2011) The telomeric part of the human
chromosome 21 from Cstb to Prmt2 is not necessary for the locomotor and short-term memory deficits
observed in the Tc1 mouse model of Down syndrome. Behavioural Brain Research 217: 271281. doi:
10.1016/j.bbr.2010.10.023 PMID: 21047530
30. Sahún I, Marechal D, Lopes Pereira P, Nalesso V, Gruart A, et al. (2014) Cognition and Hippocampal
Plasticity in the Mouse Is Altered by Monosomy of a Genomic Region Implicatedin Down Syndrome.
Genetics.
31. Hoelter SM, Dalke C, Kallnik M, Becker L, Horsch M, et al. (2008) Sighted C3Hmicea tool for analys-
ing the influence of vision on mouse behaviour? Frontiers in Bioscience 13: 58105823. PMID:
18508624
32. Dunlevy L, Bennett M, Slender A, Lana-Elola E, Tybulewicz VL, et al. (2010) Downs syndrome-like car-
diac developmental defects in embryos of the transchromosomic Tc1 mouse. Cardiovasc Res 88:
287295. doi: 10.1093/cvr/cvq193 PMID: 20558441
33. Duchon A, Raveau M, Chevalier C, Nalesso V, Sharp AJ, et al. (2011) Identification of the translocation
breakpoints in the Ts65Dn and Ts1Cje mouse lines: relevance for modeling down syndrome. Mamm
Genome.
34. Gubner NR, Wilhelm CJ, Phillips TJ, Mitchell SH (2010) Strain Differences in Behavioral Inhibition in a
Go/No-go Task Demonstrated Using 15 Inbred Mouse Strains. Alcoholism-Clinical and Experimental
Research 34: 13531362. doi: 10.1111/j.1530-0277.2010.01219.x PMID: 20491731
35. Bolivar VJ, Caldarone BJ, Reilly AA, Flaherty L (2000) Habituation of activity in an open field: A survey
of inbred strains and F-1 hybrids. Behavior Genetics 30: 285293. PMID: 11206083
36. Moy SS, Nadler JJ, Young NB, Perez A, Holloway LP, et al. (2007) Mouse behavioral tasks relevant to
autism: phenotypes of 10 inbred strains. Behav Brain Res 176: 420. PMID: 16971002
37. Campbell C, Landry O, Russo N, Flores H, Jacques S, et al. (2013) Cognitive flexibilityamong individu-
als with Down syndrome: assessing the influence of verbal and nonverbal abilities. Am J Intellect Dev
Disabil 118: 193200. doi: 10.1352/1944-7558-118.3.193 PMID: 23734614
38. Jones BJ, Roberts DJ (1968) QUANTITATIVE MEASUREMENT OF MOTOR INCO-ORDINATION IN
NAIVE MICE USING AN ACCELERATING ROTAROD. Journal of Pharmacy and Pharmacology 20:
302&. PMID: 4384609
39. Rustay NR, Wahlsten D, Crabbe JC (2003) Influence of task parameters on rotarod performance and
sensitivity to ethanol in mice. Behavioural Brain Research 141: 237249. PMID: 12742261
40. Kroker KS, Rast G, Giovannini R, Marti A, Dorner-Ciossek C, et al. (2012) Inhibition of acetylcholines-
terase and phosphodiesterase-9A has differential effects on hippocampal early and late LTP. Neuro-
pharmacology 62: 19641974. doi: 10.1016/j.neuropharm.2011.12.021 PMID: 22245562
41. Hutson PH, Finger EN, Magliaro BC, Smith SM, Converso A, et al. (2011) The selective phosphodies-
terase 9 (PDE9) inhibitor PF-04447943 (6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-
(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one) enhances synaptic
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 12 / 13
plasticity and cognitive function in rodents. Neuropharmacology 61: 665676. doi: 10.1016/j.
neuropharm.2011.05.009 PMID: 21619887
42. Regnier V, Billard JM, Gupta S, Potier B, Woerner S, et al. (2012) Brain Phenotype of Transgenic Mice
Overexpressing Cystathionine beta-Synthase. Plos One 7. doi: 10.1371/journal.pone.0051204 PMID:
23349657
43. Lyle R, Béna F, Gagos S, Gehrig C, Lopez G, et al. (2009) Genotype-phenotype correlations in Down
syndrome identified by array CGH in 30 cases of partial trisomy and partial monosomy chromosome
21. Eur J Hum Genet 17: 454466. doi: 10.1038/ejhg.2008.214 PMID: 19002211
44. Korbel J, Tirosh-Wagner T, Urban A, Chen X, Kasowski M, et al. (2009) The genetic architecture of
Down syndrome phenotypes revealed by high-resolution analysis of human segmental trisomies. Proc
Natl Acad Sci U S A 106: 1203112036. doi: 10.1073/pnas.0813248106 PMID: 19597142
45. Hannachi H, Mougou-Zerelli S, BenAbdallah I, Mama N, Hamdi I, et al. (2011) Clinical and molecular
characterization of a combined 17p13.3 microdeletion with partial monosomy 21q21.3 in a 26-year-old
man. Cytogenet Genome Res 135: 102110. doi: 10.1159/000330880 PMID: 21876345
46. Melis D, Genesio R, Cappuccio G, MariaGinocchio V, Casa RD, et al. (2011) Mental retardation, con-
genital heart malformation, and myelodysplasia in a patient with a complex chromosomal rearrange-
ment involving the critical region 21q22. Am J Med Genet A 155A: 16971705. doi: 10.1002/ajmg.a.
33976 PMID: 21671372
47. Roberson ED, Wohler ES, Hoover-Fong JE, Lisi E, Stevens EL, et al. (2011) Genomic analysis of par-
tial 21q monosomies with variable phenotypes. Eur J Hum Genet 19: 235238. doi: 10.1038/ejhg.
2010.150 PMID: 20823914
48. Lyle R, Bena F, Gagos S, Gehrig C, Lopez G, et al. (2009) Genotype-phenotype correlations in Down
syndrome identified by array CGH in 30 cases of partial trisomy and partial monosomy chromosome
21. European Journal of Human Genetics 17: 454466. doi: 10.1038/ejhg.2008.214 PMID: 19002211
49. Yu T, Clapcote S, Li Z, Liu C, Pao A, et al. (2010) Deficiencies in the region syntenic to human 21q22.3
cause cognitive deficits in mice. Mamm Genome 21: 258267. doi: 10.1007/s00335-010-9262-x
PMID: 20512340
ABCG1-U2AF1 Region and Behavior in Down Syndrome
PLOS ONE | DOI:10.1371/journal.pone.0115302 February 23, 2015 13 / 13
... We reviewed the clinical manifestations of the patients with truncating variants, except p.(Ser660fs)(Table S1, Supplementary text). Recurrent features include, consistently with what was reported 5,6,8,11 : moderate to severe ID, prenatal or postnatal progressive microcephaly, major speech impairment, feeding difficulties, seizures and especially history of febrile seizures, autistic traits and anxiety, delayed gross motor development with unstable gait, brain MRI abnormalities including dilated ventricles and corpus callosum hypoplasia and recurrent facial features ( Figure 1A, Figure S3). We found genital abnormalies as reported 35 but no obvious renal anomalies. ...
... Although numerous studies have unraveled some of the functions of DYRK1A, most of the works about its role in brain have been performed in mouse (Fotaki et al. 2002;Fotaki et al. 2004), drosophila (11,11,13) or more recently, in xenopus models Willsey et al. 2020). Therefore, little is known about its role in human neural precursors, and about what are, among its cellular functions, those that are critical for human brain development and functioning. ...
... Although numerous studies have unraveled some of the functions of DYRK1A, most of the works about its role in brain have been performed in mouse (Fotaki et al. 2002;Fotaki et al. 2004), drosophila (11,11,13) or more recently, in xenopus models Willsey et al. 2020). Therefore, little is known about its role in human neural precursors, and about what are, among its cellular functions, those that are critical for human brain development and functioning. ...
Thesis
La déficience intellectuelle (DI) est un trouble du neurodéveloppement et la première raison de consultation génétique. Cependant, un certain nombre de variants restent classés en tant que Variants de signification inconnue (VSI) et contribuent au phénomène d’errance diagnostique. De plus, la DI est caractérisée par une hétérogénéité génétique extrême et les mécanismes physiopathologiques associés sont généralement mal connues. Mon travail de doctorat a consisté à mettre au point des approches expérimentales permettant de reclasser l’ensemble des VSI dans les gènes DYRK1A et PQBP1. J’ai également investigué les mécanismes cellulaires dérégulés lors de l’inactivation de ces gènes dans un modèle de précurseurs neuronaux. Mes travaux de doctorat permettront donc d’améliorer le diagnostic moléculaire de la déficience intellectuelle associée à des mutations dans les gènes DYRK1A et PQBP1. Ils permettront également de mieux comprendre les mécanismes physiopathologiques impliqués dans ces troubles neurodéveloppementaux. Ces connaissances aideront à mieux prendre en charge les individus atteints et pourront permettre d’identifier de nouvelles cibles thérapeutiques.
... Since this model's inception, further analyses have combined different models to distinguish the subregions' contributions to specific DS phenotypes. [31] Its copy of Hsa21, which is nearly complete, resides in Tc1 cells and only differs in six duplications, one deletion, and 25 structural rearrangements de novo that were potentially from the model's exposure to gamma radiation during its creation. [32] The Tc1 mouse line displays phenotypes that affect the hippocampal function, [33] locomotor activities, and short term memory impairment, all of which are characteristics in a damaged Hsa21. ...
... [38] In their main study, the Ms2Yah was paired with the Tc1 model to observe how different behavioral phenotypes are affected. [31] The results indicated that the Tc1/Ms2Yah mice and the controls learned the platform location equally quickly, but the Tc1 learned it more slowly. Learning memory was unaffected, so Marechal et al. suspect that the Tc1 mice's worse performance was due to an absence of cognitive flexibility. ...
... This demonstrated the alteration of locomotor function's learning mechanisms, which could not be resolved by the Abcg1-U2af1's decreasing number of copies. [31] ...
Article
Full-text available
Telomeres are a characteristic of chromosomes that have increasingly large significance in research. They are studied in various diseases to discover potential treatment strategies. Their most vital characteristic is their length because the length can be used to describe different characteristics about the cell, such as its age. The length of telomeres can also be used as a potential way to treat disease. This review article’s purpose is to explore how te-lomeres can be potentially used as a method to treat genetic diseases such as trisomy 21 and cancer.
... This hypothesis is in agreement with the study of rare individuals, carrying Hsa21 duplications, who displayed intellectual disabilities (5,7) and should be taken into account when therapeutic assays are planned. We suggest that preclinical observation in one partial trisomic mouse model should be replicated in more genetically complex models to test potential genetic influences (39,60,96). This is probably the limit of the model, since although behaviour and memory mechanisms are common between mice and humans, the complexity of the model is lower. ...
... The Dp5Yah crossed with Tg(Dyrk1a) was tested for Y-maze and NOR at 24h. All the standard operating procedures for behavioural phenotyping have been already described (96,(100)(101)(102)(103) and are detailed in the supplementary information. ...
Article
Full-text available
Down syndrome (DS) is the most common genetic form of intellectual disability caused by the presence of an additional copy of human chromosome 21 (Hsa21). To provide novel insights into genotype–phenotype correlations, we used standardized behavioural tests, magnetic resonance imaging (MRI) and hippocampal gene expression to screen several DS mouse models for the mouse chromosome 16 region homologous to Hsa21. First, we unravelled several genetic interactions between different regions of chromosome 21 and how they contribute significantly to altering the outcome of the phenotypes in brain cognition, function and structure. Then, in-depth analysis of misregulated expressed genes involved in synaptic dysfunction highlighted 6 biological cascades centred around DYRK1A, GSK3β, NPY, SNARE, RHOA and NPAS4. Finally, we provide a novel vision of the existing altered gene–gene crosstalk and molecular mechanisms targeting specific hubs in DS models that should become central to better understanding of DS and improving the development of therapies.
... The 655 same was observed with other genes as Irgn1,Ifit1, Ifit2 or Ndufa13. This upregulation of the 656 interferonB pathways was previously reported in the Ts1Cje mouse model (79,80) The result in one partial trisomic mouse model should be replicated in more genetically complex 696 models to test potential genetic influencial factor (35,84,85). This is probably the limit of the 697 model, even if the mechanisms of behaviour and memory are common between mice and 698 human, the complexity of the system is lower. ...
... The only 709 exception is the trisomic Ts65Dn (Ts(17 16 Tg(Dyrk1a) was tested for Y-maze and NOR at 24h. All the standard operating procedures for 737 behavioural phenotyping have been already described (85,(88)(89)(90)(91) Then the brain structure was dissected and kept in PFA 4% 2mM ProHance over night at 4°C. 746 ...
Preprint
Full-text available
Down syndrome (DS) is the most common genetic form of intellectual disability caused by the presence of an additional copy of human chromosome 21. To provide novel insights into genotype–phenotype correlations, we screened the in vivo DS mouse library with standardized behavioural tests, magnetic resonance imaging (MRI) and hippocampal gene expression. Altogether this approach brings novel insights into the field. First, we unravelled several genetic interactions between different regions of the chromosome 21 and how they importantly contribute in altering the outcome of the phenotypes in brain function and structure. Then, in depth analysis of misregulated expressed genes involved in synaptic dysfunction highlitghed 6 biological cascades centered around DYRK1A, GSK3β, NPY, SNARE, RHOA and NPAS4. Finally, we provide a novel vision of the existing altered gene-gene crosstalk and molecular mechanisms targeting specific hubs in DS models that should become central to advance in our understanding of DS and therapies development. Highlights Brain function and morphology changes in DS mouse models result from multiple genetic loci Each combination of loci induces specific alteration of gene expression profile in mouse models Altered gene expression converges to a few functional pathwys in DS mouse hippocampi The synaptic pathway analysis leads to six connected biological cascades and defines a specific DS disease network
... Interestingly, as observed in the rotarod test, the locomotor phenotype of the Tc1 transchromosomic mouse model carrying an almost complete Hsa21 is rescued when the Abcg1-Cbs region is returned to two copies in Tc1/Ms2Yah mice (25). Similarly, the trisomy of a larger overlapping segment on Mmu17 from Abcg1 to Rrp1b induces an increased LTP as compared with controls in the Dp(17)Yey model (22) and was shown to genetically interact with the trisomy of the Lipi-Zbtb21 interval. ...
... The number of animals required for behavioural analysis was estimated according to similar experiments we previously performed in DS mouse models (5,21,25). All the mice used in the study were males and the controls were littermates. ...
Article
Full-text available
Identifying dosage-sensitive genes is a key to understand the mechanisms underlying intellectual disability in Down syndrome (DS). The Dp(17Abcg1-Cbs)1Yah DS mouse model (Dp1Yah) shows cognitive phenotypes that need to be investigated to identify the main genetic driver. Here, we report that three copies of the cystathionine-beta-synthase gene (Cbs) in the Dp1Yah mice are necessary to observe a deficit in the novel object recognition (NOR) paradigm. Moreover, the overexpression of Cbs alone is sufficient to induce deficits in the NOR test. Accordingly, overexpressing human CBS specifically in Camk2a-expressing neurons leads to impaired objects discrimination. Altogether, this shows that Cbs overdosage is involved in DS learning and memory phenotypes. To go further, we identified compounds that interfere with the phenotypical consequence of CBS overdosage in yeast. Pharmacological intervention in Tg(CBS) mice with one selected compound restored memory in the NOR test. In addition, using a genetic approach, we demonstrated an epistatic interaction between Cbs and Dyrk1a, another human chromosome 21-located gene (which encodes the dual-specificity tyrosine phosphorylation-regulated kinase 1a) and an already known target for DS therapeutic intervention. Further analysis using proteomic approaches highlighted several molecular pathways, including synaptic transmission, cell projection morphogenesis and actin cytoskeleton, that are affected by DYRK1A and CBS overexpression. Overall, we demonstrated that CBS overdosage underpins the DS-related recognition memory deficit and that both CBS and DYRK1A interact to control accurate memory processes in DS. In addition, our study establishes CBS as an intervention point for treating intellectual deficiencies linked to DS.
... These triplicated genes not only affect phenotypes directly, but also interact among themselves and with disomic genes throughout genomes with consequences exhibited at the molecular, cellular, and organismal levels. Some of these interactions have been speculated to occur based on established knowledge, while others have been demonstrated experimentally [18,[21][22][23][40][41][42]. To reflect these evolutionarily conserved interactions at all levels, it is desirable that all Hsa21 gene orthologs in an animal model are triplicated. ...
Article
Full-text available
Down syndrome (DS) is one of the most complex genetic disorders in humans and a leading genetic cause of developmental delays and intellectual disabilities. The mouse remains an essential model organism in DS research because human chromosome 21 (Hsa21) is orthologously conserved with three regions in the mouse genome. Recent studies have revealed complex interactions among different triplicated genomic regions and Hsa21 gene orthologs that underlie major DS phenotypes. Because we do not know conclusively which triplicated genes are indispensable in such interactions for a specific phenotype, it is desirable that all evolutionarily conserved Hsa21 gene orthologs are triplicated in a complete model. For this reason, the Dp(10)1Yey/+;Dp(16)1Yey/+;Dp(17)1Yey/+ mouse is the most complete model of DS to reflect gene dosage effects because it is the only mutant triplicated for all Hsa21 orthologous regions. Recently, several groups have expressed concerns that efforts needed to generate the triple compound model would be so overwhelming that it may be impractical to take advantage of its unique strength. To alleviate these concerns, we developed a strategy to drastically improve the efficiency of generating the triple compound model with the aid of a targeted coat color, and the results confirmed that the mutant mice generated via this approach exhibited cognitive deficits.
... Further investigations by our laboratory contributed to the analysis of the DS locomotor deficits identifying as new pathological mechanisms and adding to the complexity of the phenotype. Rescue experiment analysis by crossing the Tc1 transchromosomic model with a monosomic model for the Mmu17 Abcg1-U2af1 region partially rescued the locomotor deficit observed in the Tc1 model revealing a role of this region in locomotor function although the trisomic mouse model for this region did not present locomotor deficit (Marechal et al., 2015). We observed a mild locomotor deficit in trisomic mice for the Mmu16 proximal Hspa13-App region that was associated with decreased muscle endurance and oxidative capacity due to decreased mitochondrial content, shedding light on hypotonia and pointing at the Nrip1 gene as candidate for this phenotype (Brault et al., 2015). ...
Chapter
The genotype-phenotype relationship and the physiopathology of Down Syndrome (DS) have been explored in the last 20 years with more and more relevant mouse models. From the early age of transgenesis to the new CRISPR/CAS9-derived chromosomal engineering and the transchromosomic technologies, mouse models have been key to identify homologous genes or entire regions homologous to the human chromosome 21 that are necessary or sufficient to induce DS features, to investigate the complexity of the genetic interactions that are involved in DS and to explore therapeutic strategies. In this review we report the new developments made, how genomic data and new genetic tools have deeply changed our way of making models, extended our panel of animal models, and increased our understanding of the neurobiology of the disease. But even if we have made an incredible progress which promises to make DS a curable condition, we are facing new research challenges to nurture our knowledge of DS pathophysiology as a neurodevelopmental disorder with many comorbidities during ageing.
... Further investigations by our laboratory contributed to the analysis of the DS locomotor deficits identifying as new pathological mechanisms and adding to the complexity of the phenotype. Rescue experiment analysis by crossing the Tc1 transchromosomic model with a monosomic model for the Mmu17 Abcg1-U2af1 region partially rescued the locomotor deficit observed in the Tc1 model revealing a role of this region in locomotor function although the trisomic mouse model for this region did not present locomotor deficit (Marechal et al., 2015). We observed a mild locomotor deficit in trisomic mice for the Mmu16 proximal Hspa13-App region that was associated with decreased muscle endurance and oxidative capacity due to decreased mitochondrial content, shedding light on hypotonia and pointing at the Nrip1 gene as candidate for this phenotype (Brault et al., 2015). ...
Article
As one of the most common human genetic disorders, Down syndrome (DS) is characterized by a mild-to-moderate cognitive disability, which mainly results from genes overexpression on chromosome 21. The expression of miR-99a, a gene harboring on chromosome 21, is increased by 50 folds in DS brain samples. This study aims to investigate the effect of miR-99a overexpression in the hippocampus on mouse behaviors and explore the underlying mechanisms. Lentivirus vectors were delivered into the hippocampus for focal miR-99a overexpression in mice. Then behaviors were observed by an open field, elevated plus maze, rotarod motor test, and Morris water maze. The genes affected by miR-99a were identified by RNA sequencing (RNA-seq) and confirmed by quantitative RT-PCR (qRT-PCR) in samples isolated from the hippocampus injected with lentivirus-GFP-miR-99a or lentivirus-GFP vectors. It was found that the expression of miR-99a with intrahippocampal delivery of lentivirus-GFP-miR-99a resulted in reversal learning impairment in mice although it had no influence on motor function and anxiety. Meanwhile, RNA-seq results showed that 92 genes including mRNAs and microRNAs were significantly regulated by miR-99a, consistent with qRT-PCR consequence. Moreover, Dual-luciferase reporter assay showed that miR-99a binds directly to the 3’-untranslated regions (3’UTR) of target genes (Clic6 and Kcnj13) with an inhibitory effect on their activity. Furthermore, we also found that miR-99a overexpression affected different biological processes by bioinformatic analyses. Our study showed that miR-99a overexpression in the hippocampus leads to cognitive impairment through regulating the expressions of various genes, which reveals a novel function of miR-99a and provides new insights into understanding the pathophysiologic process of DS.
Preprint
Full-text available
Identifying dosage sensitive genes is a key to understand the mechanisms underlying intellectual disability in Down syndrome (DS). The Dp(17Abcg1-Cbs)1Yah DS mouse model (Dp1Yah) show cognitive phenotype and needs to be investigated to identify the main genetic driver. Here, we report that, in the Dp1Yah mice, 3 copies of the Cystathionine-beta-synthase gene ( Cbs) are necessary to observe a deficit in the novel object recognition (NOR) paradigm. Moreover, the overexpression of Cbs alone is sufficient to induce NOR deficit. Accordingly targeting the overexpression of human CBS, specifically in Camk2a-expressing neurons, leads to impaired objects discrimination. Altogether this shows that Cbs overdosage is involved in DS learning and memory phenotypes. In order to go further, we identified compounds that interfere with the phenotypical consequence of CBS overdosage in yeast. Pharmacological intervention in the Tg( CBS ) with one selected compound restored memory in the novel object recognition. In addition, using a genetic approach, we demonstrated an epistatic interaction between Cbs and Dyrk1a , another human chromosome 21 gene encoding the dual-specificity tyrosine phosphorylation-regulated kinase 1a and an already known target for DS therapeutic intervention. Further analysis using proteomic approaches highlighted several pathways, including synaptic transmission, cell projection morphogenesis, and actin cytoskeleton, that are affected by DYRK1A and CBS overexpression. Overall we demonstrated that CBS overdosage underpins the DS-related recognition memory deficit and that both CBS and DYRK1A interact to control accurate memory processes in DS. In addition, our study establishes CBS as an intervention point for treating intellectual deficiencies linked to DS. SIGNIFICANT STATEMENT Here, we investigated a region homologous to Hsa21 and located on mouse chromosome 17. We demonstrated using three independent genetic approaches that the overdosage of the Cystathionine-beta-synthase gene ( Cbs ) gene, encoded in the segment, is necessary and sufficient to induce deficit in novel object recognition (NR). In addition, we identified compounds that interfere with the phenotypical consequence of CBS overdosage in yeast and in mouse transgenic lines. Then we analyzed the relation between Cbs overdosage and the consequence of DYRK1a overexpression, a main driver of another region homologous to Hsa21 and we demonstrated that an epistatic interaction exist between Cbs and Dyrk1a affecting different pathways, including synaptic transmission, cell projection morphogenesis, and actin cytoskeleton.
Article
Full-text available
Down syndrome (DS) is due to increased copy number of human chromosome 21. The contribution of different genetic regions has been tested using mouse models. As shown previously, the Abcg1-U2af1 genetic region contributes to cognitive defects in working and short-term recognition memory in Down syndrome mouse models. Here we analyzed the impact of monosomy of the same genetic interval using a new mouse model, named Ms2Yah. We used several cognitive paradigms, and did not detect defects in the object recognition nor the Morris water maze tests. However, surprisingly, Ms2Yah mice displayed increased associative memory in a pure contextual fear conditioning test, and decreased social novelty interaction along with a larger long term potentiation recorded in the CA1 area following stimulation of Schaffer collaterals. Whole genome expression studies carried out on hippocampus showed that only the transcription of a small number of genes is affected, mainly from the genetic interval (Cbs, Rsph1, Wdr4), with a few additional ones, including the postsynaptic Gabrr2, Gabbr1, Grid2p, Park2 and Dlg1 and the components of the Ubiquitin mediated proteolysis (Anapc1, Rnf7, Huwe1, Park2). The Abcg1-U2af1 region is undeniably encompassing dosage sensitive genes or elements whose change in copy number directly impact learning and memory, synaptic function and autistic related behavior.
Article
Full-text available
Trisomy 21 is the most frequent genetic cause of cognitive impairment. To assess the perturbations of gene expression in trisomy 21, and to eliminate the noise of genomic variability, we studied the transcriptome of fetal fibroblasts from a pair of monozygotic twins discordant for trisomy 21. Here we show that the differential expression between the twins is organized in domains along all chromosomes that are either upregulated or downregulated. These gene expression dysregulation domains (GEDDs) can be defined by the expression level of their gene content, and are well conserved in induced pluripotent stem cells derived from the twins' fibroblasts. Comparison of the transcriptome of the Ts65Dn mouse model of Down's syndrome and normal littermate mouse fibroblasts also showed GEDDs along the mouse chromosomes that were syntenic in human. The GEDDs correlate with the lamina-associated (LADs) and replication domains of mammalian cells. The overall position of LADs was not altered in trisomic cells; however, the H3K4me3 profile of the trisomic fibroblasts was modified and accurately followed the GEDD pattern. These results indicate that the nuclear compartments of trisomic cells undergo modifications of the chromatin environment influencing the overall transcriptome, and that GEDDs may therefore contribute to some trisomy 21 phenotypes.
Article
Full-text available
Genetic factors are important in the pathogenesis of fractures. Notably, estrogen receptor α (ESR1) has been suggested as a possible candidate gene for hip fractures; however, published studies of ESR1 gene polymorphisms have been hampered by small sample sizes and inconclusive or ambiguous results. The aim of this meta-analysis is to investigate the associations between two novel common ESR1 polymorphisms (intron 1 polymorphisms PvuII-rs2234693: C>T and XbaI-rs9340799: A>G) and hip fracture. Crude odds ratios (ORs) with 95% confidence intervals (CIs) were used to evaluate the strength of the association. Five case-control and three cohort studies were assessed, including a total of 1,838 hip fracture cases and 14,972 healthy controls. This meta-analysis revealed that the PvuII T allele is a highly significant risk factor for hip fracture susceptibility, with an effect magnitude similar in male and pre-menopausal and post-menopausal female patients. In stratified analysis based on ethnicity, the PvuII T allele remained significantly correlated with increased risk of hip fracture in Caucasian populations; this correlation, however, was not found in Asian populations. Unlike the PvuII polymorphism, we did not find significant differences in the XbaI (A>G) polymorphism allele or genotype distributions of hip fracture patients and controls. We also found no obvious association between the XbaI polymorphism and hip fracture in any of the racial or gender subgroups. Our findings show that the ESR1 PvuII T allele may increase the risk of hip fracture and that the XbaI polymorphism is not associated with hip fracture.
Article
Full-text available
Abstract The influences of verbal mental age (VMA) and performance mental age (PMA) on cognitive flexibility were examined among a group of participants with Down syndrome (DS), in order to disentangle the relative contributions of each. The impaired cognitive flexibility typically observed among individuals with DS in combination with uneven VMA and PMA development suggests an opportunity to further understand the developmental relationship between VMA, PMA, and cognitive flexibility. We examined the performance of 22 participants with DS on the Flexible Item Selection Task (FIST), used for measuring cognitive flexibility among preschoolers. Partial correlations revealed that only VMA was related to the FIST after controlling for PMA, highlighting the role of verbal abilities in the development of cognitive flexibility.
Article
Full-text available
Down syndrome (DS) is caused by trisomy of chromosome 21 (Hsa21) and presents a complex phenotype that arises from abnormal dosage of genes on this chromosome. However, the individual dosage-sensitive genes underlying each phenotype remain largely unknown. To help dissect genotype - phenotype correlations in this complex syndrome, the first fully transchromosomic mouse model, the Tc1 mouse, which carries a copy of human chromosome 21 was produced in 2005. The Tc1 strain is trisomic for the majority of genes that cause phenotypes associated with DS, and this freely available mouse strain has become used widely to study DS, the effects of gene dosage abnormalities, and the effect on the basic biology of cells when a mouse carries a freely segregating human chromosome. Tc1 mice were created by a process that included irradiation microcell-mediated chromosome transfer of Hsa21 into recipient mouse embryonic stem cells. Here, the combination of next generation sequencing, array-CGH and fluorescence in situ hybridization technologies has enabled us to identify unsuspected rearrangements of Hsa21 in this mouse model; revealing one deletion, six duplications and more than 25 de novo structural rearrangements. Our study is not only essential for informing functional studies of the Tc1 mouse but also (1) presents for the first time a detailed sequence analysis of the effects of gamma radiation on an entire human chromosome, which gives some mechanistic insight into the effects of radiation damage on DNA, and (2) overcomes specific technical difficulties of assaying a human chromosome on a mouse background where highly conserved sequences may confound the analysis. Sequence data generated in this study is deposited in the ENA database, Study Accession number: ERP000439.
Article
Full-text available
A mouse model for Down syndrome, Ts1Cje, has been developed. This model has made possible a step in the genetic dissection of the learning, behavioral, and neurological abnormalities associated with segmental trisomy for the region of mouse chromosome 16 homologous with the so-called “Down syndrome region” of human chromosome segment 21q22. Tests of learning in the Morris water maze and assessment of spontaneous locomotor activity reveal distinct learning and behavioral abnormalities, some of which are indicative of hippocampal dysfunction. The triplicated region in Ts1Cje, from Sod1 to Mx1, is smaller than that in Ts65Dn, another segmental trisomy 16 mouse, and the learning deficits in Ts1Cje are less severe than those in Ts65Dn. In addition, degeneration of basal forebrain cholinergic neurons, which was observed in Ts65Dn, was absent in Ts1Cje.
Article
Full-text available
Impaired left ventricular (LV) function has been shown by strain rate (SR) imaging in patients with coronary artery disease (CAD). Our aim was to investigate global and regional, systolic and diastolic left atrial (LA) and right atrial (RA) longitudinal deformation in CAD using velocity vector imaging. Echocardiographic and velocity vector imaging studies were performed in 20 patients with mild CAD, 40 patients with severe CAD and 25 controls. Maximal atrial volume, peak atrial longitudinal strain (ε(s)) and SR during LV systole (SRs), SR during early LV filling (SRe) and late LV filling (SRa) were measured. Longitudinal strain during atrial contraction (ε(a)) was obtained at the onset of P-wave on electrocardiography, and ε(a)/ε(s) was calculated. Longitudinal peak ε(s) and SRs of LA showed decreased trend among CAD patients. The global and lateral LA SRe were prominently lower, while RA ε(a), SRa and ε(a)/ε(s) were prominently higher in 2 CAD groups than control group (P value <0.05). As compared with controls and patients with other single-vessel disease, LA SRa and ε(a)/ε(s) ratio were significantly increased among patients with exclusively left anterior descending coronary artery (LAD) stenosis (SRa 1.14±0.38 s(-1), 1.10±0.41 s(-1), 1.45±0.46 s(-1), P value<0.05; ε(a)/ε(s) 0.44±0.11, 0.44±0.20, 0.57±0.12, P value<0.01). Apparently decreased SRe of LA and increased ε(a), SRa and ε(a)/ε(s) of RA were found in CAD patients with preserved LVEF and E/E' in gray zone. SRa and ε(a)/ε(s) of LA were found to significantly increase in those with LAD stenosis.
Article
Full-text available
The X chromosome is present as a single copy in the heterogametic sex, and this hemizygosity is expected to drive unusual patterns of evolution on the X relative to the autosomes. For example, the hemizgosity of the X may lead to a lower chromosomal effective population size compared to the autosomes, suggesting that the X might be more strongly affected by genetic drift. However, the X may also experience stronger positive selection than the autosomes, because recessive beneficial mutations will be more visible to selection on the X where they will spend less time being masked by the dominant, less beneficial allele-a proposal known as the faster-X hypothesis. Thus, empirical studies demonstrating increased genetic divergence on the X chromosome could be indicative of either adaptive or non-adaptive evolution. We measured gene expression in Drosophila species and in D. melanogaster inbred strains for both embryos and adults. In the embryos we found that expression divergence is on average more than 20% higher for genes on the X chromosome relative to the autosomes; but in contrast, in the inbred strains, gene expression variation is significantly lower on the X chromosome. Furthermore, expression divergence of genes on Muller's D element is significantly greater along the branch leading to the obscura sub-group, in which this element segregates as a neo-X chromosome. In the adults, divergence is greatest on the X chromosome for males, but not for females, yet in both sexes inbred strains harbour the lowest level of gene expression variation on the X chromosome. We consider different explanations for our results and conclude that they are most consistent within the framework of the faster-X hypothesis.