You are on page 1of 14

International Journal of Developmental Neuroscience 67 (2018) 19–32

Contents lists available at ScienceDirect

International Journal of Developmental Neuroscience


journal homepage: www.elsevier.com/locate/ijdevneu

Beneficial effects of environmental enrichment on behavior, stress reactivity T


and synaptophysin/BDNF expression in hippocampus following early life
stress
Εvgenia Dandia, Aikaterini Kalamaria, Olga Touloumic, Rosa Lagoudakic,
⁎ ⁎
Evangelia Nousiopoulouc, Constantina Simeonidoub, Evangelia Spandoub, , Despina A. Tataa,
a
Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki 541 24, Greece
b
Laboratory of Experimental Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 541 24, Greece
c
Laboratory of Neuroimmunology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 541 24, Greece

A R T I C LE I N FO A B S T R A C T

Keywords: Exposure to environmental enrichment can beneficially influence the behavior and enhance synaptic plasticity.
Early stress The aim of the present study was to investigate the mediated effects of environmental enrichment on postnatal
Enriched environment stress-associated impact with regard to behavior, stress reactivity as well as synaptic plasticity changes in the
Visuospatial learning dorsal hippocampus. Wistar rat pups were submitted to a 3 h maternal separation (MS) protocol during postnatal
Spatial memory
days 1–21, while another group was left undisturbed. On postnatal day 23, a subgroup from each rearing
Recognition memory
Corticosterone
condition (maternal separation, no-maternal separation) was housed in enriched environmental conditions until
postnatal day 65 (6 weeks duration). At approximately three months of age, adult rats underwent behavioral
testing to evaluate anxiety (Elevated Plus Maze), locomotion (Open Field Test), spatial learning and memory
(Morris Water Maze) as well as non-spatial recognition memory (Novel Object Recognition Test). After com-
pletion of behavioral testing, blood samples were taken for evaluation of stress-induced plasma corticosterone
using an enzyme-linked immunosorbent assay (ELISA), while immunofluorescence was applied to evaluate
hippocampal BDNF and synaptophysin expression in dorsal hippocampus. We found that environmental en-
richment protected against the effects of maternal separation as indicated by the lower anxiety levels and the
reversal of spatial memory deficits compared to animals housed in standard conditions. These changes were
associated with increased BDNF and synaptophysin expression in the hippocampus. Regarding the neuroendo-
crine response to stress, while exposure to an acute stressor potentiated corticosterone increases in maternally-
separated rats, environmental enrichment of these rats prevented this effect. The current study aimed at in-
vestigating the compensatory role of enriched environment against the negative outcomes of adverse experi-
ences early in life concurrently on emotional and cognitive behaviors, HPA function and neuroplasticity markers.

1. Introduction paradigm. During the first postnatal days, survival of rat pups depends
exclusively on maternal care. In addition, the role of tactile stimulation,
It is well established that environmental factors during the postnatal nourishing and passive contact is considered crucial for the regulation
period have long term effects on behavior, neuroendocrine function and of Hypothalamic-Pituitary-Adrenal (HPA) axis (Levine, 2002, 2001).
neuronal plasticity (Bock and Braun, 2011; Stiles, 2011). Epidemiolo- Prolonged MS disrupts the normal interaction between mother and
gical and clinical studies support the idea that stress early in life may pups and affects brain and behavior. Reduction in neurogenesis (Korosi
lead to psychopathology in adulthood (Anda et al., 2009; Felitti et al., et al., 2012; Lajud et al., 2012) and glucocorticoid receptors’ density
1998; Heim and Binder, 2012; Heim and Nemeroff, 2001; Palagini (Aisa et al., 2008; Enthoven et al., 2008a,b) has been reported as a
et al., 2015). A well-established animal model of early stress that is result of MS. Early life stress also reduces synaptic formation and en-
widely used over the last decades is the maternal separation (MS) hances neuroendocrine stress response (Bock and Braun, 2011). At

Abbreviations: BDNF, brain-derived neurotrophic factor; CORT, corticosterone; EPM, elevated plus maze; EE, environmental enrichment; MS, maternal separation; NMS, no maternal
separation; OFT, open field test; PND, postnatal day; SYN, synaptophysin

Corresponding authors.
E-mail addresses: spandou@med.auth.gr (E. Spandou), dtata@psy.auth.gr (D.A. Tata).

https://doi.org/10.1016/j.ijdevneu.2018.03.003
Received 30 November 2017; Received in revised form 7 March 2018; Accepted 8 March 2018
Available online 12 March 2018
0736-5748/ © 2018 ISDN. Published by Elsevier Ltd. All rights reserved.
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

behavioral level, maternally separated rats show increased anxiety and expression of these two proteins in the hippocampus, a structure par-
depressive-like behaviors in adulthood (Aisa et al., 2007; Veenema ticularly vulnerable to stress.
et al., 2007; Vetulani, 2013; Wang et al., 2015a; Wigger and Neumann,
1999). Several studies have also reported impairments in various cog- 2. Materials and method
nitive functions (e.g., learning and memory), following early life stress
(Chocyk et al., 2014; Conrad, 2010; Cui et al., 2006; Mcewen, 1997; 2.1. Animals
Tata et al., 2015; Xiong et al., 2015). Based on existing evidence, these
effects have been associated with alterations in the expression of sy- Female Wistar rats on the second gestational week were in-
naptic plasticity markers, such as the brain-derived neurotrophic factor dividually housed until delivery. The day of birth was designated as
(BDNF) and synaptophysin (SYN). BDNF, a main neurotrophin in postnatal day 0 (PND 0). Totally, 47 neonates were included in the
mammals’ central nervous system, is essential for cell survival and experiment and all animals participated in behavioral testing, as well as
regulation of dendritic and synaptic plasticity, mainly in the hippo- in corticosterone and BDNF/synaptophysin measurements. In order to
campus (Fumagalli et al., 2007; Lu et al., 2005), while synaptophysin, ensure normal growth and development, body weight measurements of
an indirect indicator of the synapse number, is a synaptic vesicle pro- neonates were taken on the 3rd, 8th, 14th and 21st postnatal days. All
tein involved in neurotransmission (Valtorta et al., 2004). It has been animals were maintained on a 12:12 light/dark cycle (08:00 light/
shown that adult rats exposed to MS condition exhibited reduced ex- 20:00 dark) and standard temperature (22 ± 2 °C), with food and
pression of BDNF and SYN in various brain regions (Andersen and water available ad libitum. Handling of the pups and behavioral testing
Teicher, 2004; Choy et al., 2008; Lippmann et al., 2007; Roth et al., were performed by the same personnel since familiarity with the ex-
2009). perimenter increases consistency in animal testing (van Driel and
In contrast to the effects of early stress, specific housing conditions, Talling, 2005). Εxperimental procedures were conducted in accordance
such as environmental enrichment (EE), can exert a neuroprotective to the Institutional Animal Ethics EL 54 BIO 20.
role (Baroncelli et al., 2010; Gelfo et al., 2011; Jha et al., 2011). En-
vironmental enrichment (EΕ) promotes neurogenesis (Monteiro et al., 2.2. Rearing conditions
2014) and synaptogenesis (Rampon et al., 2000), enhances hippo-
campal LTP (Cortese et al., 2018) and increases dendritic arborization On PND 1 litters were assigned randomly to no-maternal separation
as well as spine density, thus facilitating neuronal communication (NMS) (NS = 23) or maternal separation (MS) (N = 24) conditions. The
(Leggio et al., 2005). In addition, it improves learning and memory pups of the NMS condition remained undisturbed in their cages with
(Frick and Fernandez, 2003; Harburger et al., 2007; Leggio et al., 2005; their dams until weaning (PND 23). The MS condition involved a daily
Rampon and Tsien, 2000) and protects from cognitive deficits caused 3 h separation of the pups from their dams during the three postnatal
by brain damage or exposure to stress (Hralová et al., 2013; Nozari weeks (PND1-21) (Huot et al., 2001). The maternal separation proce-
et al., 2014; Schreiber et al., 2014; Wright and Conrad, 2008) or aging dure took place between 0900 and 1400 h and was performed as pre-
(Cortese et al., 2018). Furthermore, enriched housing promotes social viously described (Tata et al., 2015). Briefly, dams were first removed
interaction, locomotor activity and exploratory behavior (Brenes Sáenz from their cages, followed by the pups, which were placed, as a litter, in
et al., 2006) and regulates emotional behavior. Rodents housed in EE plastic containers. Upon completion of the 3 h separation period, pups
show decreased anxiety and depressive-like behaviors in various be- were returned to their home cage, followed by their dam. Litter size
havioral tasks (Chapillon et al., 1999; Friske and Gammie, 2005; ranged from 6 to 10 rats.
Ishihama et al., 2010; Nicolas et al., 2015). These effects seem to be
associated with a number of neuronal and neuroendocrine changes that 2.3. Post-weaning housing conditions
may, in turn, result in greater resistance to stress later in life (Connors
et al., 2014; Crofton et al., 2015; Fox et al., 2006). On PND 23 rats from each of the two rearing conditions were ran-
These findings support the hypothesis that EE could compensate for domly assigned to either standard housing (SH) (N = 22) or enriched
the detrimental effects of stress. In fact, existing evidence indicates that environment (EE) conditions (N = 25). In the SH condition, rats in
EE attenuates the chronic stress impact on behavior and hippocampal same-gender pairs were housed in standard laboratory cages. In the EE
integrity, thus protecting against the stress-associated cognitive deficits, condition (PND23-65), same-gender groups of 5–7 rats were housed in
emotional dysregulation, dendritic atrophy and reduced neurogenesis large cages (60 cm × 45 cm × 76 cm) containing various non chewable
(Hutchinson et al., 2012; Veena et al., 2009; Wright and Conrad, 2008). toys, climbing platforms, tunnels and running wheels (Griva et al.,
Similar to adult stress, restorative effects of EE have been reported in 2017). To promote exploratory and novelty-seeking behavior, every
rodents submitted to prenatal (Laviola et al., 2008; Pascual et al., 2015; 3–4 days toys as well as food and water containers were moved to a new
Yang et al., 2007) or juvenile stress (Ilin and Richter-Levin, 2009). To location in the cage. Once a week the cages were cleaned and platforms,
the best of our knowledge, there is limited information regarding the tunnels and running wheels were rearranged, and toys were replaced by
compensatory action of EE following early stress (do Prado et al., 2016; new ones. On PND 65, animals of the EE condition were transferred in
Francis et al., 2002; Koe et al., 2016; Vivinetto et al., 2013) and so far, pairs to standard laboratory cages, where they were kept until beha-
no study has looked at the interaction of maternal separation and vioral testing began.
subsequent exposure to EE conditions concurrently at cognitive and The final four groups that emerged after experimental manipula-
emotional behavior, HPA function and markers of synaptic plasticity tions were the following: a) non-maternally separated rats housed in
(BDNF, SYN) during adulthood. The critical importance of the first standard housing conditions (NMS/SH; N = 10), b) non-maternally
postnatal weeks to normal development (Rice and Barone, 2000), fur- separated rats housed in enriched environment condition (NMS/EE;
ther supports the significance of exploring the hypothesis that EE ex- N = 13) c) maternally-separated rats housed in standard housing con-
perience may overcome behavioral and neurobiological deficits in- ditions (MS/SH; N = 12), d) maternally-separated rats housed in en-
duced by early postnatal stress. riched environment (MS/EE; N = 12). The animals of each experi-
The purpose of the current study was to explore the hypothesis that mental group were obtained from 2 to 3 litters.
EE during adolescence may protect against the negative outcomes of
maternal separation on spatial and non-spatial learning and memory, 2.4. Behavioral testing
anxiety, as well as neuroendocrine stress response during adulthood.
Furthermore, given the essential role of BDNF and SYN in cognitive Behavioral testing took place at approximately 2.5 months of age in
functions and emotional behavior, we aimed at exploring the order to examine emotional and exploratory behavior as well as

20
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

learning and memory. Anxiety-like behavior was assessed by the corners of the arena at a distance of 25 cm from the wall. The objects
Elevated Plus Maze (EPM) (PND 67), locomotion and exploratory be- used were made of metal or glass and were heavy enough so that they
havior was estimated by the Open Field Test (OFT) (PND 69), while could not be displaced by the rats. In addition, we ensured that the
non-spatial and spatial (visual) learning and memory were assessed by objects were of similar attractiveness for the rats. Object exploration
the Novel Object Recognition (NORT) (PND 70) and the Morris Water was operationally defined as directly attending to the object with the
Maze (MWM) (PND72-76) tests, respectively. Behavioral testing oc- head no more than 2 cm from the object (Ennaceur and Delacour,
curred during the light cycle between 09:00–15:00. Rat behavior was 1988). In trial 2 (choice phase; 3 min duration) one of the two objects
recorded by means of a ceiling-mounted camera placed 160 cm above was replaced by a new one of similar height (Bevins and Besheer, 2006;
the experimental arena. Animals’ swimming behavior in the MWM was Ennaceur and Delacour, 1988). Under normal conditions, rats tend to
analyzed by a data acquisition system (Ethovision v.2.3, Noldus explore more the novel object, a preference that is indicative of non-
Information Technology), while in case of NORT and EPM testing re- spatial memory. The length of the specific inter-trial interval (ITI) was
corded behavior was manually scored by two researchers. based on finding that under control conditions rats can still recognize
Experimenters were blind to experimental conditions both during the novel object and subsequently the danger of floor effects due to
behavioral testing and data analysis. All experimental apparatuses were longer ITIs is eliminated (Ennaceur et al., 1997). In the current study,
cleaned thoroughly with a 25% ethanol solution after each trial to we estimated the discrimination ratio, defined as the exploration time
eliminate odor cues. of the novel object in trial 2 divided by the total time exploring both
objects in the same trial. Exploration time of each object during the
2.4.1. Elevated plus maze (EPM) sample phase was also calculated in order to ensure that any preference
EPM is a behavioral test that is used to measure anxiety (Handley shown for the new object would be due to its novelty and not to dif-
and Mithani, 1984) based on rodents’ innate preference of dark and ference in the exploration during the sample phase.
enclosed spaces and their unconditioned fear of height/open areas. The
apparatus was cross-shaped, with two open and two closed arms
(50 cm × 10 cm); the height of the walls was 40 cm and the maze was 2.4.4. Morris water maze (MWM)
positioned 50 cm from the floor. The animals were placed in the open Morris Water Maze is used to estimate spatial learning and memory
central square (10 cm × 10 cm) formed at the four arms intersection, (Morris, 1984). In the present study the maze consisted of a plastic
facing the open arm opposite to experimenter, and were left un- circular tank (1.6 m/diameter × 50 cm/height) filled with water in
disturbed for 5 min to explore the maze. Increased time and number of standard temperature (25 ± 1 °C) and was virtually divided into four
entries in the open arms (Walf and Frye, 2007) as well as smaller oc- quadrants: north-west (NW), north-east (NE), south-west (SW) and
currence of risk assessment behaviors (i.e., stretch attend posture, south-east (SE). An escape platform (10 cm × 10 cm) made of trans-
closed arms returns) (Cruz et al., 1994; Doremus et al., 2006; Rodgers parent Plexiglas was placed in the middle of one of the quadrants (SW),
and Dalvi, 1997) indicate lower levels of anxiety. In the present study half-way between the center and the wall, and 1.5 cm below the water
assessment of anxiety was based on a) the ratio of open arms entries surface. Pictures on the walls and the furniture of the room were used as
(open arms entries/total entries), b) the ratio of the open arms time extra-maze cues. Animals entered the maze facing the wall of the pool
(open arms time/total arms time), c) the number of stretch attend and were allowed to swim for 60 s in order to locate the submerged
posture/SAP (the animal’s two hind legs remain in an arm while it platform. In case an animal failed to locate the platform within the
elongates its head and shoulders out of the arm and then returns to its specified period, it was gently guided to the platform by the experi-
initial position) and d) the number of returns to closed arms (the ani- menter; once the animal had reached the platform it was allowed to
mal’s half front body exits a closed arm followed by return to the closed remain there for 15 s. The interval between trials of each day was 1 min
arm). In addition, the total number of arm entries was estimated as an (including the 15 s period spent on the platform). The animals’ swim-
index of general activity. ming behavior was recorded by a video camera connected to a data
acquisition system (Ethovision v.2.3, Noldus Information Technology).
2.4.2. Open field test (OFT)
Open field test is used to measure locomotion and exploration (Lau
et al., 2008). The apparatus used in the current study consisted of a 2.4.4.1. Spatial acquisition phase (spatial learning). In this phase animals
wooden square arena (100 cm × 100 cm) surrounded by 40 cm high had to learn to navigate to the platform based on external cues. Spatial
wall to prevent escape. The arena’s floor was divided into 16 equally acquisition phase lasted 4 days and animals were given 4 trials/day
sized sectors (25 cm × 25 cm). The four squares in the center formed (totally 16 trials). The platform was permanently located in the SW
the inner zone of the arena, while the twelve outer squares formed the quadrant of the maze and start position was different for each trial. A
outer zone (peripheral squares). Rats were placed facing the wall and semi-random approach of start positions for each trial was chosen
they were allowed to freely explore the arena for 6 min. (Vorhees and Williams, 2006). Time to reach the platform (escape
Two behavioral parameters were analyzed in order to estimate latency; s) was measured to estimate visuospatial learning. Normally,
general activity and exploration, the number a) of square visits (am- animals spend less time to locate the platform from day to day as a
bulation) (Bubser et al., 1992; Suchecki et al., 2000) and b) rearings result of learning (Morris, 1984). Swim speed (velocity; cm/s) was also
(Gamberini et al., 2015; Kulesskaya and Voikar, 2014), respectively. A estimated in order to ensure that any differences in time to locate the
square visit was recorded if at least half of animal’s torso was in a platform was attributed to learning acquisition and not to differences in
sector, while rearing when animals kept hind paws on the floor with the swimming speed.
front limbs off it.

2.4.3. Novel object recognition test (NORT) 2.4.4.2. Probe trial. In order to assess spatial memory, a probe trial was
This task was administered in order to examine the non-spatial administered 24 h after the last acquisition day. During this trial (60 s),
episodic memory (Ennaceur and Meliani, 1992). It took place in a the escape platform was removed and each animal entered the maze
wooden open field arena (see above, 2.4.2.), and it was preceded by two from a novel position (180° from the platform position during the
habituation sessions (6 min duration each). acquisition phase). The time spent to the target quadrant (SW) as well
The testing consisted of two trials separated by a 70 min delay. In as the frequency of entries in the area where the platform used to be
trial 1 (sample phase; 4 min duration), rats were allowed to freely ex- located during the acquisition phase were used as indices of spatial
plore two identical objects that were placed at the two adjacent back memory (D’Hooge and De Deyn, 2001; Shinohara and Hata, 2014).

21
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

Fig. 1. (A) Entries and (B) time spent in the open


arms expressed as a ratio of the total entries and time
spent in both open and closed arms of the EPM.
Maternal separation significantly decreased number
of entries and time spent in the open arms in SH
animals (NMS/SH vs MS/SH, entries:**p < 0.001,
time:*p < 0.01), but exposure to EE reversed this
outcome (MS/SH vs. MS/EE, #p < 0.05). (C)
Number of returns to closed arms of the EPM. MS
animals reared in standard housing performed more
closed arms returns, compared to NMS treated ani-
mals (NMS/SH vs. MS/SH: *p < 0.01). Exposure of
maternally-separated animals to EE decreased the
number of returns compared to MS animals of the SH
condition (MS/SH vs. MS/EE: #p < 0.001). (D)
Among animals raised in SH condition, MS rats ex-
hibited increased number of Stretch-Attend Postures
(SAPs) in relation to non-maternally separated ani-
mals (NMS/SH vs. MS/SH, *p < 0.01), but EE re-
duced occurrence of this behavior (MS/SH vs. MS/
EE: #p < 0.001).

2.5. Corticosterone assessment microscope Zeiss Axioplan-2 using a CCD camera (Nikon DS–5 M). Two
sections per animal were chosen with care to allow comparison of si-
One week after completion of behavioral testing blood was collected milar regions across experimental conditions. Immunoreactivity for
from the rats’ carotid artery in order to determine corticosterone levels BDNF and SYN was estimated within the CA3, CA2 and CA1 stratum
before and after stress induction. Stress was induced by placing the rat radiatum and the dentate gyrus (DG) molecular layer with 40x objec-
cage on a rotating disk (78 revolutions per minute) for a 15 min period. tive lens. Approximately sixteen microscopic fields per section/animal
According to existing evidence, cage rotation for durations of 10 or were analyzed. Expression of BDNF and SYN was measured using the
20 min can produce significant increase in circulating corticosterone ImageJ software (version 1.45b, NIH) and is presented as mean
concentrations (Gein and Sharavieva, 2016; Riley, 1981). In order to Integrated Density (arbitrary density units) of all four hippocampal
avoid circadian variability, blood sampling was carried out between subregions.
0900 and 1100 for all control and experimental groups. Samples were
centrifuged (12.000 r.p.m. for 10 min at 4 °C) and plasma was extracted 2.7. Statistical analyses
and maintained at −80 °C until assay. Corticosterone concentrations
were assessed using an enzyme-linked immunosorbent assay (ELISA) kit All statistical analyses were performed using the statistical program
(MB Biomedicals, 07DE9922). SPSS Statistics (v. 22). Effects of the two experimental conditions
(rearing: NMS, MS and housing: SH, EE) on behavior (elevated plus
2.6. Tissue preparation and immunofluorescence maze, open field test, novel object recognition, MWM retention phase),
plasma corticosterone levels and immunofluorescence markers (BDNF,
All animals aged approximately 3.5 months were euthanized fol- SYN) were tested using 2 × 2 ANOVAs with rearing and housing as the
lowing anaesthetization. Brains were removed immediately and post- between-subjects factors. A three factor repeated-measures ANOVA
fixed (4% paraformaldehyde in 0.1 M phosphate-buffer saline, 3 × 24 h (between-subjects factors: rearing, housing; within-subjects factor: day)
at 4 °C). Coronal blocks were gradually hydrated and embedded in was applied to analyze the latency to locate the platform (MWM; ac-
paraffin. Serial coronal sections of 5 μm thick were taken at the level of quisition phase). Body weight measurement during the first three
dorsal hippocampus (−3.24 mm to −3.36 mm posterior to bregma) postnatal weeks (3rd, 8th, 14th, 21st postnatal days) were analyzed by
(Paxinos and Watson, 2007). Expression of BDNF and SYN on these a two factor repeated-measures ANOVA (between-subjects factors:
sections was evaluated using immunofluorescence. Following depar- rearing; within-subjects factor: postnatal day). Pairwise comparisons
affinization and hydration, steamer was used for antigen retrieval with Bonferroni correction were used in order to explore simple effects
(pH = 6, 1 h). Next, sections were incubated to blocking buffer (10% (i.e., effect of each factor within each level of the other factor). Data are
Fetal Bovine Serum, 2% Normal Goat Serum, 30 min) and treated with a presented as mean values ( ± SEM). Statistical significance was set at
primary antibody against BDNF (1:400, rabbit polyclonal, Santa Cruz), a < 0.05 for all measures.
or synaptophysin (1:100, mouse monoclonal, CloneSY38, DAKO)
overnight (4 °C). Depending on the primary antibody, sections were 3. Results
exposed to goat anti-rabbit IgC (Biotium 488) or goat Anti-MouseIgG
(Biotium 555) for BDNF or SYN, respectively. Nuclei were counter- 3.1. Body weight
stained with DAPI and mounted with the corresponding Biotium
medium (Biotium 23004). A repeated-measures analysis showed a significant increase in body
Tissue images were captured from sections with a fluorescent weight over the three postnatal weeks [F(3, 126) = 1593.8, p < 0.001,

22
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

0.327 (0.021)] (Fig. 1B).The two manipulations significantly interacted


with each other [MS × EE: F(1, 43) = 52.85, p < 0.001, partial
η2 = 0.551]. Maternal separation in standard-housed condition de-
creased the time spent in open arms [F(1, 43) = 19.47, p < 0.01,
partial η2 = 0.19, NMS/SH: 0.276 (0.033) vs. MS/SH: 0.134 (0.030)].
Housing of MS rats in EE condition significantly reversed the decreased
time seen in MS animals that lived in SH conditions [F(1, 43) = 65.264,
p < 0.001; MS/SH: 0.134 (0.030) vs. MS/EE: 0.478 (0.030)].
Closed arm returns: Housing conditions affected the specific beha-
vior, with animals exposed to EE performing less returns compared to
SH animals [F(1, 43) = 23.48, p < 0.001, partial η2 = 0.353]
(Fig. 1C). Results revealed a tendency of the effect of rearing on closed
arm returns, which did not reach statistical significance [F(1,
43) = 3.73, p = 0.06, partial η2 = 0.08]. The two factors interacted
with each other [F(1, 43) = 8.23, p < 0.01, partial η2 = 0.161]. Spe-
cifically, MS increased closed arms returns for SH animals [F(1,
43) = 10.793, p < 0.01; NMS/SH: 2.5 (0.525) vs. MS/SH: 4.833
(0.479)], but exposure of MS rats to EE condition totally reversed this
effect [F(1, 43) = 16.898, p < 0.001; MS/SH: 4.833 (0.479) vs. MS/
EE: 1.083 (0.479)].
Stretch Attend Posture (SAP): There were no significant effects for
the number of SAPs caused by either the rearing or housing manip-
ulations [MS: F(1, 43) = 0.276, p > 0.05, partial η2 = 0.006; EE: F(1,
43) = 2.28, p > 0.05, partial η2 = 0.05]. However, the effect of EE
differed as a function of rearing, as indicated by the significant inter-
action [F(1, 43) = 17.78, p < 0.001, partial η2 = 0.292]. MS increased
the SAPs in the standard-housed condition [F(1, 43) = 10.532,
p < 0.01, partial η2 = 0.197; NMS/SH: 2.8 (0.785) vs. MS/SH: 6.25
(0.717)]. While EE condition did not affect occurrence of this behavior
in the NMS rats [F(1, 43) = 3.556, p > 0.05, partial η2 = 0.076], it
caused a significant decrease in the MS groups [F(1, 43) = 16.898,
p < 0.001, partial η2 = 0.282; MS/SH: 6.25 (0.717), MS/EE: 2.083
(0.717)] (Fig. 1D).
Total arm entries: Neither MS nor EE conditions affected the total
arm entries [MS: F(1, 43) = 0.110, p > .05, partial η2 = 0.003, NMS:
16.45 (0.641), MS: 16.75 (0.622); EE: F (1, 43) = 0.092, p > .05,
partial η2 = 0.002, SH: 16.47 (0.653), EE: 16.74 (0.61)].
Fig. 2. (A) Number of rearings in the outer zone of the open field during the 6-min period.
Environmental enrichment significantly increased the number of rearings (#p < 0.05, EE
main effect). (B) Total entries (ambulation). There were no significant effects for the total 3.3. Open field
number of square visits caused by either the MS or EE manipulations (p > 0.05) and the
two factors did not interact with each other (p > 0.05) (Fig. 2B). Rearings (vertical activity): Analysis of rearings performed in the
outer zone revealed a significant effect of EE [F(1, 43) = 4.943,
partial η2 = 0.974; 3rd day: 9.19 (0.255), 8th day: 17.43 (0.481), 14th p < 0.05, partial η2 = 0.103]. EE treated animals performed more
day: 30.65 (0.783), 21st day: 47.1 (0.974)]. Maternal separation did not rearings than SH treated animals [EE: 12.18 (1.22), SH: 8.22 (1.3)]. MS
affect the body weight [F(1, 42) = 1.602, partial η2 = 0.037] neither condition did not affect vertical activity [F(1, 43) = 2.264, p > 0.005,
interacted with postnatal day [F(3, 126) = 2.541, p > 0.05, partial partial η2 = 0.050]. The effect of EE was not a function of rearing, as
η2 = 0.057]. indicated by the non-significant interaction [F(1, 43) = 0.064,
p > 0.05, partial η2 = 0.001] (Fig. 2A).
Ambulation: There were no significant effects for the total number
3.2. Elevated plus maze of square visits caused by either the MS [F(1, 43) = 1.373, p > 0.05,
partial η2 = 0.031] or EE manipulations [F(1, 43) = 2.103, p > 0.05,
Ratio of open arm entries: Analysis revealed a significant main effect partial η2 = 0.047], and the two factors did not interact with each other
of EE [F(1, 43) = 11.68, p < 0.01, partial η2 = 0.214; SH: [F(1, 43) = 0.101, p > 0.05, partial η2 = 0.002] (Fig. 2B).
0.262(0.021), EE: 0.361(0.02)], but not of MS condition [F(1,
43) = 0.23, p > 0.05, partial η2 = 0.005, NMS: 0.305 (0.021.), MS: 3.4. Novel object recognition task (NORT)
0.319 (0.02)] (Fig. 1A). Pairwise comparisons to explore the significant
MS × EE interaction [F(1, 43) = 47.97, p < 0.001, partial η2 = 0.527] MS or EE did not differentiate the groups in exploration time of the
showed that while maternal separation in standard-housed animals object A1 [MS: F(1, 43) = 3.102, p > 0.05, partial η2 = 0.067; EE: F(1,
decreased the number of entries [F (1, 43) = 19.47, p < 0.001, partial 43) = 0.188, p > 0.05, partial η2 = 0.004] or object A2 [MS: F(1,
η2 = 0.312, NMS/SH: 0.355 (0.031) vs. MS/SH: 0.169(0.028)], this 43) = 2.431, p > 0.05, partial η2 = 0.054; EE: F(1, 43) = 0.520,
effect was completely reversed by EE housing [F(1, 43) = 55.13, p > 0.05, partial η2 = 0.012] during the sample phase (trial 1).
p < 0.001, MS/SH: 0.169 (0.028) vs. MS/EE: 0.468 (0.028)]. Analysis of the exploration time from the choice phase (trial 2) in-
Ratio of time spent in open arms: Both MS and EE conditions af- dicated no significant effect of MS [F(1, 40) = 0.494, p > 0.05, partial
fected time spent in open arms [MS: F(1, 43) = 6.90, p < 0.05, partial η2 = 0.012], EE [F(1, 40) = 1.635, p > 0.05, partial η2 = 0.039] or a
η2 = 0.138, NMS: 0.226 (0.022.), MS: 0.306 (0.021); EE: F(1, significant interaction between the two factors [F(1, 40) = 1.48,
43) = 15.87, p < 0.001, partial η2 = 0.270, SH: 0.205 (0.022), EE: p > 0.05, partial η2 = 0.036] on discrimination ratio (Fig. 3). This

23
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

p > 0.05, partial η2 = 0.011, day 3: F(1, 43) = 0.82, p > 0.05, partial
η2 = 0.019, day 4: F(1, 43) = 2.76, p > 0.05, partial η2 = 0.06].
Probe trial: A two-way ANOVA revealed a significant effect for MS
on time spent in the target quadrant (SW) [F(1, 43) = 6.74, p < 0.05,
partial η2 = 0.135] as well as on frequency of entries into the platform
area [F(1, 43) = 9.113 p < 0.01, partial η2 = 0.175] (Fig. 5). MS an-
imals spend less time and did fewer entries than the non-maternally
separated animals. The EE condition did not affect any of the two
measures [time: F(1, 43) = 0.43, p > 0.05, partial η2 = 0.01; fre-
quency: F(1, 43) = 0.866, p > 0.05, partial η2 = 0.020]. Interestingly,
housing in EE seems to protect against stress-induced impairments, as
indicated by the significant interaction between MS and ΕΕ for the
duration [F(1, 43) = 7.57, p < 0.01, partial η2 = 0.150] and fre-
quency of entries [F(1, 43) = 4.474, p < 0.05, partial η2 = 0.094].
Specifically, MS rats raised in standard housing conditions exhibited
poorer performance compared to NMS rats as indicated by duration
Fig. 3. Mean values of the discrimination ratio of all groups in the novel object re- time [F(1, 43) = 13.395, p < 0.01; NMS/SH: 23.544 (1.183) vs. MS/
cognition. Preference for the novel object did not differ among groups as a function of SH: 17.68 (1.08)] and frequency [F(1, 43) = 12.349, p < 0.01; NMS/
maternal separation or environmental enrichment (p > 0.05). SH: 3.30 (1.418) vs. MS/SH: 1.33 (1.154). Housing of maternally se-
parated rats in EE reversed this effect [duration: F(1, 43) = 5.93,
finding indicates that all animals reacted similarly to novelty regardless p < 0.05; MS/SH: 17.68 (1.08) vs. MS/EE: 21.42 (1.080); frequency: F
of experimental conditions. (1, 43) = 4.78, p < 0.05, MS/SH: 1.33 (1.154) vs. MS/EE: 2.50
(1.243)]. As far as the time spent in the remaining (non-target) quad-
rants (NE, NW, SE), there was no difference as a function of MS or EE in
3.5. Morris water maze time spent in the a) NE quadrant [MS: F(1, 43) = 1.24, p > 0.05,
partial η2 = 0.028; EE: F (1, 43) = 0.012, p > 0.05, partial η2 = 0.00;
Spatial learning: Spatial learning was significantly affected by EE, NMS/SH = 11.15 (1.21), NMS/EE = 12.05 (1.18), MS/SH = 13.53
with EE treated animals being faster to locate the platform compared to (1.36), MS/EE = 12.37 (1.02)], b) NW quadrant [MS: F(1, 43) = 0.057,
the SH condition [F(1, 43) = 62.47, p < 0.001, partial η2 = 0.592] p > 0.05, partial η2 = 0.001; EE: F(1, 43) = 0.417, p > 0.05, partial
(Fig. 4). The rearing condition exerted no influence on latency [MS: F(1, η2 = 0.01; NMS/SH = 11.58 (0.65), NMS/EE = 13.41 (1.32), MS/
43) = 0.848, p > 0.05, partial η2 = 0.019] neither interacted with EE SH = 14.57 (1.62), MS/EE = 11.04 (1.26)] and c) SE quadrant [MS: F
[MS × EE: F(1, 43) = 1.419, p > 0.05, partial η2 = 0.032]. The per- (1, 43) = 0.819, p > 0.05, partial η2 = 0.019; EE: F (1.43) = 0.006,
formance of rats was improved over the 4-day period [day: F(3, p > 0.05, partial η2 = 0.00; NMS/SH: 12.34 (1.07), NMS/EE: 12.28
129) = 174.309, p < 0.001, partial η2 = 0.802]. Bonferroni’s pairwise (1.51), MS/SH: 13.41 (1.32), MS/EE: 13.68 (1.38)].
comparisons revealed a gradual decrease in latency from the first to the
third day (p < 0.001), but no significant difference was found between 3.6. Plasma corticosterone analysis
the third and the fourth day (p > 0.05). The “day” factor interacted
with housing [day × EE: F(3, 129) = 6.42, p < 0.001, partial Basal levels: There were no significant effects for the basal corti-
η2 = 0.13]. costerone levels by either MS [F (1, 38) = 1.04, p > 0.05, partial
In order to ensure that differences in latency to locate the platform η2 = 0.027] or EE manipulations [F(1, 38) = 0.028, p > 0.005, partial
were not due to differences in swim speed, a two-way ANOVA η2 = 0.001], and the two factors did not interact with each other [F(1,
(MS × EE) was conducted on mean velocity/day. No statistically sig- 38) = 0.000, p > 0.05, partial η2 = 0.000; NMS/SH: 25.58 (6.695),
nificant difference was found as a result of MS [day 1: F(1, 43) = 3.72, NMS/EE: 24.59 (5.316), MS/SH: 19.73 (5.484), MS/EE: 18.11 (4.854)].
p > 0.05, partial η2 = 0.08, day 2: F(1, 43) = 4.01, p > 0.05, partial Stress-induced levels: Analysis revealed a significant effect of
η2 = 0.085), day 3: F(1, 43) = 3.17, p > 0.05, partial η2 = 0.07, day 4: housing condition on corticosterone elevations in response to acute
F(1, 43) = 0.08, p > 0.05, partial η2 = 0.002] or EE [day 1: F(1, stress [F(1, 41) = 6.168, p < 0.017, partial η2 = 0.131], with the SH
43) = 1.86, p > 0.05, partial η2 = 0.041, day 2: F(1, 43) = 0.44, rats expressing significantly higher levels than the animals of the EE
manipulation (Fig. 6). The effect of EE was a function of MS as indicated
by their significant interaction [MS × EE: F(1, 41) = 7.920, p < 0.01,
partial η2 = 0.162]. Pairwise comparisons revealed that maternal se-
paration caused significant elevations in animals of the SH condition
compared to NMS rats [F(1, 41) = 4.887, p < 0.05; NMS/SH: 261
(20.102) vs. MS/SH: 321.167 (18.35)]. On the contrary, exposure to EE
blocked this effect [F(1, 41) = 13.788, p < 0.01, partial η2 = 0.252;
MS/SH: 321.167 (18.35) vs. MS/EE: 220.100 (20.102)].

3.7. Immunoreactivity of BDNF and synaptophysin in the hippocampus

BDNF: Analysis of variance revealed a significant main effect of MS


on the expression of hippocampal BDNF [F (1, 19) = 9.134, p < 0.001,
partial η2 = 0.325], with MS animals expressing lower levels compared
to NMS (Fig. 7). In addition, immunoreactivity was significantly higher
Fig. 4. Latency to find the platform of all groups in the spatial learning protocol of the in animals that were housed in enriched environment, as indicated by
Morris water maze. All animals improved their performance over the 4-day period the main effect of the EE condition [F(1, 19) = 27.068, p < 0.001,
(#
p < 0.05). EE treated animals exhibited lower latencies compared to SH treated ani- partial η2 = 0.588]. In NMS condition, BDNF expression was sig-
mals (*p < 0.001).
nificantly higher in rats that were exposed to enriched conditions

24
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

Fig. 5. (A) Time spent in the target quadrant and (B) frequency of entries in the platform area during the probe trial of the Morris water maze. MS rats housed in standard conditions spent
less time and did fewer entries, compared to NMS/SH rats (*p < 0.01), while exposure to EE increased the time and the number of entries for MS rats (MS/SH vs MS/EE, #p < 0.05).

form of maternal separation (MS). In particular, we investigated the


effects of these environmental manipulations as well as their interaction
on cognition, emotionality, stress reactivity and expression of BDNF
and synaptophysin in the hippocampus.

4.1. Effect of maternal separation and environmental enrichment on anxiety


and stress reactivity

Emotionality was evaluated by the EPM task, a reliable behavioral


test of anxiety. Analysis of four behavioral indices (open-arms time and
entries, closed-arms returns and SAPs) showed that daily MS during the
first three postnatal weeks increased anxiety-like behavior during
adulthood. Specifically, our data suggest that MS potentiated the
Fig. 6. MS animals housed in SH had higher plasma corticosterone levels after exposure standardly housed (SH) rodent’s unconditioned fear towards open
to acute stress, compared to NMS animals (NMS/SH vs MS/SH,*p < 0.05). Rearing of spaces and increased closed arm returns as well as SAPs, a behavioral
maternally-separated animals in EE significantly reduced these corticosterone elevations
profile indicative of increased anxiety. The reduced open-arms pre-
(MS/SH vs MS/EE, #p < 0.05).
ference seems not to be associated with a general decrease in activity,
as indicated by the comparable ambulation scores and arm entries in
compared to standard housing [F(1, 19) = 7.470, p < 0.05, NMS/SH: our Open Field and EPM task, respectively, among the MS and NMS
21.64 (1.91) vs NMS/EE: 28.72 (1.74), partial η2 = 0.282]. Pairwise groups. Specifically, there was no significant difference between NMS
comparisons revealed that maternal separation significantly reduced and MS groups with respect to the horizontal (ambulation) or vertical
the BDNF levels in SH animals [F(1, 19) = 9.545, p < 0.001, partial (rearing) activity, a finding also reported previously (Markostamou
η2 = 0.334, NMS/SH: 21.64 (1.91) vs. MS/SH: 13.9 (1.62)]. Environ- et al., 2016; Shalev and Kafkafi, 2002; Vivinetto et al., 2013).
mental enrichment of maternally separated rats counteracted this MS- Interestingly, post-weaning housing of maternally separated rats in
associated decrease [F(1, 19) = 21.696, p < 0.001, partial η2 = 0.533, EE conditions acted beneficially, decreasing significantly the risk as-
MS/SH: 13.9 (1.62) vs MS/EE: 25.57 (1.91)]. sessment behaviors (i.e., stretch attend posture, closed arms returns)
Synaptophysin (SYN): Both MS and EE conditions significantly af- while increased open arms entries and time. These behavioral effects of
fected the SYN immunoreactivity in the hippocampus [MS: F(1, MS and EE conditions appear to be mediated by alterations in the HPA
19) = 14.172, p < 0.01, partial η2 = 0.427; EE: F(1, 19) = 25.848, axis reactivity. Indeed, maternally separated rats that were housed in
p < 0.001, partial η2 = 0.57], with MS decreasing and EE increasing standard post-weaning conditions (MS/SH group) expressed sig-
its levels (Fig. 8). Additionally, environmental enrichment significantly nificantly higher levels of corticosterone in response to a stress chal-
increased SYN expression in the non-maternally separated rats [F(1, lenge as adults. On the contrary, housing in EE condition (MS/EE
19) = 12.769, p < 0.01, NMS/SH: 27.59 (2.21) vs NMS/EE: 38.31 group) counterbalanced the MS effects, thus attenuating adrenocortical
(2.02), partial η2 = 0.402]. In SH rats, maternal separation significantly responses to levels similar to those of the NMS group. It should be also
decreased SYN expression [F(1, 19) = 7.125, p < 0.05, partial reported that basal corticosterone concentrations did not differ sig-
η2 = 0.273; NMS/SH: 27.59 (2.85), MS/SH: 19.84 (1.74)]. Housing of nificantly as a function of rearing or housing.
maternally-separated rats in enriched conditions reversed this effect [F Our findings are in accordance with previous studies reporting the
(1, 19) = 13.093, p < 0.01, partial η2 = 0.408; MS/SH: 19.84 (1.74) detrimental effects of early adversity on emotionality and stress re-
vs. MS/EE: 30.34 (2.80)]. activity. In fact, it has been shown that MS over the first 2–3 postnatal
weeks significantly increases anxiety and depression-like behaviors and
4. Discussion enhances HPA responses to stressors during adulthood (Aisa et al.,
2007; Kalinichev et al., 2002; Koe et al., 2016; Liu et al., 2000;
It is well known that environmental conditions can exert a strong Veenema et al., 2007). In our experiment, the potentiated corticos-
influence on brain development, behavior and neuroplasticity. In par- terone increase of MS/SH rats in response to an acute stressor (i.e., cage
ticular, early adverse experiences may lead to psychopathology and rotation) was not associated with elevated basal corticosterone in MS/
cognitive impairments that can persist until adulthood, while environ- SH compared to NMS/SH groups. This finding is in accordance with
mental enrichment (EE) appears to have a beneficial role on different studies reporting no differences in basal concentrations in animals
aspects of brain function and behavior. Τhe aim of the present study previously exposed to early-life stress (Knuth and Etgen, 2007; Koe
was to explore whether Environmental Enrichment (EE) during ado- et al., 2016; Rüedi-Bettschen et al., 2005; Slotten et al., 2006; Wigger
lescence can counterbalance the negative impact of early stress in the and Neumann, 1999). Given the significant role of the hippocampus,

25
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

Fig. 7. (A) BDNF expression in the dorsal hippocampus expressed as integrated density. MS animals raised in SH had lower BDNF levels compared to NMS animals of the same rearing
condition (NMS/SH vs MS/SH, *p < 0.001). Environmental enrichment significantly increased protein expression in standard housed rats (NMS/SH vs NMS/EE, #p < 0.05) and
counteracted the decreased levels in maternally-separated rats (MS/SH vs MS/EE, ##p < 0.001). (B) Representative photomicrographs of BDNF immunofluorescence staining in the
hippocampal CA1 region. Total magnification 400x, scale bar = 100 μm.

particularly its glucocorticoid receptor (GR) system, in the HPA nega- effects of MS. Specifically, both recognition memory deficits and de-
tive feedback mechanism, it may be suggested that hippocampal al- pressive-like behavior are completely reversed after administration of a
terations may be associated with the potentiated stress response. In fact, GR antagonist (Aisa et al., 2008, 2007), while treatment with the cor-
decreases in the GR density and mRNA expression have been reported ticosterone synthesis inhibitor, metyrapone, reduces the increased
following early stress (Aisa et al., 2008; Enthoven et al., 2008b). This vulnerability to kindling epileptogenesis seen in maternally separated
downregulation appears to dysregulate the negative feedback me- rats (Koe et al., 2016).
chanism, thus leading to further elevations of plasma corticosterone Contrary to early stress, EE appears to have beneficial behavioral
and increased anxiety (Sampedro-Piquero et al., 2014). Existing data and neurobiological effects. A body of evidence suggests that EE
suggest that glucocorticoids elevations play an important role in the housing attenuates anxiety and HPA-mediated endocrine responses

26
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

Fig. 8. (A) Synaptophysin (SYN) expression in the dorsal hippocampus expressed as integrated density. Environmental enrichment significantly increased protein expression in standard
housed rats (NMS/SH vs NMS/EE, #p < 0.01). Maternal separation significantly decreased SYN expression in rats of the SH condition (NMS/SH vs MS/SH, *p < 0.05). Enriched
environment reversed the decreased SYN levels in maternally separated rats (MS/SH vs MS/EE, #p < 0.01). Representative photomicrographs of SYN immunofluorescence staining in
the hippocampal CA1 region. Total magnification 400x, scale bar = 100 μm.

evoked by stressors in adulthood (Chapillon et al., 1999; Fox et al., compensatory role of EE mainly refer to chronic stress paradigms ad-
2006), while it increases GR expression in hippocampal areas (Vivinetto ministered during adulthood. Specifically, EE housing prior to or fol-
et al., 2013). The regulatory impact of EE is also supported by studies of lowing periods of adult chronic stress reduces emotionality and HPA
reduced anxiety in animal models of anxiety, depression or mental activity to an acute stressor (Wright and Conrad, 2008). Recently, it has
disorders (e.g., schizophrenia and ADHD) (Brenes Sáenz et al., 2006; been shown that a short episode of EE during adulthood reduces an-
Ishihama et al., 2010; Nicolas et al., 2015) as well as in gestational xiety-like behavior in MS rats (Koe et al., 2016). The increases in GR
inflammation-induced HPA axis hyperactivity in young rats (Connors expression seen in EE-housed rats that were subsequently exposed to
et al., 2014). The majority of existing data highlighting the both chronic and acute stress as adults appear to contribute to a more

27
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

adaptive response to chronic stress (Zanca et al., 2015). To the best of post-weaning EE on NORT performance. Although EE housing of 3-
our knowledge, so far only one study has investigated the role of EE on month old adult rats for 3 weeks (PND90-111) improves novel object
enhanced HPA reactivity in maternally-separated rats, reporting a total recognition (Bechara and Kelly, 2013), EE exposure for a longer period
reversal of corticosterone to normal levels under conditions of EE of time during an earlier developmental stage (PND21-60) does not
(Francis et al., 2002). seem to affect it (Vivinetto et al., 2013). These findings probably sug-
gest that the effectiveness of EE on recognition memory may be a
4.2. Effect of maternal separation and environmental enrichment on function of time of exposure and duration of exposure.
cognitive function
4.3. Effects of maternal separation and environmental enrichment on BDNF
Behavioral testing in the Morris Water Maze (MWM) revealed a and synaptophysin expression in the hippocampus
beneficial effect of EE on visuospatial learning during adulthood. The
groups that were housed in post-weaning EE conditions exhibited Increasing evidence underlines the essential role of hippocampal
shorter latencies, compared to the SH rats, to locate the platform, an BDNF and synaptophysin (SYN) in cognition as well as anxiety and
indication of improved spatial acquisition. Our findings are in line with depression-related behaviors (Heldt et al., 2007; Liu et al., 2005;
previous studies demonstrating the beneficial effects of environmental Mitchelmore and Gede, 2014). In the present study, MS rats expressed
enrichment on cognitive function. In fact, EE has been associated with behaviors indicative of impaired memory and increased anxiety, while
improvement in spatial learning, memory, and orientation in adult and EE exerted a beneficial effect in the MS group. In order to estimate
aged rodents (Frick and Fernandez, 2003; Harburger et al., 2007; whether these behavioral changes were mediated by alterations in
Hullinger et al., 2015; Leggio et al., 2005) as well as protection against neuronal plasticity, we evaluated the expression of these two markers
age-associated impairment in LTP induction (Stein et al., 2016). (BDNF and SYN) in the dorsal hippocampus. BDNF plays an essential
MS did not impair spatial acquisition, a finding also reported by role in synapse formation, neuronal survival and growth and it has been
other studies following postnatal stress (Aisa et al., 2009; Akatsu et al., suggested to be involved in transducing the effects of environmental
2015; Grace et al., 2009). However, it did impair spatial memory, as manipulation on brain function during early developmental stages
indicated by our probe trial data. These long term negative con- (Cirulli et al., 2003). In addition, BDNF expression in multiple brain
sequences in memory (spatial or fear-associated) are in line with pre- regions is sensitive to adverse life experiences (Han et al., 2011; Meng
vious reports demonstrating spatial and fear-associated memory im- et al., 2011).
pairments following maternal separation (Chocyk et al., 2014; Sousa According to our analysis, MS significantly decreased the expression
et al., 2014; Xiong et al., 2015). This finding may imply that spatial of BDNF in the dorsal hippocampus. This finding is in line with previous
memory is more vulnerable than visuospatial learning to the effect of studies showing a downregulation in BDNF mRNA or protein expression
early adversity, as it has been also suggested regarding the effects of following 2–3 weeks of MS (Aisa et al., 2009; Lippmann et al., 2007;
chronic stress (Conrad, 2010). In fact, it has been reported that the MacQueen et al., 2003). However, the investigation of the MS impact
spatial learning deficits seen in juvenile rats following postnatal social on BDNF expression has yielded inconsistent results due to the in-
isolation are restored in adulthood, an over-time improvement possibly volvement of various factors. The duration of adverse postnatal ma-
associated with compensatory changes that take place (i.e., damaged nipulation appears to be a determining factor, since postnatal stress of
dendrites are repaired, new ones are sprouting) within the hippo- shorter duration did not affect BDNF levels (i.e., a single episode of 24 h
campus (Frisone et al., 2002). maternal deprivation, or 6-day MS) (Choy et al., 2008; Markostamou
Interestingly, post weaning EE offered total protection against the et al., 2016; Roceri et al., 2002). Furthermore, the developmental
spatial memory deficits associated with MS. While MS rats spent less period of the animals as well as the brain region seem to influence MS
time in the target quadrant and made fewer frequencies of entries into effects. In fact, it has been shown that MS induces different patterns (up
the platform area, compared to NMS group, EE completely reversed this or down-regulation) of BDNF expression along with age in different
effect. As far as the impact of EE on spatial memory of MS rats, previous brain regions (Pardon et al., 2009; Récamier-Carballo et al., 2017;
findings also support its compensatory role against cognitive deficits Wang et al., 2015b). This differential impact of MS on BDNF expression
following chronic adult (Hutchinson et al., 2012; Veena et al., 2009; reflects the complex functional consequences of adverse early life ex-
Wright and Conrad, 2008), juvenile (Ilin and Richter-Levin, 2009) or periences in synaptic plasticity mechanisms.
early stress induced by MS or limited nesting/bedding material (Cui In addition to the BDNF changes, we found that prolonged MS
et al., 2006; do Prado et al., 2016; Vivinetto et al., 2013). However, the significantly decreased SYN in the dorsal hippocampus. SYN, a synaptic
present study is the first to demonstrate the ameliorating role of EE vesicle-associated protein, is a general synaptic marker and its presence
against spatial memory impairments in rodents that experienced early indicates the efficiency of synaptic transmission (Thiel, 1993; Valtorta
stress in the form of maternal separation. et al., 2004). It has been previously reported that MS can cause down-
Behavioral analysis of the NORT data revealed that neither the type regulation of SYN during adulthood, and this effect might be re-
of rearing nor housing manipulations affected non-spatial recognition sponsible for impaired hippocampal neurotransmission and behavioral
memory. While a number of studies showed impairments in recognition changes (Aisa et al., 2009; Andersen and Teicher, 2004).
memory following early adversity (Aisa et al., 2008; Daniels et al., The possibility that BDNF and SYN expression may have been af-
2009; Hulshof et al., 2011), other investigators report no effect of fected by the acute stress (i.e., rotation) applied at the end of the ex-
postnatal stress on this type of memory (Grace et al., 2009; Mourlon periments cannot be excluded. Previous studies have shown that acute
et al., 2010; Vivinetto et al., 2013). It is possible that differences in stress alters the expression of neurotrophic factors and synaptic reg-
duration and type of postnatal stress as well as time of testing may ulatory proteins and these alterations may be responsible for some of
account for this discrepancy. It should be mentioned that the absence of the morphological and behavioral changes observed after stress ex-
deficits in the non-spatial recognition memory reported here is in line posure (Amin et al., 2015; Gao et al., 2006; Smith et al., 1995; Thome
with previous research conducted in our lab (Tata et al., 2015). The fact et al., 2001). Specifically, acute stress induces a brain region-dependent
that MS rats were impaired in the probe trial of the MWM, but not in differential expression of BDNF (Lakshminarasimhan and Chattarji,
the NOR task, implies that our MS paradigm selectively affects spatial 2012). Furthermore, the type of stressor, the duration of stress or the
memory. In fact, Leret and colleagues have also shown impairments in age of the animals are factors that also affect the outcome (Badowska-
spatial memory but not object recognition both in adolescent and adult Szalewska et al., 2017; Fuchikami et al., 2009; Molteni et al., 2009).
rats that had been exposed to MS (Leret et al., 2010). Regarding the role However, the present study did not intend to explore the possible effect
of housing, so far there is limited information concerning the effect of of acute stress on markers of synaptic plasticity or its functional

28
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

consequences. Furthermore, it should be mentioned that all animals Medical Athens S.A) for his excellent technical assistance in corticos-
were subjected to the acute stress condition, which implies that all they terone measurements (ELISA).
would have been equally affected.
Contrary to the effects of MS, exposure to EE increased the levels of References
both SYN and BDNF expression in the non-stressed group. Previous
studies have also supported the beneficial effects of EE as indicated by Aisa, B., Tordera, R., Lasheras, B., Del Río, J., Ramírez, M.J., 2007. Cognitive impairment
the enhanced BDNF both at mRNA and protein levels (Cao et al., 2014; associated to HPA axis hyperactivity after maternal separation in rats.
Psychoneuroendocrinology 32, 256–266. http://dx.doi.org/10.1016/j.psyneuen.
Griva et al., 2017; Ickes et al., 2000; Mosaferi et al., 2015). In addition, 2006.12.013.
our analysis showed that EE upregulated SYN in control animals, an Aisa, B., Tordera, R., Lasheras, B., Del Río, J., Ramírez, M.J., 2008. Effects of maternal
increase which is in agreement with previous studies (Birch et al., 2013; separation on hypothalamic-pituitary-adrenal responses, cognition and vulnerability
to stress in adult female rats. Neuroscience 154, 1218–1226. http://dx.doi.org/10.
Lambert et al., 2005; Nithianantharajah et al., 2004) and indicates the 1016/j.neuroscience.2008.05.011.
involvement of this synaptic protein in experience-dependent neuro- Aisa, B., Elizalde, N., Tordera, R., Lasheras, B., Del Río, J., Ramírez, M.J., 2009. Effects of
plasticity. neonatal stress on markers of synaptic plasticity in the hippocampus: implications for
spatial memory. Hippocampus 19, 1222–1231. http://dx.doi.org/10.1002/hipo.
In our study, the beneficial role of EE is mainly supported by its
20586.
compensatory effect against the MS-associated BDNF and SYN de- Akatsu, S., Ishikawa, C., Takemura, K., Ohtani, A., Shiga, T., 2015. Effects of prenatal
creases. Specifically, the levels in the stressed rats exposed to EE were stress and neonatal handling on anxiety, spatial learning and serotonergic system of
male offspring mice. Neurosci. Res. http://dx.doi.org/10.1016/j.neures.2015.07.002.
significantly higher than those in the MS animals housed in standard
Amin, S.N., El-Aidi, A.A., Ali, M.M., Attia, Y.M., Rashed, L.A., 2015. Modification of
conditions, and did not differ from those in the non-stressed groups. hippocampal markers of synaptic plasticity by memantine in animal models of acute
Existing evidence supports the compensatory effect of EE on brain da- and repeated restraint stress: implications for memory and behavior. Neuromolecular
mage in models of chronic stress or aging. In particular, EE protects Med. 17, 121–136. http://dx.doi.org/10.1007/s12017-015-8343-0.
Anda, R.F., Dong, M., Brown, D.W., Felitti, V.J., Giles, W.H., Perry, G.S., Valerie, E.J.,
from dendritic atrophy as well as reductions in neurogenesis and neu- Dube, S.R., 2009. The relationship of adverse childhood experiences to a history of
rotrophin expression following adult stress (Hutchinson et al., 2012; premature death of family members. BMC Public Health 9, 106. http://dx.doi.org/
Shilpa et al., 2017; Veena et al., 2009; Vega-Rivera et al., 2016). Pre- 10.1186/1471-2458-9-106.
Andersen, S.L., Teicher, M.H., 2004. Delayed effects of early stress on hippocampal de-
vious studies have also shown that exposure to EE during a critical velopment. Neuropsychopharmacology 29, 1988–1993. http://dx.doi.org/10.1038/
developmental period (0–2 postnatal months), is more effective at in- sj.npp.1300528.
creasing BDNF levels (Jha et al., 2016). In this respect, our BDNF in- Badowska-Szalewska, E., Ludkiewicz, B., Krawczyk, R., Melka, N., Moryś, J., 2017.
Comparison of the influence of two models of mild stress on hippocampal brain-
creases are in line with existing evidence, indicating a beneficial role of derived neurotrophin factor (BDNF) immunoreactivity in old age rats. Acta
post-weaning EE (PND23-65) against MS-associated decreases in BDNF Neurobiol. Exp. (Wars) 77, 68–76.
expression. Given that EE completely restored spatial memory impair- Baroncelli, L., Braschi, C., Spolidoro, M., Begenisic, T., Sale, a, Maffei, L., 2010. Nurturing
brain plasticity: impact of environmental enrichment. Cell Death Differ. 17,
ments in MS group and improved spatial acquisition, it could be sug-
1092–1103. http://dx.doi.org/10.1038/cdd.2009.193.
gested that EE-associated increases in SYN and BDNF may be a pre- Bechara, R.G., Kelly, Á.M., 2013. Exercise improves object recognition memory and in-
requisite for these behavioral effects. In fact, higher levels of BDNF and duces BDNF expression and cell proliferation in cognitively enriched rats. Behav.
Brain Res. 245, 96–100. http://dx.doi.org/10.1016/j.bbr.2013.02.018.
SYN have been correlated with cognitive improvement (Chen et al.,
Bennett, J.C., McRae, P.a., Levy, L.J., Frick, K.M., 2006. Long-term continuous, but not
2013; Frick and Fernandez, 2003), while according to others may not daily, environmental enrichment reduces spatial memory decline in aged male mice.
be a prerequisite (Bennett et al., 2006; Griva et al., 2017; Pereira et al., Neurobiol. Learn. Mem. 85, 139–152. http://dx.doi.org/10.1016/j.nlm.2005.09.003.
2009). Bevins, R.a., Besheer, J., 2006. Object recognition in rats and mice: a one-trial non-
matching-to-sample learning task to study recognition memory. Nat. Protoc. 1,
1306–1311. http://dx.doi.org/10.1038/nprot.2006.205.
5. Conclusion Birch, A.M., Mcgarry, N.B., Kelly, Á.M., 2013. Short-term environmental enrichment, in
the absence of exercise, improves memory, and increases NGF concentration, early
neuronal survival, and synaptogenesis in the dentate gyrus in a time-dependent
In the current study we explored the interaction of a 3-week MS and manner. Hippocampus 23, 437–450. http://dx.doi.org/10.1002/hipo.22103.
subsequent exposure to enriched conditions on various behaviors, Bock, J., Braun, K., 2011. The impact of perinatal stress on the functional maturation of
neuroendocrine stress response and two plasticity markers, BDNF and prefronto-cortical synaptic circuits: implications for the pathophysiology of ADHD?
Prog. Brain Res. 189, 155–169. http://dx.doi.org/10.1016/B978-0-444-53884-0.
synaptophysin. To date, existing studies suggest the negative impact of 00023-3.
MS or, on the contrary, the beneficial role of EE on emotional and Brenes Sáenz, J.C., Villagra, O.R., Fornaguera Trías, J., 2006. Factor analysis of Forced
cognitive behaviors, stress response and brain anatomy and neu- Swimming test, Sucrose Preference test and Open Field test on enriched, social and
isolated reared rats. Behav. Brain Res. 169, 57–65. http://dx.doi.org/10.1016/j.bbr.
rochemistry. Besides the main effects of the two environmental condi- 2005.12.001.
tions on learning and memory, corticosterone levels and BDNF and SYN Bubser, M., Keseberg, U., Notz, P.K., Schmidt, W.J., 1992. Differential behavioural and
expression, here we report that EE can compensate against stress-as- neurochemical effects of competitive and non-competitive NMDA receptor antago-
nists in rats. Eur. J. Pharmacol. 229, 75–82. http://dx.doi.org/10.1016/0014-
sociated spatial memory deficits, increased anxiety and downregulation
2999(92)90288-F.
of BDNF and SYN. To the best of our knowledge, this is the first study to Cao, W., Duan, J., Wang, X., Zhong, X., Hu, Z., Huang, F., Wang, H., Zhang, J., Li, F.,
show that MS and EE interact with each other, modulating adult’s be- Zhang, J., Luo, X., Li, C.Q., 2014. Early enriched environment induces an increased
havior and neuroendocrine response to stress as well as neuroplasticity. conversion of proBDNF to BDNF in the adult rat’s hippocampus. Behav. Brain Res.
265, 76–83. http://dx.doi.org/10.1016/j.bbr.2014.02.022.
Given the important role of early experiences, the need to better un- Chapillon, P., Manneché, C., Belzung, C., Caston, J., 1999. Rearing environmental en-
derstand the underlying mechanisms that mediate behavioral changes richment in two inbred strains of mice: 1. Effects on emotional reactivity. Behav.
is further emphasized. Genet. 29, 41–46. http://dx.doi.org/10.1023/A:1021437905913.
Chen, A., Xiong, L.-J., Tong, Y., Mao, M., 2013. The neuroprotective roles of BDNF in
hypoxic ischemic brain injury. Biomed. Rep. 1, 167–176. http://dx.doi.org/10.3892/
Funding br.2012.48.
Chocyk, A., Przyborowska, A., Makuch, W., Majcher-Maślanka, I., Dudys, D., Wędzony,
K., 2014. The effects of early-life adversity on fear memories in adolescent rats and
This research has been financially supported by the General their persistence into adulthood. Behav. Brain Res. 264, 161–172. http://dx.doi.org/
Secretariat for Research and Technology (GSRT) and the Hellenic 10.1016/j.bbr.2014.01.040.
Foundation for Research and Innovation (HFRI) (Scholarship Code: Choy, K.H.C., de Visser, Y., Nichols, N.R., van den Buuse, M., 2008. Combined neonatal
stress and young-adult glucocorticoid stimulation in rats reduce BDNF expression in
95144 to E.D). hippocampus: effects on learning and memory. Hippocampus 18, 655–667. http://dx.
doi.org/10.1002/hipo.20425.
Acknowledgements Cirulli, F., Berry, A., Alleva, E., 2003. Early disruption of the mother–infant relationship:
effects on brain plasticity and implications for psychopathology. Neurosci. Biobehav.
Rev. 27, 73–82. http://dx.doi.org/10.1016/S0149-7634(03)00010-1.
The authors are grateful to Alexandros Antonellos (Microanalysi

29
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

Connors, E.J., Shaik, a. N., Migliore, M.M., Kentner, a. C., 2014. Environmental enrich- potential and production of IL-1β, TNFα, and IL-10 by peritoneal macrophages under
ment mitigates the sex-specific effects of gestational inflammation on social en- stress conditions. Bull. Exp. Biol. Med. 161, 339–343. http://dx.doi.org/10.1007/
gagement and the hypothalamic pituitary adrenal axis-feedback system. Brain Behav. s10517-016-3409-z.
Immun. 42, 178–190. http://dx.doi.org/10.1016/j.bbi.2014.06.020. Gelfo, F., Cutuli, D., Foti, F., Laricchiuta, D., De Bartolo, P., Caltagirone, C., Petrosini, L.,
Conrad, C.D., 2010. A critical review of chronic stress effects on spatial learning and Angelucci, F., 2011. Enriched environment improves motor function and increases
memory. Prog. Neuropsychopharmacol. Biol. Psychiatry 34, 742–755. http://dx.doi. neurotrophins in hemicerebellar lesioned rats. Neurorehabil. Neural Rep. 25,
org/10.1016/j.pnpbp.2009.11.003. 243–252. http://dx.doi.org/10.1177/1545968310380926.
Cortese, G.P., Olin, A., O’Riordan, K., Hullinger, R., Burger, C., 2018. Environmental Grace, L., Hescham, S., Kellaway, L.a., Bugarith, K., Russell, V.a., 2009. Effect of exercise
enrichment improves hippocampal function in aged rats by enhancing learning and on learning and memory in a rat model of developmental stress. Metab. Brain Dis. 24,
memory, LTP, and mGluR5-Homer1c activity. Neurobiol. Aging 63, 1–11. http://dx. 643–657. http://dx.doi.org/10.1007/s11011-009-9162-5.
doi.org/10.1016/J.NEUROBIOLAGING.2017.11.004. Griva, M., Lagoudaki, R., Touloumi, O., Nousiopoulou, E., Karalis, F., Georgiou, T.,
Crofton, E.J., Zhang, Y., Green, T.A., 2015. Inoculation stress hypothesis of environmental Kokaraki, G., Simeonidou, C., Tata, D.A., Spandou, E., 2017. Long-term effects of
enrichment. Neurosci. Biobehav. Rev. 49, 19–31. http://dx.doi.org/10.1016/j. enriched environment following neonatal hypoxia-ischemia on behavior, BDNF and
neubiorev.2014.11.017. synaptophysin levels in rat hippocampus: effect of combined treatment with G-CSF.
Cruz, a P., Frei, F., Graeff, F.G., 1994. Ethopharmacological analysis of rat behavior on Brain Res. 1667, 55–67. http://dx.doi.org/10.1016/j.brainres.2017.05.004.
the elevated plus-maze. Pharmacol. Biochem. Behav. 49, 171–176. http://dx.doi.org/ Han, X., Wang, W., Xue, X., Shao, F., Li, N., 2011. Brief social isolation in early adoles-
10.1016/0091-3057(94)90472-3. cence affects reversal learning and forebrain BDNF expression in adult rats. Brain Res.
Cui, M., Yang, Y., Yang, J., Zhang, J., Han, H., Ma, W., Li, H., Mao, R., Xu, L., Hao, W., Bull. 86, 173–178. http://dx.doi.org/10.1016/J.BRAINRESBULL.2011.07.008.
Cao, J., 2006. Enriched environment experience overcomes the memory deficits and Handley, S.L., Mithani, S., 1984. Archives of phamnacology effects of alpha-adrenoceptor
depressive-like behavior induced by early life stress. Neurosci. Lett. 404, 208–212. agonists and antagonists in a maze-exploration model of ’ fear ’–motivated
http://dx.doi.org/10.1016/j.neulet.2006.05.048. behaviour. pp. 1–5.
D’Hooge, R., De Deyn, P.P., 2001. Applications of the Morris water maze in the study of Harburger, L.L., Lambert, T.J., Frick, K.M., 2007. Age-dependent effects of environmental
learning and memory. Brain Res. Rev. http://dx.doi.org/10.1016/S0165-0173(01) enrichment on spatial reference memory in male mice. Behav. Brain Res. 185, 43–48.
00067-4. http://dx.doi.org/10.1016/j.bbr.2007.07.009.
Daniels, W.M.U., Fairbairn, L.R., van Tilburg, G., McEvoy, C.R.E., Zigmond, M.J., Russell, Heim, C., Binder, E.B., 2012. Current research trends in early life stress and depression:
V.a., Stein, D.J., 2009. Maternal separation alters nerve growth factor and corticos- review of human studies on sensitive periods, gene-environment interactions, and
terone levels but not the DNA methylation status of the exon 1(7) glucocorticoid epigenetics. Exp. Neurol. 233, 102–111. http://dx.doi.org/10.1016/j.expneurol.
receptor promoter region. Metab. Brain Dis. 24, 615–627. http://dx.doi.org/10. 2011.10.032.
1007/s11011-009-9163-4. Heim, C., Nemeroff, C.B., 2001. The role of childhood trauma in the neurobiology of
do Prado, C.H., Narahari, T., Holland, F.H., Lee, H.-N., Murthy, S.K., Brenhouse, H.C., mood and anxiety disorders: preclinical and clinical studies. Biol. Psychiatry 49,
2016. Effects of early adolescent environmental enrichment on cognitive dysfunction, 1023–1039.
prefrontal cortex development, and inflammatory cytokines after early life stress. Heldt, S.A., Stanek, L., Chhatwal, J.P., Ressler, K.J., 2007. Hippocampus-specific deletion
Dev. Psychobiol. 58, 482–491. http://dx.doi.org/10.1002/dev.21390. of BDNF in adult mice impairs spatial memory and extinction of aversive memories.
Doremus, T.L., Varlinskaya, E.I., Spear, L.P., 2006. Factor analysis of elevated plus-maze Mol. Psychiatry 12, 656–670. http://dx.doi.org/10.1038/sj.mp.4001957.
behavior in adolescent and adult rats. Pharmacol. Biochem. Behav. 83, 570–577. Hralová, M., Angerová, Y., Gueye, T., Bortelová, J., Švestková, O., Zima, T., Lippertová-
http://dx.doi.org/10.1016/j.pbb.2006.03.019. Grünerová, M., 2013. Long-term results of enriched environment and erythropoietin
Ennaceur, A., Delacour, J., 1988. A new one-trial test for neurobiological studies of after hypobaric hypoxia in rats. Physiol. Res. 62, 463–470 (932354 [pii]).
memory in rats. 1: behavioral data. Behav. Brain Res. 31, 47–59. Hullinger, R., O’Riordan, K., Burger, C., 2015. Environmental enrichment improves
Ennaceur, A., Meliani, K., 1992. A new one-trial test for neurobiological studies of learning and memory and long-term potentiation in young adult rats through a
memory in rats. III. Spatial vs. non-spatial working memory. Behav. Brain Res. 51, mechanism requiring mGluR5 signaling and sustained activation of p70s6k.
83–92. http://dx.doi.org/10.1016/S0166-4328(05)80315-8. Neurobiol. Learn. Mem. 125, 126–134. http://dx.doi.org/10.1016/j.nlm.2015.08.
Ennaceur, A., Neave, N., Aggleton, J.P., 1997. Spontaneous object recognition and object 006.
location memory in rats: the effects of lesions in the cingulate cortices the medial Hulshof, H.J., Novati, A., Sgoifo, A., Luiten, P.G.M., den Boer, J.a., Meerlo, P., 2011.
prefrontal cortex, the cingulum bundle and the fornix. Exp. Brain Res. 113, 509–519. Maternal separation decreases adult hippocampal cell proliferation and impairs
Enthoven, L., de Kloet, E.R., Oitzl, M.S., 2008a. Effects of maternal deprivation of CD1 cognitive performance but has little effect on stress sensitivity and anxiety in adult
mice on performance in the water maze and swim stress. Behav. Brain Res. 187, Wistar rats. Behav. Brain Res. 216, 552–560. http://dx.doi.org/10.1016/j.bbr.2010.
195–199. http://dx.doi.org/10.1016/j.bbr.2007.08.037. 08.038.
Enthoven, L., de Kloet, E.R., Oitzl, M.S., 2008b. Differential development of stress system Huot, R.L., Thrivikraman, K.V., Meaney, M.J., Plotsky, P.M., 2001. Development of adult
(re)activity at weaning dependent on time of disruption of maternal care. Brain Res. ethanol preference and anxiety as a consequence of neonatal maternal separation in
1217, 62–69. http://dx.doi.org/10.1016/j.brainres.2008.04.009. Long Evans rats and reversal with antidepressant treatment. Psychopharmacology
Felitti, V.J., Anda, R.F., Nordenberg, D., Williamson, D.F., Spitz, A.M., Edwards, V., Koss, (Berl.) 158, 366–373. http://dx.doi.org/10.1007/s002130100701.
M.P., Marks, J.S., 1998. Household dysfunction to many of the leading causes of Hutchinson, K.M., McLaughlin, K.J., Wright, R.L., Bryce Ortiz, J., Anouti, D.P., Mika, A.,
death in adults the adverse childhood experiences (ACE) study. Am. J. Prev. Med. 14, Diamond, D.M., Conrad, C.D., 2012. Environmental enrichment protects against the
245–258. effects of chronic stress on cognitive and morphological measures of hippocampal
Fox, C., Merali, Z., Harrison, C., 2006. Therapeutic and protective effect of environmental integrity. Neurobiol. Learn. Mem. 97, 250–260. http://dx.doi.org/10.1016/j.nlm.
enrichment against psychogenic and neurogenic stress. Behav. Brain Res. 175, 1–8. 2012.01.003.
http://dx.doi.org/10.1016/j.bbr.2006.08.016. Ickes, B.R., Pham, T.M., Sanders, L.A., Albeck, D.S., Mohammed, A.H., Granholm, A.-C.,
Francis, D.D., Diorio, J., Plotsky, P.M., Meaney, M.J., 2002. Environmental enrichment 2000. Long-term environmental enrichment leads to regional increases in neuro-
reverses the effects of maternal separation on stress reactivity. J. Neurosci. 22, trophin levels in rat brain. Exp. Neurol. 164, 45–52. http://dx.doi.org/10.1006/exnr.
7840–7843. 2000.7415.
Frick, K.M., Fernandez, S.M., 2003. Enrichment enhances spatial memory and increases Ilin, Y., Richter-Levin, G., 2009. Enriched environment experience overcomes learning
synaptophysin levels in aged female mice. Neurobiol. Aging 24, 615–626. http://dx. deficits and depressive-Like behavior induced by juvenile stress. PLoS One 4, e4329.
doi.org/10.1016/S0197-4580(02)00138-0. http://dx.doi.org/10.1371/journal.pone.0004329.
Friske, J.E., Gammie, S.C., 2005. Environmental enrichment alters plus maze, but not Ishihama, T., Ago, Y., Shintani, N., Hashimoto, H., Baba, A., Takuma, K., Matsuda, T.,
maternal defense performance in mice. Physiol. Behav. 85, 187–194. http://dx.doi. 2010. Environmental factors during early developmental period influence psycho-
org/10.1016/j.physbeh.2005.03.022. behavioral abnormalities in adult PACAP-deficient mice. Behav. Brain Res. 209,
Frisone, D.F., Frye, C.A., Zimmerberg, B., 2002. Social isolation stress during the third 274–280. http://dx.doi.org/10.1016/j.bbr.2010.02.009.
week of life has age-dependent effects on spatial learning in rats. Behav. Brain Res. Jha, S., Dong, B., Sakata, K., 2011. Enriched environment treatment reverses depression-
128, 153–160. like behavior and restores reduced hippocampal neurogenesis and protein levels of
Fuchikami, M., Morinobu, S., Kurata, A., Yamamoto, S., Yamawaki, S., 2009. Single im- brain-derived neurotrophic factor in mice lacking its expression through promoter IV.
mobilization stress differentially alters the expression profile of transcripts of the Transl. Psychiatry 1, e40. http://dx.doi.org/10.1038/tp.2011.33.
brain-derived neurotrophic factor (BDNF) gene and histone acetylation at its pro- Jha, S., Dong, B.E., Xue, Y., Delotterie, D.F., Vail, M.G., Sakata, K., 2016. Antidepressive
moters in the rat hippocampus. Int. J. Neuropsychopharmacol. 12, 73. http://dx.doi. and BDNF effects of enriched environment treatment across ages in mice lacking
org/10.1017/S1461145708008997. BDNF expression through promoter IV. Transl. Psychiatry 6, e896. http://dx.doi.org/
Fumagalli, F., Molteni, R., Racagni, G., Riva, M.A., 2007. Stress during development: 10.1038/tp.2016.160.
impact on neuroplasticity and relevance to psychopathology. Prog. Neurobiol. 81, Kalinichev, M., Easterling, K.W., Plotsky, P.M., Holtzman, S.G., 2002. Long-lasting
197–217. http://dx.doi.org/10.1016/j.pneurobio.2007.01.002. changes in stress-induced corticosterone response and anxiety-like behaviors as a
Gamberini, M.T., Rodrigues, D.S., Rodrigues, D., Pontes, V.B., 2015. Effects of the aqu- consequence of neonatal maternal separation in Long-Evans rats. Pharmacol.
eous extract of Pimpinella anisum L. seeds on exploratory activity and emotional Biochem. Behav. 73, 131–140.
behavior in rats using the open field and elevated plus maze tests. J. Ethnopharmacol. Knuth, E.D., Etgen, A.M., 2007. Long-term behavioral consequences of brief, repeated
1–5. http://dx.doi.org/10.1016/j.jep.2015.03.053. neonatal isolation. Brain Res. 1128, 139–147. http://dx.doi.org/10.1016/j.brainres.
Gao, Y., Bezchlibnyk, Y.B., Sun, X., Wang, J.-F., McEwen, B.S., Young, L.T., 2006. Effects 2006.10.054.
of restraint stress on the expression of proteins involved in synaptic vesicle exocytosis Koe, A.S., Ashokan, A., Mitra, R., 2016. Short environmental enrichment in adulthood
in the hippocampus. Neuroscience 141, 1139–1148. http://dx.doi.org/10.1016/J. reverses anxiety and basolateral amygdala hypertrophy induced by maternal se-
NEUROSCIENCE.2006.04.066. paration. Transl. Psychiatry 6, e729. http://dx.doi.org/10.1038/tp.2015.217.
Gein, S.V., Sharavieva, I.L., 2016. Effect of opiate receptors blockade on microbicidal Korosi, A., Naninck, E.F.G., Oomen, C. a., Schouten, M., Krugers, H., Fitzsimons, C.,

30
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

Lucassen, P.J., 2012. Early-life stress mediated modulation of adult neurogenesis and Nicolas, S., Veyssière, J., Gandin, C., Zsürger, N., Pietri, M., Heurteaux, C., Glaichenhaus,
behavior. Behav. Brain Res. 227, 400–409. http://dx.doi.org/10.1016/j.bbr.2011.07. N., Petit-Paitel, A., Chabry, J., 2015. Neurogenesis-independent antidepressant-like
037. effects of enriched environment is dependent on adiponectin.
Kulesskaya, N., Voikar, V., 2014. Assessment of mouse anxiety-like behavior in the light- Psychoneuroendocrinology 57, 72–83. http://dx.doi.org/10.1016/j.psyneuen.2015.
dark box and open-field arena: role of equipment and procedure. Physiol. Behav. 133, 03.017.
30–38. http://dx.doi.org/10.1016/j.physbeh.2014.05.006. Nithianantharajah, J., Levis, H., Murphy, M., 2004. Environmental enrichment results in
Lajud, N., Roque, A., Cajero, M., Gutiérrez-Ospina, G., Torner, L., 2012. Periodic maternal cortical and subcortical changes in levels of synaptophysin and PSD-95 proteins.
separation decreases hippocampal neurogenesis without affecting basal corticos- Neurobiol. Learn. Mem. 81, 200–210. http://dx.doi.org/10.1016/j.nlm.2004.02.002.
terone during the stress hyporesponsive period, but alters HPA axis and coping be- Nozari, M., Shabani, M., Hadadi, M., Atapour, N., 2014. Enriched environment prevents
havior in adulthood. Psychoneuroendocrinology 37, 410–420. http://dx.doi.org/10. cognitive and motor deficits associated with postnatal MK-801 treatment.
1016/j.psyneuen.2011.07.011. Psychopharmacology (Berl.) 231, 4361–4370. http://dx.doi.org/10.1007/s00213-
Lakshminarasimhan, H., Chattarji, S., 2012. Stress leads to contrasting effects on the le- 014-3580-8.
vels of brain derived neurotrophic factor in the hippocampus and amygdala. PLoS Palagini, L., Drake, C.L., Gehrman, P., Meerlo, P., Riemann, D., 2015. Early-life origin of
One 7, e30481. http://dx.doi.org/10.1371/journal.pone.0030481. adult insomnia: does prenatal–early-life stress play a role? Sleep Med. 16, 446–456.
Lambert, T.J., Fernandez, S.M., Frick, K.M., 2005. Different types of environmental en- http://dx.doi.org/10.1016/j.sleep.2014.10.013.
richment have discrepant effects on spatial memory and synaptophysin levels in fe- Pardon, M.-C., Sarmad, S., Rattray, I., Bates, T.E., Scullion, G.A., Marsden, C.A., Barrett,
male mice. Neurobiol. Learn. Mem. 83, 206–216. http://dx.doi.org/10.1016/j.nlm. D.A., Lowe, J., Kendall, D.A., 2009. Repeated novel cage exposure-induced im-
2004.12.001. provement of early Alzheimer’s-like cognitive and amyloid changes in TASTPM mice
Lau, A.A., Crawley, A.C., Hopwood, J.J., Hemsley, K.M., 2008. Open field locomotor is unrelated to changes in brain endocannabinoids levels. Neurobiol. Aging 30,
activity and anxiety-related behaviors in mucopolysaccharidosis type IIIA mice. 1099–1113. http://dx.doi.org/10.1016/j.neurobiolaging.2007.10.002.
Behav. Brain Res. 191, 130–136. http://dx.doi.org/10.1016/j.bbr.2008.03.024. Pascual, R., Valencia, M., Bustamante, C., 2015. Purkinje cell dendritic atrophy induced
Laviola, G., Hannan, A.J., Macrì, S., Solinas, M., Jaber, M., 2008. Effects of enriched by prenatal stress is mitigated by early environmental enrichment. Neuropediatrics
environment on animal models of neurodegenerative diseases and psychiatric dis- 46, 37–43. http://dx.doi.org/10.1055/s-0034-1395344.
orders. Neurobiol. Dis. 31, 159–168. http://dx.doi.org/10.1016/j.nbd.2008.05.001. Paxinos, G., Watson, C., 2007. The Rat Brain in Stereotaxic Coordinates, 6th ed. Elevier,
Leggio, M.G., Mandolesi, L., Federico, F., Spirito, F., Ricci, B., Gelfo, F., Petrosini, L., Boston.
2005. Environmental enrichment promotes improved spatial abilities and enhanced Pereira, L.O., Nabinger, P.M., Strapasson, A.C.P., Nardin, P., Gonçalves, C.A.S., Siqueira,
dendritic growth in the rat. Behav. Brain Res. 163, 78–90. http://dx.doi.org/10. I.R., Netto, C.A., 2009. Long-term effects of environmental stimulation following
1016/j.bbr.2005.04.009. hypoxia-ischemia on the oxidative state and BDNF levels in rat hippocampus and
Leret, M.L., Pérez-Hernández, M., Sanz, N., Díaz, S., Hernández-Tristán, R., Rua, C., 2010. frontal cortex. Brain Res. 1247, 188–195. http://dx.doi.org/10.1016/j.brainres.2008.
[P2.52] Maternal separation effects in spatial and non-spatial memory in adolescent 10.017.
and adult rats. Int. J. Dev. Neurosci. 28, 704–705. http://dx.doi.org/10.1016/j. Récamier-Carballo, S., Estrada-Camarena, E., López-Rubalcava, C., 2017. Maternal se-
ijdevneu.2010.07.182. paration induces long-term effects on monoamines and brain-derived neurotrophic
Levine, S., 2001. Primary social relationships influence the development of the hy- factor levels on the frontal cortex, amygdala, and hippocampus: differential effects
pothalamic–pituitary–adrenal axis in the rat. Physiol. Behav. 73, 255–260. after a stress challenge. Behav. Pharmacol. 28, 545–557. http://dx.doi.org/10.1097/
Levine, S., 2002. Regulation of the hypothalamic-pituitary-adrenal axis in the neonatal FBP.0000000000000324.
rat: the role of maternal behavior. Neurotox. Res. 4, 557–564. http://dx.doi.org/10. Rüedi-Bettschen, D., Pedersen, E.-M., Feldon, J., Pryce, C.R., 2005. Early deprivation
1080/10298420290030569. under specific conditions leads to reduced interest in reward in adulthood in Wistar
Lippmann, M., Bress, A., Nemeroff, C.B., Plotsky, P.M., Monteggia, L.M., 2007. Long-term rats. Behav. Brain Res. 156, 297–310. http://dx.doi.org/10.1016/j.bbr.2004.06.001.
behavioural and molecular alterations associated with maternal separation in rats. Rampon, C., Tsien, J.Z., 2000. Genetic analysis of learning behavior-induced structural
Eur. J. Neurosci. 25, 3091–3098. http://dx.doi.org/10.1111/j.1460-9568.2007. plasticity. Hippocampus 10, 605–609. http://dx.doi.org/10.1002/1098-1063(2000)
05522.x. 10:5<605:AID-HIPO11>3.0.CO;2-3.
Liu, D., Caldji, C., Sharma, S., Plotsky, P.M., Meaney, M.J., 2000. Influence of neonatal Rampon, C., Rampon, C., Jiang, C.H., Jiang, C.H., Dong, H., Dong, H., Tang, Y.P., Tang,
rearing conditions on stress-induced adrenocorticotropin responses and nor- Y.P., Lockhart, D.J., Lockhart, D.J., Schultz, P.G., Schultz, P.G., Tsien, J.Z., Tsien,
epinepherine release in the hypothalamic paraventricular nucleus. J. J.Z., Hu, Y., Hu, Y., 2000. Effects of environmental enrichment on gene expression in
Neuroendocrinol. 12, 5–12. the brain. Proc. Natl. Acad. Sci. U. S. A. 97, 12880–12884. http://dx.doi.org/10.
Liu, H.-X., Zhang, J.-J., Zheng, P., Zhang, Y., 2005. Altered expression of MAP-2, GAP-43, 1073/pnas.97.23.12880.
and synaptophysin in the hippocampus of rats with chronic cerebral hypoperfusion Rice, D., Barone, S., 2000. Critical periods of vulnerability for the developing nervous
correlates with cognitive impairment. Brain Res. Mol. Brain Res. 139, 169–177. system: evidence from humans and animal models. Environ. Health Perspect.
http://dx.doi.org/10.1016/j.molbrainres.2005.05.014. 511–533.
Lu, B., Pang, P.T., Woo, N.H., 2005. The yin and yang of neurotrophin action. Nat. Rev. Riley, V., 1981. Psychoneuroendocrine influences on immunocompetence and neoplasia.
Neurosci. 6, 603–614. http://dx.doi.org/10.1038/nrn1726. Science 212, 1100–1109. http://dx.doi.org/10.1126/SCIENCE.7233204.
MacQueen, G.M., Ramakrishnan, K., Ratnasingan, R., Chen, B., Young, L.T., 2003. Roceri, M., Hendriks, W., Racagni, G., Ellenbroek, B.a., Riva, M.a., 2002. Early maternal
Desipramine treatment reduces the long-term behavioural and neurochemical se- deprivation reduces the expression of BDNF and NMDA receptor subunits in rat
quelae of early-life maternal separation. Int. J. Neuropsychopharmacol. 6, 391–396. hippocampus. Mol. Psychiatry 7, 609–616. http://dx.doi.org/10.1038/sj.mp.
http://dx.doi.org/10.1017/S1461145703003729. 4001036.
Markostamou, I., Ioannidis, A., Dandi, E., Mandyla, M.A., Nousiopoulou, E., Simeonidou, Rodgers, R.J., Dalvi, A., 1997. Anxiety, defence and the elevated plus-maze. Neurosci.
C., Spandou, E., Tata, D.A., 2016. Maternal separation prior to neonatal hypoxia- Biobehav. Rev. 21, 801–810. http://dx.doi.org/10.1016/S0149-7634(96)00058-9.
ischemia: impact on emotional aspects of behavior and markers of synaptic plasticity Roth, T.L., Lubin, F.D., Funk, A.J., Sweatt, J.D., 2009. Lasting epigenetic influence of
in hippocampus. Int. J. Dev. Neurosci. 52, 1–12. http://dx.doi.org/10.1016/j. early-life adversity on the BDNF gene. Biol. Psychiatry 65, 760–769. http://dx.doi.
ijdevneu.2016.04.002. org/10.1016/j.biopsych.2008.11.028.
Mcewen, B.S., 1997. Stress and hippocampal LTD. Neuroscience 3http://dx.doi.org/10. Sampedro-Piquero, P., Begega, A., Arias, J.L., 2014. Increase of glucocorticoid receptor
1177/107385849700300601. (355-355). expression after environmental enrichment: relations to spatial memory, exploration
Meng, Q., Li, N., Han, X., Shao, F., Wang, W., 2011. Effects of adolescent social isolation and anxiety-related behaviors. Physiol. Behav. 129, 118–129. http://dx.doi.org/10.
on the expression of brain-derived neurotrophic factors in the forebrain. Eur. J. 1016/j.physbeh.2014.02.048.
Pharmacol. 650, 229–232. http://dx.doi.org/10.1016/J.EJPHAR.2010.09.061. Schreiber, S., Lin, R., Haim, L., Baratz-Goldstien, R., Rubovitch, V., Vaisman, N., Pick,
Mitchelmore, C., Gede, L., 2014. Brain Derived Neurotrophic Factor: epigenetic regula- C.G., 2014. Enriched environment improves the cognitive effects from traumatic
tion in psychiatric disorders. Brain Res. 1586, 162–172. http://dx.doi.org/10.1016/j. brain injury in mice. Behav. Brain Res. 271, 59–64. http://dx.doi.org/10.1016/j.bbr.
brainres.2014.06.037. 2014.05.060.
Molteni, R., Calabrese, F., Cattaneo, A., Mancini, M., Gennarelli, M., Racagni, G., Riva, Shalev, U., Kafkafi, N., 2002. Repeated maternal separation does not alter sucrose-re-
M.A., 2009. Acute stress responsiveness of the neurotrophin BDNF in the rat hippo- inforced and open-field behaviors. Pharmacol. Biochem. Behav. 73, 115–122.
campus is modulated by chronic treatment with the antidepressant duloxetine. Shilpa, B., Bhagya, V., Harish, G., Srinivas Bharath, M., Shankaranarayana Rao, B., 2017.
Neuropsychopharmacology 34, 1523–1532. http://dx.doi.org/10.1038/npp.2008. Environmental enrichment ameliorates chronic immobilisation stress-induced spatial
208. learning deficits and restores the expression of BDNF, VEGF, GFAP and glucocorticoid
Monteiro, B.M.M., Moreira, F.a., Massensini, A.R., Moraes, M.F.D., Pereira, G.S., 2014. receptors. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 76, 88–100. http://dx.doi.
Enriched environment increases neurogenesis and improves social memory persis- org/10.1016/j.pnpbp.2017.02.025.
tence in socially isolated adult mice. Hippocampus 24, 239–248. http://dx.doi.org/ Shinohara, K., Hata, T., 2014. Post-acquisition hippocampal NMDA receptor blockade
10.1002/hipo.22218. sustains retention of spatial reference memory in Morris water maze. Behav. Brain
Morris, R., 1984. Developments of a water-maze procedure for studying spatial learning Res. 259, 261–267. http://dx.doi.org/10.1016/j.bbr.2013.11.016.
in the rat. J. Neurosci. Methods 11, 47–60. http://dx.doi.org/10.1016/0165- Slotten, H.a., Kalinichev, M., Hagan, J.J., Marsden, C.a., Fone, K.C.F., 2006. Long-lasting
0270(84)90007-4. changes in behavioural and neuroendocrine indices in the rat following neonatal
Mosaferi, B., Babri, S., Mohaddes, G., Khamnei, S., Mesgari, M., 2015. Post-weaning maternal separation: gender-dependent effects. Brain Res. 1097, 123–132. http://dx.
environmental enrichment improves BDNF response of adult male rats. Int. J. Dev. doi.org/10.1016/j.brainres.2006.04.066.
Neurosci. 46, 108–114. http://dx.doi.org/10.1016/j.ijdevneu.2015.07.008. Smith, M.A., Makino, S., Kvetnansky, R., Post, R.M., 1995. Stress and glucocorticoids
Mourlon, V., Baudin, A., Blanc, O., Lauber, A., Giros, B., Naudon, L., Daugé, V., 2010. affect the expression of brain-derived neurotrophic factor and neurotrophin-3 mRNAs
Maternal deprivation induces depressive-like behaviours only in female rats. Behav. in the hippocampus. J. Neurosci. 15, 1768–1777.
Brain Res. 213, 278–287. http://dx.doi.org/10.1016/j.bbr.2010.05.017. Sousa, V.C., Vital, J., Costenla, A.R., Batalha, V.L., Sebastião, A.M., Ribeiro, J.a., Lopes,

31
Ε. Dandi et al. International Journal of Developmental Neuroscience 67 (2018) 19–32

L.V., 2014. Maternal separation impairs long term-potentiation in CA1-CA3 synapses mild stress persistent after enrichment cessation in six-month-old female Balb/C
and hippocampal-dependent memory in old rats. Neurobiol. Aging 35, 1680–1685. mice. Behav. Brain Res. 301, 72–83. http://dx.doi.org/10.1016/j.bbr.2015.12.028.
http://dx.doi.org/10.1016/j.neurobiolaging.2014.01.024. Vetulani, J., 2013. Early maternal separation: a rodent model of depression and a pre-
Stein, L.R., O’Dell, K.A., Funatsu, M., Zorumski, C.F., Izumi, Y., 2016. Short-term en- vailing human condition. Pharmacol. Rep. 65, 1451–1461. http://dx.doi.org/10.
vironmental enrichment enhances synaptic plasticity in hippocampal slices from aged 1016/S1734-1140(13)71505-6.
rats. Neuroscience 329, 294–305. http://dx.doi.org/10.1016/j.neuroscience.2016. Vivinetto, A.L., Suárez, M.M., Rivarola, M.A., 2013. Neurobiological effects of neonatal
05.020. maternal separation and post-weaning environmental enrichment. Behav. Brain Res.
Stiles, J., 2011. Brain development and the nature versus nurture debate. Progress in 240, 110–118. http://dx.doi.org/10.1016/j.bbr.2012.11.014.
Brain Research, 1st ed. Elsevier B.V.http://dx.doi.org/10.1016/B978-0-444-53884-0. Vorhees, C.V., Williams, M.T., 2006. Morris water maze: procedures for assessing spatial
00015-4. and related forms of learning and memory. Nat. Protoc. 1, 848–858. http://dx.doi.
Suchecki, D., Duarte Palma, B., Tufik, S., 2000. Pituitary-adrenal axis and behavioural org/10.1038/nprot.2006.116.
responses of maternally deprived juvenile rats to the open field. Behav. Brain Res. Walf, A.a., Frye, C.a., 2007. The use of the elevated plus maze as an assay of anxiety-
111, 99–106. related behavior in rodents. Nat. Protoc. 2, 322–328. http://dx.doi.org/10.1038/
Tata, D.A., Markostamou, I., Ioannidis, A., Gkioka, M., Simeonidou, C., Anogianakis, G., nprot.2007.44.
Spandou, E., 2015. Effects of maternal separation on behavior and brain damage in Wang, Q., Li, M., Du, W., Shao, F., Wang, W., 2015a. The different effects of maternal
adult rats exposed to neonatal hypoxia-ischemia. Behav. Brain Res. 280, 51–61. separation on spatial learning and reversal learning in rats. Behav. Brain Res. 280,
http://dx.doi.org/10.1016/j.bbr.2014.11.033. 16–23. http://dx.doi.org/10.1016/j.bbr.2014.11.040.
Thiel, G., 1993. Synapsin I, synapsin II, and synaptophysin: marker proteins of synaptic Wang, Q., Shao, F., Wang, W., 2015b. Maternal separation produces alterations of fore-
vesicles. Brain Pathol. 3, 87–95. http://dx.doi.org/10.1111/j. 1750–3639.1993. brain brain-derived neurotrophic factor expression in differently aged rats. Front.
tb00729.x. Mol. Neurosci. 8, 49. http://dx.doi.org/10.3389/fnmol.2015.00049.
Thome, J., Pesold, B., Baader, M., Hu, M., Gewirtz, J.C., Duman, R.S., Henn, F.A., 2001. Wigger, A., Neumann, I.D., 1999. Periodic maternal deprivation induces gender-depen-
Stress differentially regulates synaptophysin and synaptotagmin expression in hip- dent alterations in behavioral and neuroendocrine responses to emotional stress in
pocampus. Biol. Psychiatry 50, 809–812. http://dx.doi.org/10.1016/S0006- adult rats. Physiol. Behav. 66, 293–302.
3223(01)01229-X. Wright, R.L., Conrad, C.D., 2008. Enriched environment prevents chronic stress-induced
van Driel, K.S., Talling, J.C., 2005. Familiarity increases consistency in animal tests. spatial learning and memory deficits. Behav. Brain Res. 187, 41–47. http://dx.doi.
Behav. Brain Res. 159, 243–245. http://dx.doi.org/10.1016/j.bbr.2004.11.005. org/10.1016/j.bbr.2007.08.025.
Valtorta, F., Pennuto, M., Bonanomi, D., Benfenati, F., 2004. Synaptophysin: leading actor Xiong, G.-J., Yang, Y., Cao, J., Mao, R.-R., Xu, L., 2015. Fluoxetine treatment reverses the
or walk-on role in synaptic vesicle exocytosis? Bioessays 26, 445–453. http://dx.doi. intergenerational impact of maternal separation on fear and anxiety behaviors.
org/10.1002/bies.20012. Neuropharmacology 92, 1–7. http://dx.doi.org/10.1016/j.neuropharm.2014.12.026.
Veena, J., Srikumar, B.N., Mahati, K., Bhagya, V., Raju, T.R., Shankaranarayana Rao, B.S., Yang, J., Hou, C., Ma, N., Liu, J., Zhang, Y., Zhou, J., Xu, L., Li, L., 2007. Enriched
2009. Enriched environment restores hippocampal cell proliferation and ameliorates environment treatment restores impaired hippocampal synaptic plasticity and cog-
cognitive deficits in chronically stressed rats. J. Neurosci. Res. 87, 831–843. http:// nitive deficits induced by prenatal chronic stress. Neurobiol. Learn. Mem. 87,
dx.doi.org/10.1002/jnr.21907. 257–263. http://dx.doi.org/10.1016/j.nlm.2006.09.001.
Veenema, A.H., Bredewold, R., Neumann, I.D., 2007. Opposite effects of maternal se- Zanca, R.M., Braren, S.H., Maloney, B., Schrott, L.M., Luine, V.N., Serrano, P.A., 2015.
paration on intermale and maternal aggression in C57BL/6 mice: link to hypotha- Environmental enrichment increases glucocorticoid receptors and decreases GluA2
lamic vasopressin and oxytocin immunoreactivity. Psychoneuroendocrinology 32, and protein kinase M zeta (PKMζ) trafficking during chronic stress: a protective
437–450. http://dx.doi.org/10.1016/j.psyneuen.2007.02.008. mechanism? Front. Behav. Neurosci. 9, 303. http://dx.doi.org/10.3389/fnbeh.2015.
Vega-Rivera, N.M., Ortiz-López, L., Gómez-Sánchez, A., Oikawa-Sala, J., Estrada- 00303.
Camarena, E.M., Ramírez-Rodríguez, G.B., 2016. The neurogenic effects of an en-
riched environment and its protection against the behavioral consequences of chronic

32

You might also like