You are on page 1of 14

822141

research-article2019
JOP0010.1177/0269881118822141Journal of PsychopharmacologyMillard et al.

Original Article

Perinatal exposure to fluoxetine increases


anxiety- and depressive-like behaviours and
alters glutamatergic markers in the prefrontal Journal of Psychopharmacology

cortex and hippocampus of male adolescent


1­–14
© The Author(s) 2019
Article reuse guidelines:
rats: A comparison between Sprague-Dawley sagepub.com/journals-permissions
DOI: 10.1177/0269881118822141
https://doi.org/10.1177/0269881118822141

rats and the Wistar-Kyoto rat model of journals.sagepub.com/home/jop

depression

Samuel J Millard1,2, Jeremy S Lum1,2, Francesca Fernandez3, Katrina


Weston-Green1,2 and Kelly A Newell1,2

Abstract
Background: With approximately 10% of pregnant women prescribed antidepressant drugs for the treatment of depressive disorders, there is growing
concern regarding the potential long-term effects of this exposure on offspring. Research is needed in clinically relevant models to determine the
effects on offspring behaviour and associated neurobiological systems.
Aim: The aim of this study was to determine the effects of maternal fluoxetine treatment on anxiety-like and depressive-like behaviours in adolescent
offspring as well as associated glutamatergic markers, using a clinically relevant rodent model of depression.
Methods: Wistar-Kyoto (model of innate depression) and Sprague-Dawley rats were treated with fluoxetine (10 mg/kg) from gestational day 0
to postnatal day 14. Male offspring underwent behavioural testing (open field, elevated plus maze, forced swim test) at adolescence followed by
quantitative immuno-detection of glutamatergic markers in the prefrontal cortex and ventral hippocampus.
Results: Perinatal fluoxetine exposure exacerbated the anxiety-like and depressive-like phenotype in Wistar-Kyoto offspring and induced an anxiety-
like and depressive-like phenotype in Sprague-Dawley offspring. Wistar-Kyoto offspring showed reductions in NMDA receptor NR1, NR2A and NR2B
subunits, as well as post-synaptic density 95 (PSD-95) and metabotropic glutamate receptor subtype 1 (mGluR1) in the prefrontal cortex; perinatal
fluoxetine exposure further reduced NR1, NR2A, PSD-95 and mGluR1 expression in Wistar-Kyoto as well as Sprague-Dawley offspring. In the ventral
hippocampus perinatal fluoxetine exposure reduced PSD-95 and increased metabotropic glutamate receptor subtype 5 (mGluR5) and Homer1b/c in
both Sprague-Dawley and Wistar-Kyoto strains.
Conclusion: These findings suggest that maternal fluoxetine treatment exacerbates effects of underlying maternal depression on offspring behaviour,
which may be mediated through alterations in the glutamatergic system. Further research investigating how to minimise these effects, whilst ensuring
optimal treatment for mothers, is essential to move the field forward.

Keywords
Maternal depression, offspring, perinatal selective serotonin reuptake inhibitor, fluoxetine, antidepressant, Wistar-Kyoto, glutamate, N-methyl-D-
aspartate receptor, metabotropic glutamate receptor, post-synaptic density 95

Introduction the most frequently prescribed antidepressants during pregnancy


(in 80% of cases) are the selective serotonin reuptake inhibitors
Major depressive disorder (MDD) is one of the most common (SSRIs) (Mitchell et al., 2011; Oberlander et al., 2009). SSRIs
mental illnesses affecting an estimated 300 m people worldwide such as fluoxetine (FLX), bind to the serotonin transporter,
(World Health Organization, 2017). The overall incidence of
MDD has risen significantly over the last several decades
(Huybrechts et al., 2013; Kessler, 2003). Women display twice 1Molecular Horizons and School of Medicine, University of Wollongong,
the risk ratio compared to men, with the greatest risk of develop- Wollongong, NSW, Australia
2Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
ing the disorder being at childbearing age (Huybrechts et al.,
3School of Science, Australian Catholic University, Brisbane, QLD, Australia
2013; Marcus and Heringhausen, 2009). Untreated maternal
MDD and stress-related disorders are associated with adverse Corresponding author:
outcomes for mother and child (Deave et al., 2008; Ronald et al., Kelly A Newell, School of Medicine, Faculty of Science, Medicine and
2011). Approximately 10% of pregnant women are prescribed Health, University of Wollongong, Northfields Ave, Wollongong, NSW
antidepressant drugs for the treatment of depressive and stress- 2522, Australia.
related disorders (Cooper et al., 2007; Huybrechts et al., 2013); Email: knewell@uow.edu.au
2 Journal of Psychopharmacology 00(0)

blocking the reabsorption of serotonin by the presynaptic neuron (vHPC) in neurodevelopmental and psychiatric disorders and their
resulting in increased serotonin levels in the synaptic cleft biological sensitivity to the action of SSRIs (Brandon and McKay,
(Hiemke and Härtter, 2000). It has been shown that SSRIs admin- 2015; Dusi et al., 2015), these brain areas were investigated. By
istered during pregnancy cross the placenta (by as much as ~45– comparing the effects of maternal SSRI treatment in a rodent
70% of maternal levels) (Heikkinen et al., 2002) and are excreted model of innate depression (Wistar-Kyoto (WKY)) to that of a
in breast milk (Hendrick et al., 2001; Kim et al., 2006), exposing standard laboratory rat strain (Sprague-Dawley (SD)), we aimed to
the developing foetus and newborn. test the hypothesis that maternal depressive phenotypes (which
The serotonergic system plays an important role in early brain model depressive disorders) could influence the effects of SSRI
development facilitating a number of neurodevelopmental pro- exposure on offspring neurobiology and behaviour.
cesses such as neuronal proliferation, migration and differentia-
tion (Vitalis and Parnavelas, 2003); of particular interest, the
serotonergic system regulates the development and maturation of Materials and methods
the brain’s primary excitatory neurotransmitter system, the gluta-
Animals
matergic system (Bonnin et al., 2007; Edgar and Price, 2001;
Lavdas et al., 1997; Vitalis and Parnavelas, 2003). Dysfunction of All experiments were approved by the University of Wollongong’s
the glutamatergic system, in particular the N-methyl-D-aspartate Animal Ethics Committee in accordance with the Australian Code for
(NMDA) ionotropic receptors and Group I metabotropic recep- the Care and Use of Animals for Scientific Purposes (AE14/31). To
tors, has long been linked to neurodevelopmental and psychiatric separate the effects of maternal FLX treatment from that of innate
disorders including autism, attention deficit hyperactivity disorder maternal depression, SD (control strain) and WKY (model of innate
(ADHD), depression and schizophrenia (Deschwanden et al., depression) rats were used in the study. The WKY rat strain has long
2011; Hashimoto et al., 2007; Lakhan et al., 2013; Matosin et al., been established as a model of innate depression, displaying exagger-
2013; Pérez de la Mora et al., 2006; Yuan et al., 2015). In line with ated stress responses, increased anhedonia, learned helplessness,
this, SSRI use during pregnancy has been associated with decreased locomotor activity and increased anxiety- and depressive-
increased risk of autism (Boukhris et al., 2016; Croen et al., 2011; like behaviours (López-Rubalcava and Lucki, 2000; Nam et al.,
Gidaya et al., 2014; Harrington et al., 2014), ADHD (Clements 2014; Pare and Tejani-Butt, 1996; Rittenhouse et al., 2002; Will et al.,
et al., 2015; Figueroa, 2010), depression (Malm et al., 2016) and 2003). The WKY strain is also characterised by alterations in stress
anxiety (Brandlistuen et al., 2015) in exposed children. However, hormones and serotonergic alterations similar to those previously
current evidence is conflicting and a number of studies have reported in depressed individuals (Arango et al., 2001; Lemos et al.,
reported no significant association between maternal SSRI expo- 2011; López-Figueroa et al., 2004; Pardon et al., 2002). Studies have
sure and risk of neurodevelopmental or neuropsychiatric disor- reported increases in stress hormones such as basal corticosterone
ders in offspring (for review see Millard et al., 2017). Additionally, and ACTH (Pardon et al., 2002; Will et al., 2003) and increased levels
there is an inherent difficulty in discerning the effects of SSRI of pro-opiomelanocortin in response to acute stress in comparison to
exposure from that of the innate maternal depression in clinical other strains (Redei et al., 1994; Will et al., 2003). The WKY strain
studies, especially given that maternal depression and adversity also shows decreased basal serotonin (5HT) concentrations,
are themselves associated with an increased risk for altered neu- decreased 5-hydroxyindoleacetic acid (5HIAA; metabolite of 5HT),
rodevelopment and associated disorders in offspring (Deave et al., decreased serotonin transporter (SERT) density in hippocampus and
2008; Glover, 2014; Ronald et al., 2011). cortical regions, decreased 5HT1A receptor and a reduction in the rate-
Preclinical studies investigating the effects of developmental limiting serotonin-producing enzyme, tryptophan hydroxylase
SSRI exposure on offspring behaviour in rodents have reported (Lemos et al., 2011; Pare and Tejani-Butt, 1996; Scholl et al., 2010).
alterations in social interaction, cognitive function, anxiety- and Male and female SD and WKY rats (12 weeks of age) were
depressive-like behaviours in adolescent and adult offspring (for obtained from the Animal Resource Centre, Western Australia. Rats
review see Millard et al., 2017). However, there has been no were housed at the University of Wollongong Animal Housing
investigation of the effects of SSRI exposure in rodent models of Facility in a temperature-controlled environment (20±20C) under a
innate depression that closely model MDD. Determining the 12:12-hour light-dark cycle (lights on at 07:00). The rats were pro-
effects of maternal SSRI treatment on offspring behaviour in vided with ad libitum access to water and laboratory chow. Females
rodent models of innate depression and understanding the neuro- were housed one per cage (cage size 38×25×16 cm) to allow mon-
biological changes will be key to determining the clinical risk itoring of drug intake; males were housed two per cage of the same
associated with maternal SSRI use in context of the underlying strain. Following a minimum of one-week acclimatisation, male
maternal psychopathology. and female SD or WKY rats were paired to elicit mating. At the
The aim of this study was to investigate the effects of maternal conclusion of the mating period, pregnant dams were randomly
SSRI (FLX) treatment on offspring behaviour and neurobiology assigned to a FLX or vehicle (VEH) (water) treatment group.
relevant to neurodevelopmental and psychiatric disorders. Following the birth of pups, which was designated as post-natal day
Specifically, given the key role of serotonin in the development of (PN) 0, pups were housed with their dam and litter mates until
the glutamatergic system and the importance of the glutamatergic PN21–28 days of age, at which time they were weaned and housed
system in neurodevelopmental disorders, we sought to investigate 2–3 per cage with their littermates of the same sex. Perinatal FLX
the potential influence that maternal SSRI treatment has on key treatment did not influence litter sizes (VEH: 10.3±0.88 pups/litter;
glutamatergic markers at adolescence, particularly associated with FLX: 9.3±0.98 pups/litter; p>0.05), however dams of the WKY
the NMDA and Group I metabotropic glutamate receptors and strain did have significantly smaller litters compared to SD dams
associated scaffolding proteins. Furthermore, given the signifi- (SD: 12.4±0.66 pups/litter; WKY: 7.6±0.63 pups/litter; p<0.001).
cance of the prefrontal cortex (PFC) and ventral hippocampus Pup body weights were determined at PN7, PN14 and PN42 as
Millard et al. 3

Figure 1.  Treatment paradigm and experimental timeline.


EPM: elevated plus maze; FLX: fluoxetine; FST: forced swim test; GD: gestational day; OFT: open field test; PFC: prefrontal cortex; PN: postnatal day; SD: Sprague-Dawley;
VEH: vehicle; vHPC: ventral hippocampus; WKY: Wistar-Kyoto.

measures of potential SSRI teratogenicity. Behavioural tests and FLX treated dams compared to VEH treated dams (173.5±7.7 vs
immunoblot analyses were performed at adolescence. Male off- 206.6±10.3 g water / kg body weight; p=0.021).
spring were the initial focus of this study, given the increased preva- Bottles were replaced with fresh FLX or vehicle solution
lence in males for neurodevelopmental disorders such as autism daily to minimise drug degradation while also allowing dosage to
(4:1) and ADHD (2.3:1), (Ramtekkar et al., 2010; Werling and be adjusted based on dams body weight and water intake. The
Geschwind, 2013), and the reported association between maternal dose of 10 mg/kg was chosen based on previous animal models
FLX treatment and the risk for these disorders (Millard et al., 2017). that have shown this dose to produce antidepressant-like effects
However, it will be important to investigate the potential effects of in rodents (Dulawa et al., 2004; Kiryanova et al., 2016; Thompson
perinatal FLX exposure on female offspring in future studies. et al., 2004). Moreover, human neuroimaging studies have dem-
onstrated that clinically efficacious doses of SSRIs have resulted
in 80% or greater SERT occupancy; a paradigm that has been
Maternal SSRI treatment achieved previously in rodents with dosages of 8–12 mg/kg/day
Pregnant SD and WKY dams were dosed with FLX hydrochloride of FLX (Capello et al., 2011). The rationale behind the selected
(LKT Laboratories, St Paul, Minnesota, USA) (10 mg/kg dis- drug administration method was based on the desire to minimise
solved in tap water) or VEH (tap water), for the full gestational unwarranted stress to the animals that may occur using subcuta-
term and a further two weeks of the lactation period (i.e. five neous or intraperitoneal injections as well as to model a more
weeks in total), with the last day of treatment corresponding to clinically relevant scenario in which FLX is primarily adminis-
PN14 of the offspring, equivalent to birth in humans (Semple tered orally (Koran et al., 1996). Finally, this method and dosage
et al., 2013). In total, 27 dams (and litters) were used in this study is generally well accepted within the current literature (Ansorge
comprising 12 FLX-treated dams (6×SD, 6×WKY) and 15 et al., 2008, 2004; Bairy et al., 2006; Cabrera-Vera and Battaglia,
VEH-treated dams (7×SD, 8×WKY) (Figure 1). FLX was 1998; Karpova et al., 2009; Kiryanova et al., 2013; Olivier et al.,
administered in light-resistant gravity-fed water bottles to mini- 2011; Rodriguez-Porcel et al., 2011; Thompson et al., 2004).
mise drug degradation via photolysis (Lam et al., 2005). Dam
water consumption and body weight were recorded daily through-
Behavioural testing
out the treatment period to ensure the correct dosage. While WKY
dams weighed less than SD dams (~25%), there were no differ- Male offspring underwent a series of behavioural tests to meas-
ences between the strains or the treatment groups in percentage of ure primarily anxiety- and depressive-like behaviours using the
body weight change during gestation (p=0.818) or lactation open field test (OFT), elevated plus maze (EPM) and forced
(p=0.599) periods. The average daily water consumption over the swim test (FST), (n=8–14/group) (Figure 1). Behavioural testing
treatment period was lower in WKY dams compared to SD dams began at PN35, corresponding to the adolescent period in rodents
(42.4±3.7 vs 53.8±2.1 ml/day; p=0.002) and lower in the FLX (Semple et al., 2013) and was conducted between 12:00–1700.
treated dams compared to the VEH treated dams (43.3±2.6 vs The procedures for behavioural testing in the OFT, EPM and FST
57.6±2.6 ml/day; p=0.001). However, when accounting for differ- were based on protocols previously implemented by du Bois
ences in body weight between the dams there was no significant et al. (2008), Foldi et al. (2011) and Turner and Burne (2014).
difference in average water intake in WKY vs SD dams over the Testing was conducted from least stressful to most stressful in the
treatment period (177.4±9.5 vs 202.7±8.7 g water/kg bodyweight; order of OFT<EPM<FST, to limit any behavioural influences
p=0.068); there was however a 16% lower water intake in the that may result from prior testing (McIlwain et al., 2001).
4 Journal of Psychopharmacology 00(0)

Additionally, rat offspring were given one day of rest in between between subjects and the procedure repeated. Data was quantified
behavioural tests as suggested by Paylor et al. (2006). based on the duration of time that the rat spent unengaged in
escape-driven activity (floating in water without active move-
ments of forepaws) represented by total immobility time (passive
OFT behaviour).
The OFT was used as a measure of locomotion, however time Behaviour from all three tests was video-recorded. For the OFT
spent in different zones of the arena also provides information on and EPM, behaviour was analysed using video tracking software
anxiety-like behaviours (Prut and Belzung, 2003). Parameters (Ethovision, Noldus Information Technology). For the FST, immo-
analysed included total distance travelled (as a measure of loco- bility time was measured manually by two assessors blinded to
motor activity), time spent in the centre of the arena and time experimental conditions, inter-rater reliability between the two
spent in corners (as measures of anxiety-like behaviour). The assessors was determined using SPSS (Intraclass correlation coef-
OFT apparatus was made up of a 60 cm×60 cm square arena ficient=0.998, p<0.001). Baseline mean immobility scores for SD
with 40 cm high walls. Lighting remained constant throughout and WKY strains were similar to those previously reported (Abe
the procedure with light intensity set at ~20 lux. Upon com- et al., 2007; Calvo et al., 2011; Jama et al., 2008; Morley-Fletcher
mencement of the test a single rat was placed within the centre et al., 2011; Nam et al., 2014).
arena of the OFT, and remained within the apparatus for a testing
duration of 10 min. Immunoblotting
Rats were euthanised 48 h after their final behavioural test
EPM (PN42) via carbon dioxide (CO2) asphyxiation and the PFC
(Bregma +3.70 mm) and vHPC (Bregma −2.80 mm) were dis-
The EPM was used as a measure of anxiety-like behaviour, explo-
sected according to a standard rat brain atlas (Paxinos and
ration and locomotor activity (Walf and Frye, 2007). The main
Watson, 2007). Equal amounts of protein (5 ug/well) were sepa-
determining parameter of anxiety-like behaviour was the amount
rated in 4–20% Tris-Glycine-eXtended (TGX) precast gels (Bio-
of activity in the open arm of the apparatus versus that of the
Rad, Australia) under non-reducing conditions (Lum et al.,
closed arm, corresponding to increased or decreased anxiety-like
2016). All protein-loading concentrations were in the linear
behaviours as the animal engages in or refrains from exploratory
range of detection for each antibody. Following electrophoresis,
behaviour (Walf and Frye, 2007). The EPM apparatus comprised
proteins were transferred onto polyvinylidene difluoride mem-
of one pair of closed, and one pair of open arms, with respective
branes (Bio-Rad) and membranes blocked with 5% bovine
dimensions 50 cm×7 cm×30 cm (length×width×height) and 50
serum albumin or skim milk (w/v) for 60 min at room tempera-
cm×7 cm×1 cm. There was an open roof and the apparatus was
ture. The membranes were incubated overnight in primary poly-
arranged around a centre platform, with the arms perpendicular to
clonal antibodies (NR1, NR2A, NR2B, post-synaptic density 95
each other. The maze was elevated 60 cm above the ground and
(PSD-95), mGluR1, mGluR5, Homer1b/c and β-Actin;
the light intensity set at a constant of ~100 lux across the open
Supplementary Material Table 1). Membranes were subse-
arms. Rats were tested individually for five minutes, beginning
quently incubated with horseradish peroxidase conjugated sec-
with the placement of the subject within the centre of the appara-
ondary antibodies. Bands were visualised using Amersham ECL
tus facing an open arm.
Western blotting detection reagent (GE Healthcare, Australia)
and membranes exposed to Hyperfilm (GE Healthcare,
Australia). Films were scanned using a GS-800 scanner (Bio-
FST
Rad) and densitometry values were quantified. Relative densi-
The FST is widely implemented to assess escape-driven behav- tometry values for each protein were normalised to their
iour in the face of an inescapable aversive situation (Commons respective β-actin levels and an internal control value, to account
et al., 2017; Slattery and Cryan, 2012). Passive behaviour (i.e. for protein loading and gel-gel variability, respectively. Each
immobility) is typically measured, and was the focus of the FST sample (n=7/group) was run in triplicate.
given that it can be clearly defined and is distinguishable from
active behaviour and coping measures (such as swimming and
climbing), which can be difficult to reliably measure across exper-
Statistical analysis
imental observers (Slattery and Cryan, 2012). Increased immobil- Two-way analyses of variance (ANOVAs) were used to deter-
ity has previously been associated with ‘depressive-like’ behaviour mine any effects of treatment (FLX or VEH) or strain (WKY or
due to the observation that antidepressants increase the duration SD) on body weight (PN42), behavioural parameters and relative
of time the rodent spends engaged in active and escape-driven protein densities. Where significant interactions were found, one-
behaviour (Castagné et al., 2011). Rats were tested under ceiling way ANOVAs were used to identify differences between groups.
lighting (500–750 lux) in an opaque cylinder (30 cm diameter Kruskal–Wallis tests were used to determine the effects of treat-
×50 cm depth) filled to a level 1.25×length of the rat (snout to ment and strain on offspring body weight (PN7+PN14) and
tail) with (25±1oC) water (Slattery and Cryan, 2012). The rats behavioural data (OFT: Time spent in centre arena), where data
were tested over two days, the first day of which involved famil- showed non-normal distribution (Kolmogorov–Smirnov
iarising the rat to the test environment by placing the subject in the p<0.05). Where significant interactions were found, a post hoc
water for 15 min. On the second day (24-hour post-habituation), pairwise comparison test was performed to detect significant dif-
the subject was placed in the test apparatus filled to a predeter- ferences between groups. Significance was set at an alpha level
mined level with water and observed for 5 min; water was changed of p=0.05.
Millard et al. 5

Figure 2.  Average offspring body weight at (a) postnatal day (PN)7, (b) PN14 and (c) PN42. Data expressed as mean grams (g)+standard error of
the mean (SEM); **p<0.010, ***p<0.001. SD: Sprague-Dawley; WKY: Wistar-Kyoto.

Figure 3.  (a) Distance travelled, (b) time spent in the centre arena and (c) time spent in corners of the open field test (OFT). Data expressed as
mean centimetres (cm) or seconds (sec)+standard error of the mean (SEM); ^p=0.056, *p<0.05, **p<0.010, ***p<0.001; ###p<0.001 refers to
significant difference in Wistar-Kyoto (WKY) compared to Sprague-Dawley (SD) (overall strain effect).

Results (–61%; p<0.01) but this did not reach significance in WKY off-
spring. Similarly, there was a significant effect of TREATMENT
Offspring body weight (χ2=162.0; p<0.05), STRAIN (χ2=93.0; p<0.05) and a
Kruskal-Wallis tests revealed significant effects of TREATMENT×STRAIN interaction (χ2=18.824; p<0.001) on
TREATMENT (PN7: χ2=15.553; p<0.001; PN14: χ2=9.579; time spent in the centre of the OFT arena; WKY-VEH offspring
p<0.05), STRAIN (PN7: χ2=77.046; p<0.001; PN14: spent a substantially decreased amount of time in the centre
χ2=41.175; p<0.001) and significant TREATMENT×STRAIN arena compared to SD-VEH offspring (-95%; p<0.05) while
interactions (PN7: χ2=96.702; p<0.001; PN14: χ2=69.244; FLX-exposed SD, but not WKY, rats spent less time in the cen-
p<0.001) on body weight of offspring at PN7 and PN14. FLX- tre of the OFT area compared to SD-VEH rats (–90%; p=0.056).
exposed offspring of both strains weighed significantly less There were also significant effects of TREATMENT and
compared to VEH offspring at PN7 (SD: –12%, p<0.05; WKY: STRAIN on time spent in the corners of the OFT, with WKY
–10%, p<0.001) and PN14 (SD: –8%, p<0.05; WKY: –17%, offspring spending increased time in the corners compared to
p<0.01). This effect extended to PN42 in male SD offspring, SD offspring (+10%; F1,47 =14.448; p<0.001) and FLX-
but not WKY offspring, with SD FLX offspring weighing sig- exposed offspring of both strains overall spending increased
nificantly less compared to SD VEH offspring (–15%, p<0.05). time in the corners compared to VEH (+10%; F1,47=12.781;
WKY VEH offspring also weighed less than SD VEH offspring p<0.05). There was no TREATMENT×STRAIN interaction on
at PN7 (–19%, p<0.001), PN14 (–11.1%, p<0.001) and PN42 time spent in corners (Figure 3).
(–42%, p<0.001) (Figure 2).
EPM.  Adolescent WKY offspring travelled less in the EPM com-
Adolescent behaviour pared to SD offspring (–27%; F1,48=8.974; p<0.004) and FLX-
exposed offspring travelled less in the EPM compared to VEH
OFT.  Locomotor activity in the OFT was influenced by TREAT- (–54%; F1,48=34.618; p<0.001). There were no significant
MENT (F1,47=13.526; p<0.001), STRAIN (F1,47=22.344; TREATMENT×STRAIN interactions on distance travelled in
p<0.001) and a significant interaction between the EPM. WKY offspring spent less time in the open arms of the
TREATMENT×STRAIN (F3,47=6.463; p<0.05). WKY-VEH EPM compared to SD rats (–52%; F1,47=4.781; p<0.05); simi-
rats travelled less than SD-VEH rats (–82%; p<0.001). FLX larly, FLX-exposed offspring of both strains overall spent a
exposure reduced distance travelled in the OFT in SD offspring reduced amount of time exploring the open arms compared to
6 Journal of Psychopharmacology 00(0)

Figure 4.  (a) Distance travelled, (b) time spent in the open arms and (c) time spent in closed arms of the elevated plus maze (EPM). Data expressed
as mean centimetres (cm) or seconds (sec)+standard error of the mean (SEM); *p<0.05, ***p<0.001, #p<0.05; ##p<0.010 Wistar-Kyoto (WKY)
compared to Sprague-Dawley (SD) (overall strain effect).

(–28%; F1,26=22.028; p<0.001) and NR2B (–29%;


F1,23=15.113; p<0.001) in the PFC compared to offspring of
the SD strain, as well as reductions in the post-synaptic density
marker, PSD-95, a protein that anchors NMDA receptors
(–17%; F1,27=10.741; p<0.05). Furthermore, offspring of both
SD and WKY strains that were exposed to FLX expressed
reduced relative protein levels of NR1 (–12%; F1,25=4.515;
p<0.05), NR2A (–13%; F1,25=5.063; p<0.05) and PSD-95
(–17%; F1,27=8.389; p<0.01) compared to VEH offspring. In
the vHPC there were similar reductions in PSD-95 in WKY
offspring compared to SD offspring (–23%; F1,27=8.098;
p<0.01) as well as reductions following FLX exposure (–18%;
F1,27=5.213; p<0.05). There were no significant differences
between groups in any NMDA receptor subunits in the vHPC,
and no significant TREATMENT×STRAIN interactions
detected in either brain region (Figure 6).
Figure 5.  Time spent immobile in the forced swim test. Data Overall, the WKY strain showed reduced mGluR1 mono-
expressed as mean seconds (sec)+standard error of the mean (SEM); mer levels in the PFC compared to SD offspring (–41%;
***p<0.001; #p<0.05 Wistar-Kyoto (WKY) compared to Sprague- F1,27=17.826; p<0.01) and FLX-exposed offspring of both
Dawley (SD) (overall strain effect). strains collectively showed reductions in relative levels of
mGluR1 monomer in the PFC compared to VEH (–37%;
F1,27=6.886; p<0.05). There were no TREATMENT×STRAIN
VEH offspring (–81%; F1,47=18.025; p<0.001). There was no interactions. In the vHPC mGluR1 monomer expression was
STRAIN×TREATMENT interaction on time spent in open arms. influenced by TREATMENT (F1,26= 19.932, p<0.001),
There were significant TREATMENT (F1,48=55.379, p<0.001) STRAIN (F1,26=5.839, p<0.05) and a significant
and STRAIN (F1,48=22.994, p<0.001) effects on time spent in TREATMENT×STRAIN interaction (F3,26=6.933; p<0.05);
the closed arms and a significant STRAIN×TREATMENT inter- relative mGluR1 monomer levels in the vHPC were signifi-
action (F3,48=7.69, p=0.008). Post hoc analysis showed that WKY cantly reduced in WKY VEH offspring compared to SD VEH
VEH offspring spent an increased amount of time in the closed (–51%, p<0.05) and FLX exposure reduced mGluR1 mono-
arms compared to SD VEH offspring (+26%, p<0.001) and that mer expression in SD offspring (–70%, p<0.001) but not
FLX-exposed SD and WKY offspring spent significantly WKY offspring. There were no significant effects of treat-
increased time in the closed arms of the EPM compared to their ment, strain or interaction between the factors, on mGluR1
respective SD (+91%, p<0.001) and WKY (+25%, p<0.05) dimer levels in the PFC or vHPC (Figure 7).
VEH control groups (Figure 4). In both the PFC and vHPC, there were significant effects of
STRAIN (PFC: F1,25=16.908, p<0.001; vHPC: F1,27=11.440,
FST. Overall, offspring of the WKY strain spent more time p<0.01) and TREATMENT (PFC: F1,25=11.116, p<0.01;
immobile in the FST compared to SD offspring (+308%; vHPC: F1,27=40.567, p<0.001) on mGluR5 monomer levels
F1,44=45.2; p<0.05). FLX-exposed offspring of both strains also and significant interactions (PFC: F3,25=5.608, p<0.05; vHPC:
showed an increased amount of time spent immobile compared F3,27 =11.693, p<0.01). Post hoc analyses revealed that WKY
to VEH (+27%; F1,44=6.404; p<0.001). There were no signifi- VEH offspring express reduced mGluR5 monomer levels com-
cant TREATMENT × STRAIN interactions (Figure 5). pared to SD VEH (PFC: p<0.01; –58%; vHPC: p<0.001;
–52%) and that FLX-exposed SD offspring express reduced
Immunoblot. Adolescent offspring of the WKY strain had mGluR5 monomer levels compared to SD VEH in both brain
reduced levels of NR1 (–15%; F1,25=8.359; p<0.05), NR2A regions (PFC: p<0.01; –50%; vHPC: p<0.001; –75%), an
Millard et al.

Figure 6.  Relative levels of N-methyl-D-aspartate (NMDA) receptor subunits NR1, NR2A, NR2B and scaffolding protein post-synaptic density 95 (PSD-95) in the prefrontal cortex (PFC) ((a)–(d)) and
ventral hippocampus (vHPC) ((e)–(h)) at PN42. Data expressed as mean values+standard error of the mean (SEM). *p<0.05, ***p<0.001, #p<0.05; ###p<0.001 Wistar-Kyoto (WKY) compared to
Sprague-Dawley (SD) (overall strain effect).
7
8 Journal of Psychopharmacology 00(0)

Figure 7.  Relative levels of metabotropic glutamate receptor subtype 1 (mGluR1) monomer and dimeric forms in the prefrontal cortex (PFC) ((a) and
(b)) and ventral hippocampus (vHPC) ((c) and (d)) at post-natal day (PN) 42. Data expressed as mean values+standard error of the mean (SEM).
*p<0.05, ***p<0.001; ##p<0.01 Wistar-Kyoto (WKY) compared to Sprague-Dawley (SD) (overall strain effect). KDa: kilodalton.

Figure 8.  Relative levels of metabotropic glutamate receptor subtype 5 (mGluR5) monomer and dimeric forms, and scaffolding protein Homer1bc in the prefrontal
cortex (PFC) ((a)–(c)) and ventral hippocampus (vHPC) ((f)–(h)) at post-natal day (PN) 42. Data expressed as mean values+standard error of the mean (SEM).
*p<0.05, ***p<0.001; #p<0.05, ##p<0.010 Wistar-Kyoto (WKY) compared to Sprague-Dawley (SD) (overall strain effect). KDa: kilodalton.

effect that was not seen in FLX exposed WKY offspring. Discussion
mGluR5 dimers in the PFC were not affected by treatment,
strain or interaction effects (p>0.05); in the vHPC however, There is growing concern regarding the consequences of maternal
there was a significant effect of TREATMENT, with FLX- SSRI treatment on offspring neurodevelopment. Animal models,
such as that used in the present study design, allow us to distin-
exposed offspring overall showing increased levels of mGluR5
guish the effects of maternal SSRI treatment on offspring com-
dimers in the vHPC (+53%; F1,27=5.102; p<0.05) (Figure 8).
pared to, or in conjunction with, the underlying effects of maternal
In line with this increase, FLX-exposed offspring had signifi-
depression. Overall, our findings show that male adolescent off-
cantly increased vHPC levels of Homer1b/c, a scaffolding pro- spring of the WKY strain display increased anxiety- and depres-
tein that increases cell surface expression of mGluR5 (Mao sive-like behaviours compared to offspring of the SD strain.
et al., 2005), compared to VEH offspring (+9.6%: F1,25=5.233; Furthermore, this study showed for the first time that maternal
p<0.05). No other significant effects of TREATMENT, FLX treatment exacerbated the anxiety- and depressive-like phe-
STRAIN or interactions were detected for Homer1b/c levels in notype observed in the WKY offspring and induced these behav-
the PFC or vHPC (Figure 8). iours in the SD offspring. These FLX-induced behavioural changes
Millard et al. 9

were accompanied by altered protein expression of glutamatergic treatment between the two studies (paroxetine vs fluoxetine) or the
receptors and associated structural proteins in the PFC and vHPC ages of offspring examined (PN75 vs PN37). Several studies have
of both SD and WKY offspring. These novel findings suggest that utilised models of maternal stress (intense light exposure, restraint,
maternal FLX treatment exacerbates effects of the underlying confinement and other stressors) during various periods of gestation
maternal depression on offspring behaviour, which may be medi- coupled with FLX exposure (Boulle et al., 2016a, 2016b; Kiryanova
ated through alterations in the glutamatergic system. et al., 2016; Rayen et al., 2011). Consistent with our findings in
adolescent offspring, it has been reported that prenatal stress (gesta-
tional day (GD)15–21) coupled with postnatal FLX treatment
Maternal FLX treatment reduces offspring (post-partum day 1 to weaning) increases immobility time in the
body weight FST in male, as well as female, adolescent offspring, however does
not change OFT measures (Rayen et al., 2011). While there is a lack
Maternal FLX treatment was associated with reduced bodyweight of further investigation of adolescent offspring, one study in adult
of juvenile offspring at PN7 and PN14 in both SD and WKY rats. offspring reported increased immobility time during the FST in
While this finding is consistent with several previous studies in female (but not male) offspring born to FLX-treated (post-partum
FLX treated healthy rodents (for review see Hutchison et al., day 1 to weaning) and maternally stressed dams (GD15–21), com-
2018), this is the first report of offspring body weight followed pared to offspring of untreated, stressed dams (Boulle et al., 2016a,
throughout early developmental time periods in a rodent model of 2016b), suggesting possible sex effects; they however reported no
innate depression. In the clinic it is well reported that maternal significant effects for either sex on anxiety-like measures in the
FLX exposure can cause low birth weight (Olivier et al., 2013), OFT or Elevated Zero Maze in the adult offspring. Collectively,
however these studies are often complicated by confounders such these findings suggest that maternal SSRI treatment in models of
as maternal depression and anxiety, which are also risk factors for maternal depression or stress can influence offspring anxiety-like
perinatal complications such as low birth weight (Bansil et al., and depressive-like behaviour, especially in adolescent offspring
2010; Davalos et al., 2012; Field et al., 2004). In the clinic, there and that the sex and age of offspring can influence these effects.
are no investigations into offspring body weight beyond the early Indeed, given these observations, an emphasis on the differences
childhood years (Hutchison et al., 2018); we report in the present between innate and environmental models of depression, and the
study that the effect of FLX exposure on body weight extends to influence of offspring sex and age should be taken into considera-
adolescence in male offspring, but only in the SD strain, suggest- tion for further studies investigating the effects of maternal SSRI
ing no long-term effects of perinatal FLX exposure in the depres- treatment on offspring.
sion model. In support of these findings, Gemmel et al. (2018) The findings of increased immobility in the FST observed in
recently reported that maternal FLX treatment in a model of pre- the present study need to be taken with consideration given the
gestational stress reduces offspring body weight at pre-adoles- absence of additional behavioural tests assessing other facets of
cence but this does not extend to adulthood, similar to our findings depressive-like phenotypes (e.g. anhedonia in the sucrose prefer-
which indicate no long-term effect on body weight. ence test). It has been argued that immobility in the FST repre-
sents a switch from active to passive behaviour in the face of an
aversive stressor, and represents a behavioural adaption rather
Perinatal FLX exposure exacerbates the than depressive-like behaviours (Molendijk and de Kloet, 2015).
anxiety-like and depressive-like phenotype in Given that a decrease in locomotor activity as the result of mater-
male adolescent WKY offspring and induces nal FLX exposure was noted in the OFT and EPM, it is possible
an anxiety-like and depressive-like phenotype that the increased immobility time in FST could represent a
in male SD offspring ‘false-positive’ (Slattery and Cryan, 2012). Despite this, other
studies investigating developmental SSRI exposure on offspring
Maternal FLX treatment caused significant increases in anxiety- behaviour have noted increased immobility times in the FST in
like and depressive-like behaviour in male adolescent offspring of the presence and absence of changes in locomotor activity in the
both SD and WKY strains in the OFT, EPM and FST. While dis- OFT/EPM (for review see, Millard et al., 2017), suggesting that
tance travelled and time spent in the centre arena during the OFT developmental SSRI exposure induces changes in immobility
did not reach statistical significance in the FLX exposed WKY off- time in the FST, independent of changes in locomotor activity. To
spring, it should be noted that the WKY offspring were already further define the behavioural phenotype, future studies should
showing low measures in these parameters making it difficult to assess behaviours related to neuropsychiatric and neurodevelop-
detect further reductions. These findings suggest that perinatal FLX mental disorders in a wider battery of tests. It must also be
exposure in a model of depression exacerbates the anxiety-like and acknowledged that the effects of maternal FLX treatment on dam
depressive-like phenotype at adolescence. To our knowledge there behaviour were not assessed in the present study. Given that
is only one other study modelling maternal SSRI treatment in a maternal behaviour and care are known to influence offspring
rodent model showing an innate depressive phenotype. Glover et al. development and behaviour (Ahmadiyeh et al., 2004; Curley and
(2015) showed that paroxetine-exposed adult (PN75) male off- Champagne, 2016), this should be considered in future investiga-
spring born of SD dams exhibiting increased anxiety-like pheno- tions. Previous work in models of maternal stress have shown
types (termed bred low responders (bLRs)) had significantly greater that treatment with FLX at the same dose as the present study,
immobility times in the FST, similar to our finding, but report no does not influence maternal care behaviours (Gemmel et al.,
effect of paroxetine exposure on time spent in the open arms of the 2018) but does improve anxiety-like behaviours in the dam
EPM in the bLR offspring, in contrast to our finding. This contrast (Pawluski et al., 2012). There is no evidence to suggest that FLX
may be related to the difference in rat strains used, the different drug treatment in models of depression further exacerbate maternal
10 Journal of Psychopharmacology 00(0)

depressive- or anxiety-like behaviours, thus it is unlikely that the 2007), with glutamate signalling and BDNF production intri-
behaviours observed in the FLX exposed offspring in the present cately linked (Mattson, 2008). The BDNF signalling system is
study are a result of changes in maternal behaviour. crucial for normal neurodevelopment and ongoing regulation of
synaptic plasticity (Martinowich and Lu, 2007). Studies have
shown that mRNA expression of BDNF and its receptor,
Perinatal FLX exposure alters NMDA and group I Tropomyosin receptor kinase B (TrkB), are differentially altered
metabotropic glutamate receptors, as well as the in models of prenatal stress±postnatal exposure to FLX (Boulle
key scaffolding proteins, PSD-95 and Homer1b/c, et al., 2016a, 2016b), and that negative correlations exist between
in the cortex and ventral hippocampus of male immobility time in the FST and hippocampal BDNF mRNA
adolescent SD and WKY rats (Boulle et al., 2016b). Given our current findings of reduced
NMDA receptor subunits and PSD-95 in exposed offspring,
Whilst preclinical studies clearly implicate the NMDA receptor increased immobility time in the FST and the link between SSRIs
and associated scaffolding protein PSD-95 in the mechanism of and BDNF, it would be of interest for future investigations of
SSRI action (and to a lesser extent group 1 mGluRs) (Boyer perinatal SSRI±maternal depression, to determine whether glu-
et al., 1998; Musazzi et al., 2013; Park et al., 2014), the effects of tamatergic changes in the present study are associated with alter-
SSRI exposure during neurodevelopment on these proteins have ations in the BDNF/TrkB signalling system.
not previously been investigated. We report that maternal FLX Perturbations in glutamatergic signalling have repeatedly
exposure reduced the scaffolding protein, PSD-95, in both the been associated with anxiety- and depressive-like phenotypes.
PFC and vHPC of both rat strains. PSD-95 regulates NMDA Specifically, preclinical studies have shown reduced relative
receptor cell-surface expression and synaptic activity (Sheng, levels of NR1 and NR2A in the PFC and hippocampus (HPC) of
2001) by directly anchoring the NR2 subunits, and indirectly prenatally stressed offspring and concomitant increases in anxi-
coupling with mGluR1/5 receptor complexes (Tu et al., 1999). ety- and depressive-like behaviours (Sun et al., 2013a, 2013b).
Accordingly, NMDA receptor NR1 and NR2A subunits were also Changes in PSD-95 and related increases in anxiety-like behav-
reduced, but only in the PFC. In line with this, previous work iours have also previously been reported; anxiety-like behaviour
investigating the effects of chronic treatment with the SSRI, cit- in the novelty-induced hypophagia test and the OFT, induced by
alopram, in adult rodents, has reported reductions in mRNA lev- social isolation, has been shown to be accompanied by decreases
els encoding the NR1 (–7%) and NR2A (–35%) subunits in the in PSD-95, as well as NR1, in the frontal cortex and increases in
frontal cortex (Boyer et al., 1998). Interestingly these findings PSD-95 in the HPC and amygdala (Hermes et al., 2011; Zhang
suggest that the mechanism through which the effects of perinatal et al., 2012). Furthermore, in the present study we showed that
SSRI exposure influence NMDA receptor subunit expression in the WKY strain, which has a prominent anxiety- and depressive-
the PFC at adolescence, parallel those that occur in chronic use in like phenotype, displayed reduced glutamatergic markers
adults. FLX exposure also reduced mGluR1 and mGluR5 mono- including NR1, NR2A, NR2B and PSD-95, predominately in the
mers in both the PFC and vHPC, however these changes were PFC. Similar observations have been reported in postmortem
primarily in the SD offspring only, highlighting the potential con- brains of those suffering from depression (Feyissa et al., 2009).
sequences of maternal SSRI treatment in a healthy model and the Despite the evidence for altered glutamatergic markers in clini-
importance of using relevant models of maternal psychopathol- cal cases of MDD, these had not been previously measured in
ogy. In light of these findings, it is tempting to speculate that the the WKY strain. These findings add further support to the clini-
paralleled reductions of PSD-95 and NMDA receptor subunits in cal relevance and validity of the WKY strain as an innate depres-
the PFC, could be indicative of overall reductions in post-synap- sion model. Interestingly, others have shown that functional
tic densities, the specialised sites of glutamatergic synapses that antagonists of the NMDA receptor cause an antidepressant
harbour key proteins responsible for glutamatergic signalling and behavioural response in a model of inescapable stress (tail sus-
transduction (Sheng, 2001). However, in the vHPC, whilst we pension test) (Trullas and Skolnick, 1990), suggesting that
also report reduced PSD-95 in FLX-exposed offspring, we NMDA hyperactivity could underpin depressive-like behav-
observed no change in NMDA receptor subunits and increased iours. Indeed, NMDA antagonists such as ketamine, have rapid
levels of mGluR5 dimers and the associated scaffolding protein, and profound antidepressant properties (Iadarola et al., 2015).
Homer1b/c (which positively regulates mGluR5 signalling (Lum The observation that the WKY strain shows reduced NMDA
et al., 2018)), two proteins also indirectly coupled to PSD-95 via subunit levels in the PFC suggests that they could be resistant to
complexes deep in the post-synaptic density (Sheng and Kim, the effects of NMDA antagonists. Interestingly, however, studies
2011) suggesting that SSRI exposure induces brain region-spe- have found the contrary, with WKY rats being extremely respon-
cific effects. sive to the effects of ketamine on immobility time in the FST
While to our knowledge there have been no prior investiga- (Browne and Lucki, 2013; Tizabi et al., 2012). In the PFC, it is
tions of the glutamatergic system in models of perinatal SSRI the NMDA receptors located on gamma-aminobutyric acid
exposure, one recent study reported no effect of maternal FLX (GABA)-ergic interneurons that are reportedly more susceptible
treatment (from GD10–PN21) in SD rats on PSD-95 immunore- to the effects of ketamine (Moghaddam and Javitt, 2012); it may
activity in the PFC of pre-adolescent and adult offspring (Gemmel be that the reduction in NMDA receptor subunits observed in the
et al., 2018); they did however show reduced PSD-95 in male present study is predominantly occurring on other neuronal pop-
offspring of dams exposed to pre-gestational stress only (Gemmel ulations, such as pyramidal cells, in which case antagonism of
et al., 2018). SSRIs are well known to mediate their effects in NMDA receptors on GABA-ergic interneurons would help
adults, at least in part, by downstream effects on brain derived restore excitatory/inhibitory balance. Further investigation into
neurotrophic factor (BDNF) signalling (Martinowich and Lu, how the changes in the present study reflect specific neuronal
Millard et al. 11

populations, or whether they extend to other brain regions will neurodevelopmental disorders including autism, ADHD and
be important to further understand the potential and mechanisms mood disorders, our findings warrant further investigation into
of novel antidepressant treatments. the clinical effects of maternal SSRI exposure. Future studies
Beyond the behavioural parameters regarding anxiety- and assessing the influence of sex and maternal SSRI exposure are
depressive-like phenotypes, others have also reported that altera- also needed to provide deeper insight into the potential differ-
tions in glutamatergic signalling are associated with changes in ences of exposure between the sexes. Incidence of maternal
cognitive ability. NR1Hypo mice (expressing ~5% of normal NR1 depression is on the rise, and untreated maternal depression can
levels) (Mohn et al., 1999) have been shown to display alterations have significant negative effects on the health of both the mother
in cognition, including impairments in short- and long-term mem- and child. Antidepressants play a critical role in minimising the
ory in spatial and non-spatial tasks (Barkus et al., 2012). risks associated with untreated maternal depression. However,
Furthermore, abnormalities in PSD-95 have consistently been we are just beginning to understand the potential effects that
associated with altered cognition (VanGuilder et al., 2011). Indeed, maternal SSRIs may cause, and future studies investigating ways
two studies investigating the effects of developmental SSRI expo- to minimise these effects will be critical to ensuring healthy preg-
sure on offspring behaviour have noted alterations in cognition nancies and offspring development.
evident through changes in time taken to reach the platform in the
Morris water maze: however, these have been in standard labora- Acknowledgements
tory rodents not modelling maternal depression (Bairy et al., 2006;
Sprowles et al., 2016). Our findings of reduced NMDA receptor The authors wish to thank Rebecca Webby, Marta Ramos and Connor
Mackay for assistance with the rodent studies.
subunits and PSD-95 warrant further investigation of cognitive
behaviours using appropriate models of maternal depression.
In addition to changes observed in NMDA receptor subunits Declaration of conflicting interests
and PSD-95, our findings also suggest that mGluR5 signalling is The author(s) declared no potential conflicts of interest with respect to
fundamentally altered in the vHPC following FLX exposure. the research, authorship, and/or publication of this article.
Although the exact mechanism underlying this finding is
unknown, the relationship between anxiety-like behaviours and
Funding
mGluR5 have been well documented (Terbeck et al., 2015).
Evidence from a large number of preclinical studies has estab- The author(s) disclosed receipt of the following financial support for the
lished profound anxiolytic effects of mGluR5 antagonists research, authorship and/or publication of this article: This study has been
conducted with the support of a University of Wollongong, Faculty of
(Swanson et al., 2005). For example, following 2-methyl-
Science, Medicine and Health Target Grant to KAN and the Australian
6(phenylethynyl)pyridine (MPEP) treatment, rodents display Government Research Training Program Scholarship awarded to SJM
reduced exploratory inhibition by spending more time in the and JSL. JSL is further supported by Australian Rotary Health, in the
open arms of the EPM (Pérez de la Mora et al., 2006; form of an Ian Scott Scholarship.
Tatarczyńska et al., 2001). Previous studies have also noted
robust anxiolytic effects of selective intra-amygdaloid (a region
known to play a critical role in anxiety and fear) injections of
ORCID iD
MPEP in different rodent models of anxiety (Pérez de la Mora Kelly A Newell https://orcid.org/0000-0002-1245-5585
et al., 2006). Considering the vHPC is highly interrelated both
structurally and physiologically with the amygdala and this cir- Supplementary material
cuit mediates anxiety-like behaviour (Fanselow and Dong,
Supplementary material for this article is available online.
2010; Sah et al., 2003), it is not surprising to see changes in
mGluR5 in the vHPC of FLX-exposed offspring displaying
increased anxiety-like behaviour. Collectively our findings sug- References
gest that perinatal SSRI exposure is associated with increased Abe H, Hidaka N, Kawagoe C, et al. (2007) Prenatal psychological stress
anxiety-like and depressive-like behaviours which may be medi- causes higher emotionality, depression-like behavior, and elevated
ated by altered glutamatergic signalling. activity in the hypothalamo-pituitary-adrenal axis. Neurosci Res 59:
145–151.
Ahmadiyeh N, Slone-Wilcoxon JL, Takahashi JS, et al. (2004) Maternal
Conclusion behavior modulates X-linked inheritance of behavioral coping in the
defensive burying test. Biol Psychiatry 55: 1069–1074.
Overall, our findings show that maternal FLX treatment in a clin- Ansorge MS, Morelli E and Gingrich JA (2008) Inhibition of serotonin
ically relevant rodent model of depression induces changes in but not norepinephrine transport during development produces
body weight, behaviour and neurochemical phenotypes in delayed, persistent perturbations of emotional behaviors in mice. J
exposed offspring. Specifically, we report reduced postnatal bod- Neurosci Off J Soc Neurosci 28: 199–207.
yweight as well as increased anxiety- and depressive-like behav- Ansorge MS, Zhou M, Lira A, et al. (2004) Early-life blockade of the
iours and alterations in cortical and hippocampal glutamatergic 5-HT transporter alters emotional behavior in adult mice. Science
306: 879–881.
markers in male adolescent offspring. How the changes observed
Arango V, Underwood MD, Boldrini M, et al. (2001) Serotonin 1A
in the present study reflect glutamatergic synapses and whether receptors, serotonin transporter binding and serotonin transporter
these changes are transient in nature or persist into adulthood will mRNA expression in the brainstem of depressed suicide victims.
be key to understanding the clinical implications of maternal Neuropsychopharmacology 25: 892–903.
SSRI exposure. Given that dysfunction of key glutamatergic Bairy KL, Madhyastha S, Ashok KP, et al. (2006) Developmental and behav-
receptors and associated proteins has been linked to several ioral consequences of prenatal fluoxetine. Pharmacology 79: 1–11.
12 Journal of Psychopharmacology 00(0)

Bansil P, Kuklina EV, Meikle SF, et al. (2010) Maternal and fetal outcomes determined by [(11)C]ABP688 PET and postmortem study. Am J
among women with depression. J Womens Health 19: 329–334. Psychiatry 168: 727–734.
Barkus C, Dawson LA, Sharp T, et al. (2012) GluN1 hypomorph mice du Bois TM, Huang X-F and Deng C (2008) Perinatal administration of
exhibit wide-ranging behavioral alterations. Genes Brain Behav 11: PCP alters adult behaviour in female Sprague–Dawley rats. Behav
342–351. Brain Res 188: 416–419.
Bonnin A, Torii M, Wang L, et al. (2007) Serotonin modulates the Dulawa SC, Holick KA, Gundersen B, et al. (2004) Effects of chronic
response of embryonic thalamocortical axons to netrin-1. Nat Neu- fluoxetine in animal models in anxiety and depression. Neuropsy-
rosci 10: 588–597. chopharmacology 29: 1321–1330.
Boukhris T, Sheehy O, Mottron L, et al. (2016) Antidepressant use dur- Dusi N, Barlati S, Vita A, et al. (2015) Brain structural effects of antide-
ing pregnancy and the risk of autism spectrum disorder in children. pressant treatment in major depression. Curr Neuropharmacol 13:
JAMA Pediatr 170: 117–124. 458–465.
Boulle F, Pawluski JL, Homberg JR, et al. (2016a) Prenatal stress and Edgar JM and Price DJ (2001) Radial migration in the cerebral cortex is
early-life exposure to fluoxetine have enduring effects on anxiety enhanced by signals from thalamus. Eur J Neurosci 13: 1745–1754.
and hippocampal BDNF gene expression in adult male offspring. Fanselow MS and Dong H-W (2010) Are the dorsal and ventral hippo-
Dev Psychobiol 58: 427–438. campus functionally distinct structures? Neuron 65: 7–19.
Boulle F, Pawluski JL, Homberg JR, et al. (2016b) Developmental fluox- Feyissa AM, Chandran A, Stockmeier CA, et al. (2009) Reduced levels
etine exposure increases behavioral despair and alters epigenetic of NR2A and NR2B subunits of NMDA receptor and PSD-95 in the
regulation of the hippocampal BDNF gene in adult female offspring. prefrontal cortex in major depression. Prog Neuropsychopharmacol
Horm Behav 80: 47–57. Biol Psychiatry 33: 70–75.
Boyer P-A, Skolnick P and Fossom LH (1998) Chronic administration of Field T, Diego M, Hernandez-Reif M, et al. (2004) Prenatal maternal bio-
imipramine and citalopram alters the expression of NMDA receptor chemsitry predicts neonatal biochemistry. Int J Neurosci 114: 933–945.
subunit mRNAs in mouse brain. J Mol Neurosci 10: 219–233. Figueroa R (2010) Use of antidepressants during pregnancy and risk of
Brandlistuen RE, Ystrom E, Eberhard-Gran M, et al. (2015) Behavioural attention-deficit/hyperactivity disorder in the offspring. J Dev Behav
effects of fetal antidepressant exposure in a Norwegian cohort of dis- Pediatr 31: 641–648.
cordant siblings. Int J Epidemiol 44: 1397–1407. Foldi CJ, Eyles DW, McGrath JJ, et al. (2011) The effects of breeding
Brandon NJ and McKay R (2015) The cellular target of antidepressants. protocol in C57BL/6J mice on adult offspring behaviour. PLoS One
Nat Neurosci 18: 1537–1538. 6: e18152.
Browne CA and Lucki I (2013) Antidepressant effects of ketamine: Mecha- Gemmel M, Kokras N, Dalla C, et al. (2018) Perinatal fluoxetine prevents
nisms underlying fast-acting novel antidepressants. Front Pharmacol 4: the effect of pre-gestational maternal stress on 5-HT in the PFC, but
161. maternal stress has enduring effects on mPFC synaptic structure in
Cabrera-Vera TM and Battaglia G (1998) Prenatal exposure to fluox- offspring. Neuropharmacology 128: 168–180.
etine (Prozac) produces site-specific and age-dependent alterations Gidaya NB, Lee BK, Burstyn I, et al. (2014) In utero exposure to selec-
in brain serotonin transporters in rat progeny: Evidence from autora- tive serotonin reuptake inhibitors and risk for autism spectrum disor-
diographic studies. J Pharmacol Exp Ther 286: 1474–1481. der. J Autism Dev Disord 44: 2558–2567.
Calvo N, Cecchi M, Kabbaj M, et al. (2011) Differential effects of social Glover ME, Pugh PC, Jackson NL, et al. (2015) Early-life exposure to
defeat in rats with high and low locomotor response to novelty. Neu- the SSRI paroxetine exacerbates depression-like behavior in anxiety/
roscience 183: 81–89. depression-prone rats. Neuroscience 284: 775–797.
Capello CF, Bourke CH, Ritchie JC, et al. (2011) Serotonin transporter Glover V (2014) Maternal depression, anxiety and stress during preg-
occupancy in rats exposed to serotonin reuptake inhibitors in utero or nancy and child outcome; what needs to be done. Best Pract Res Clin
via breast milk. J Pharmacol Exp Ther 339: 275–285. Obstet Gynaecol 28: 25–35.
Castagné V, Moser P, Roux S, et al. (2011) Rodent models of depression: Harrington RA, Lee L-C, Crum RM, et al. (2014) Prenatal SSRI use and
Forced swim and tail suspension behavioral despair tests in rats and offspring with autism spectrum disorder or developmental delay.
mice. Curr Protoc Neurosci, 55: 8.10A.1-8.10A.14. Pediatrics 2013–3406.
Clements CC, Castro VM, Blumenthal SR, et al. (2015) Prenatal antide- Hashimoto K, Sawa A and Iyo M (2007) Increased levels of glutamate in
pressant exposure is associated with risk for attention deficit-hyper- brains from patients with mood disorders. Biol Psychiatry Depress
activity disorder but not autism spectrum disorder in a large health 62: 1310–1316.
system. Mol Psychiatry 20: 727–734. Heikkinen T, Ekblad U and Laine K (2002) Transplacental transfer of
Commons KG, Cholanians AB, Babb JA, et al. (2017) The rodent forced citalopram, fluoxetine and their primary demethylated metabolites
swim test measures stress-coping strategy, not depression-like in isolated perfused human placenta. Int J Obstet Gynaecol 109:
behavior. ACS Chem Neurosci 8: 955–960. 1003–1008.
Cooper WO, Willy ME, Pont SJ, et al. (2007) Increasing use of antide- Hendrick V, Stowe ZN, Altshuler LL, et al. (2001) Fluoxetine and nor-
pressants in pregnancy. Am J Obstet Gynecol 196: 544.e1–544.e5. fluoxetine concentrations in nursing infants and breast milk. Biol
Croen LA, Grether JK, Yoshida CK, et al. (2011) Antidepressant use dur- Psychiatry 50: 775–782.
ing pregnancy and childhood autism spectrum disorders. Arch Gen Hermes G, Li N, Duman C, et al. (2011) Post-weaning chronic social
Psychiatry 68: 1104–1112. isolation produces profound behavioral dysregulation with decreases
Curley JP and Champagne FA (2016) Influence of maternal care on the in prefrontal cortex synaptic-associated protein expression in female
developing brain: Mechanisms, temporal dynamics and sensitive rats. Physiol Behav 104: 354–359.
periods. Front Neuroendocrinol 40: 52–66. Hiemke C and Härtter S (2000) Pharmacokinetics of selective serotonin
Davalos DB, Yadon CA and Tregellas HC. (2012) Untreated prenatal reuptake inhibitors. Pharmacol Ther 85: 11–28.
maternal depression and the potential risks to offspring: A review. Hutchison SM, Mâsse LC, Pawluski JL, et al. (2018) Perinatal selective
Arch Womens Ment Health 15: 1–14. serotonin reuptake inhibitor (SSRI) effects on body weight at birth
Deave T, Heron J, Evans J, et al. (2008) The impact of maternal depres- and beyond: A review of animal and human studies. Reprod Toxicol
sion in pregnancy on early child development. Int J Obstet Gynaecol 77: 109–121.
115: 1043–1051. Huybrechts KF, Palmsten K, Mogun H, et al. (2013) National trends in
Deschwanden A, Karolewicz B, Feyissa AM, et al. (2011) Reduced antidepressant medication treatment among publicly insured preg-
metabotropic glutamate receptor 5 density in major depression nant women. Gen Hosp Psychiatry 35: 265–271.
Millard et al. 13

Iadarola ND, Niciu MJ, Richards EM, et al. (2015) Ketamine and other Mattson MP (2008) Glutamate and neurotrophic factors in neuronal
N-methyl-D-aspartate receptor antagonists in the treatment of plasticity and disease. Annals of the New York Academy of Sciences
depression: A perspective review. Ther Adv Chronic Dis 6: 97–114. 1144: 97–112.
Jama A, Cecchi M, Calvo N, et al. (2008) Inter-individual differences McIlwain KL, Merriweather MY, Yuva-Paylor LA, et al. (2001) The use
in novelty-seeking behavior in rats predict differential responses to of behavioral test batteries: Effects of training history. Physiol Behav
desipramine in the forced swim test. Psychopharmacology (Berl.) 73: 705–717.
198: 333–340. Millard SJ, Weston-Green K and Newell KA (2017) The effects of
Karpova NN, Lindholm J, Pruunsild P, et al. (2009) Long-lasting behav- maternal antidepressant use on offspring behaviour and brain devel-
ioural and molecular alterations induced by early postnatal fluox- opment: Implications for risk of neurodevelopmental disorders. Neu-
etine exposure are restored by chronic fluoxetine treatment in adult rosci Biobehav Rev 80: 743–765.
mice. Eur Neuropsychopharmacol 19: 97–108. Mitchell AA, Gilboa SM, Werler MM, et al.; National Birth Defects Pre-
Kessler R (2003) Epidemiology of women and depression. J Affect Dis- vention Study (2011) Medication use during pregnancy, with par-
ord 74: 5–13. ticular focus on prescription drugs: 1976–2008. Am J Obstet Gynecol
Kim J, Riggs KW, Misri S, et al. (2006) Stereoselective disposition of 205: 51.e1–8.
fluoxetine and norfluoxetine during pregnancy and breast-feeding. Moghaddam B and Javitt D (2012) From revolution to evolution: the
Br J Clin Pharmacol 61: 155–163. glutamate hypothesis of schizophrenia and its implication for treat-
Kiryanova V, McAllister BB and Dyck RH (2013) Long-term outcomes ment. Neuropsychopharmacol Off Publ Am Coll Neuropsychophar-
of developmental exposure to fluoxetine: A review of the animal lit- macol 37: 4–15.
erature. Dev Neurosci 35: 437–439. Mohn AR, Gainetdinov RR, Caron MG, et al. (1999) Mice with reduced
Kiryanova V, Meunier SJ, Vecchiarelli HA, et al. (2016) Effects of NMDA receptor expression display behaviors related to schizophre-
maternal stress and perinatal fluoxetine exposure on behavioral out- nia. Cell 98: 427–436.
comes of adult male offspring. Neuroscience 320: 281–296. Molendijk ML and de Kloet ER (2015) Immobility in the forced swim
Koran LM, Cain JW, Dominguez RA, et al. (1996) Are fluoxetine plasma test is adaptive and does not reflect depression. Psychoneuroendo-
levels related to outcome in obsessive-compulsive disorder? Am J crinology 62: 389–391.
Psychiatry 153: 1450–1454. Morley-Fletcher S, Mairesse J, Soumier A, et al. (2011) Chronic agomel-
Lakhan SE, Caro M and Hadzimichalis N (2013) NMDA receptor activ- atine treatment corrects behavioral, cellular, and biochemical abnor-
ity in neuropsychiatric disorders. Front Psychiatry 4: 52. malities induced by prenatal stress in rats. Psychopharmacology
Lam MW, Young CJ and Mabury SA (2005) Aqueous photochemical (Berl.) 217: 301.
reaction kinetics and transformations of fluoxetine. Environ Sci Musazzi L, Treccani G, Mallei A, et al. (2013) The action of antidepres-
Technol 39: 513–522. sants on the glutamate system: Regulation of glutamate release and
Lavdas AA, Blue ME, Lincoln J, et al. (1997) Serotonin promotes the differ- glutamate receptors. Biol Psychiatry 73: 1180–1188.
entiation of glutamate neurons in organotypic slice cultures of the devel- Nam H, Clinton SM, Jackson NL, et al. (2014) Learned helplessness and
oping cerebral cortex. J Neurosci Off J Soc Neurosci 17: 7872–7880. social avoidance in the Wistar-Kyoto rat. Front Behav Neurosci 8: 109.
Lemos JC, Zhang G, Walsh T, et al. (2011) Stress-hyperresponsive WKY Oberlander TF, Gingrich JA and Ansorge MS (2009) Sustained neurobe-
rats demonstrate depressed dorsal raphe neuronal excitability and havioral effects of exposure to SSRI antidepressants during devel-
dysregulated CRF-mediated responses. Neuropsychopharmacology opment: Molecular to clinical evidence. Clin Pharmacol Ther 86:
36: 721–734. 672–677.
López-Figueroa AL, Norton CS, López-Figueroa MO, et al. (2004) Sero- Olivier JDA, Åkerud H, Kaihola H, et al. (2013) The effects of maternal
tonin 5-HT1A, 5-HT1B, and 5-HT2A receptor mRNA expression in depression and maternal selective serotonin reuptake inhibitor expo-
subjects with major depression, bipolar disorder, and schizophrenia. sure on offspring. Front Cell Neurosci 7: 73.
Biol Psychiatry 55: 225–233. Olivier JDA, Vallès A, Heesch F van, et al. (2011) Fluoxetine admin-
López-Rubalcava C and Lucki I (2000) Strain differences in the behav- istration to pregnant rats increases anxiety-related behavior in the
ioral effects of antidepressant drugs in the rat forced swimming test. offspring. Psychopharmacology (Berl.) 217: 419–432.
Neuropsychopharmacology 22: 191–199. Pardon M-C, Gould GG, Garcia A, et al. (2002) Stress reactivity of the brain
Lum JS, Fernandez F, Matosin N, et al. (2016) Neurodevelopmental expres- noradrenergic system in three rat strains differing in their neuroendo-
sion profile of dimeric and monomeric group 1 mGluRs: Relevance to crine and behavioral responses to stress: Implications for susceptibility
schizophrenia pathogenesis and treatment. Sci Rep 6: srep34391. to stress-related neuropsychiatric disorders. Neuroscience 115: 229–242.
Lum JS, Millard SJ, Frank E, et al. (2018) Chronic adolescent CDPPB Pare WP and Tejani-Butt SM (1996) Effect of stress on the behavior and
treatment alters short-term, but not long-term, glutamatergic receptor 5-HT system in Sprague-Dawley and Wistar Kyoto rat strains. Integr
expression. Neurochem Res 43: 1683–1691. Physiol Behav Sci 31: 112.
Malm H, Brown AS, Gissler M, et al. (2016) Gestational exposure to Park SW, Lee JG, Seo MK, et al. (2014) Differential effects of antide-
selective serotonin reuptake inhibitors and offspring psychiatric dis- pressant drugs on mTOR signalling in rat hippocampal neurons. Int
orders: A National Register-Based Study. J Am Acad Child Adolesc J Neuropsychopharmacol 17: 1831–1846.
Psychiatry 55: 359–366. Pawluski JL, Brain UM, Underhill CM, et al. (2012) Prenatal SSRI expo-
Mao L, Yang L, Tang Q, et al. (2005) The scaffold protein Homer1b/c sure alters neonatal corticosteroid binding globulin, infant cortisol
links metabotropic glutamate receptor 5 to extracellular signal-regu- levels, and emerging HPA function. Psychoneuroendocrinology 37:
lated protein kinase cascades in neurons. J Neurosci Off J Soc Neu- 1019–1028.
rosci 25: 2741–2752. Paxinos G and Watson C (2007) The rat brain in stereotaxic coordinates.
Marcus SM and Heringhausen JE (2009) Depression in childbearing (6th Edition). San Diego: Academic Press.
women: When depression complicates pregnancy. Prim Care Clin Paylor R, Spencer CM, Yuva-Paylor LA, et al. (2006) The use of behav-
Off Pract 36: 151–165. ioral test batteries, II: Effect of test interval. Physiol Behav 87: 95–102.
Martinowich K and Lu B (2007) Interaction between BDNF and serotonin: Pérez de la Mora M, Lara-García D, Jacobsen KX, et al. (2006) Anxio-
Role in mood disorders. Neuropsychopharmacology 33: 73–83. lytic-like effects of the selective metabotropic glutamate receptor 5
Matosin N, Frank E, Deng C, et al. (2013) Metabotropic glutamate recep- antagonist MPEP after its intra-amygdaloid microinjection in three
tor 5 binding and protein expression in schizophrenia and following different non-conditioned rat models of anxiety. Eur J Neurosci 23:
antipsychotic drug treatment. Schizophr Res 146: 170–176. 2749–2759.
14 Journal of Psychopharmacology 00(0)

Prut L and Belzung C (2003) The open field as a paradigm to measure Swanson CJ, Bures M, Johnson MP, et al. (2005) Metabotropic glutamate
the effects of drugs on anxiety-like behaviors: A review. Eur J Phar- receptors as novel targets for anxiety and stress disorders. Nature
macol 463: 3–33. Reviews Drug Discovery 4: 131–144.
Ramtekkar UP, Reiersen AM, Todorov AA, et al. (2010) Sex and age Tatarczyńska E, Kłodzińska A, Chojnacka-Wójcik E, et al. (2001) Poten-
differences in attention-deficit/hyperactivity disorder symptoms and tial anxiolytic- and antidepressant-like effects of MPEP, a potent,
diagnoses: Implications for DSM-V and ICD-11. J Am Acad Child selective and systemically active mGlu5 receptor antagonist. Br J
Adolesc Psychiatry 49: 217–28.e1–3. Pharmacol 132: 1423–1430.
Rayen I, van den Hove DL, Prickaerts J, et al. (2011) Fluoxetine during Terbeck S, Akkus F, Chesterman LP, et al. (2015) The role of metabo-
development reverses the effects of prenatal stress on depressive-like tropic glutamate receptor 5 in the pathogenesis of mood disorders
behavior and hippocampal neurogenesis in adolescence. PLoS One 6: and addiction: Combining preclinical evidence with human Positron
e24003. Emission Tomography (PET) studies. Front Neurosci 9.
Redei E, Pare WP, Aird F, et al. (1994) Strain differences in hypotha- Thompson MR, Li KM, Clemens KJ, et al. (2004) Chronic fluoxetine
lamic-pituitary-adrenal activity and stress ulcer. Am J Physiol Regul treatment partly attenuates the long-term anxiety and depressive
Integr Comp Physiol 266: R353–R360. symptoms induced by MDMA (‘Ecstasy’) in rats. Neuropsychophar-
Rittenhouse PA, López-Rubalcava C, Stanwood GD, et al. (2002) Ampli- macol Off Publ Am Coll Neuropsychopharmacol 29: 694–704.
fied behavioral and endocrine responses to forced swim stress in the Tizabi Y, Bhatti BH, Manaye KF, et al. (2012) Antidepressant-like
Wistar–Kyoto rat. Psychoneuroendocrinology 27: 303–318. effects of low ketamine dose is associated with increased hippocam-
Rodriguez-Porcel F, Green D, Khatri N, et al. (2011) Neonatal exposure pal AMPA/NMDA receptor density ratio in female Wistar-Kyoto
of rats to antidepressants affects behavioral reactions to novelty and rats. Neuroscience 213: 72–80.
social interactions in a manner analogous to autistic spectrum disor- Trullas R and Skolnick P (1990) Functional antagonists at the NMDA
ders. Anat Rec Adv Integr Anat Evol Biol 294: 1726–1735. receptor complex exhibit antidepressant actions. Eur J Pharmacol
Ronald A, Pennell CE and Whitehouse AJO (2011) Prenatal maternal 185: 1–10.
stress associated with ADHD and autistic traits in early childhood. Tu JC, Xiao B, Naisbitt S, et al. (1999) Coupling of mGluR/Homer and
Front Psychol 1: 223. PSD-95 complexes by the Shank family of postsynaptic density pro-
Sah P, Faber ESL, Lopez De Armentia M, et al. (2003) The amygdaloid teins. Neuron 23: 583–592.
complex: Anatomy and physiology. Physiol Rev 83: 803–834. Turner KM and Burne THJ (2014) Comprehensive behavioural analysis
Scholl JL, Renner KJ, Forster GL, et al. (2010) Central monoamine lev- of Long Evans and Sprague-Dawley rats reveals differential effects
els differ between rat strains used in studies of depressive behavior. of housing conditions on tests relevant to neuropsychiatric disorders.
Brain Res 1355: 41–51. PLoS One 9: e93411.
Semple BD, Blomgren K, Gimlin K, et al. (2013) Brain development VanGuilder HD, Farley JA, Yan H, et al. (2011) Hippocampal dysregula-
in rodents and humans: Identifying benchmarks of maturation and tion of synaptic plasticity-associated proteins with age-related cogni-
vulnerability to injury across species. Prog Neurobiol 106–107: tive decline. Neurobiol Dis 43: 201–212.
1–16. Vitalis T and Parnavelas JG (2003) The role of serotonin in early cortical
Sheng M (2001) The postsynaptic NMDA-receptor—PSD-95 signal- development. Dev Neurosci 25: 245–256.
ing complex in excitatory synapses of the brain. J Cell Sci 114: Walf AA and Frye CA (2007) The use of the elevated plus maze as
1251. an assay of anxiety-related behavior in rodents. Nat Protoc 2:
Sheng M and Kim E (2011) The postsynaptic organization of synapses. 322–328.
Cold Spring Harb Perspect Biol 3: a005678–a005678. Werling DM and Geschwind DH (2013) Sex differences in autism spec-
Slattery DA and Cryan JF (2012) Using the rat forced swim test to trum disorders. Curr Opin Neurol 26: 146–153.
assess antidepressant-like activity in rodents. Nat Protoc 7: 1009– Will CC, Aird F and Redei EE (2003) Selectively bred Wistar-Kyoto rats:
1014. An animal model of depression and hyper-responsiveness to antide-
Sprowles JLN, Hufgard JR, Gutierrez A, et al. (2016) Perinatal exposure pressants. Mol Psychiatry 8: 925–932.
to the selective serotonin reuptake inhibitor citalopram alters spatial World Health Organization (2017) Depression and Other Common
learning and memory, anxiety, depression, and startle in Sprague- Mental Disorders: Global Health Estimates. Geneva: World Health
Dawley rats. Int J Dev Neurosci 54: 39–52. Organization.
Sun H, Guan L, Zhu Z, et al. (2013a) Reduced levels of NR1 and NR2A Yuan H, Low C-M, Moody OA, et al. (2015) Ionotropic GABA and
with depression-like behavior in different brain regions in prenatally glutamate receptor mutations and human neurologic diseases. Mol
stressed juvenile offspring. PLoS One 8: e81775. Pharmacol 88: 203.
Sun H, Jia N, Guan L, et al. (2013b) Involvement of NR1, NR2A differ- Zhang Y, Zu X, Luo W, et al. (2012) Social isolation produces anxiety-
ent expression in brain regions in anxiety-like behavior of prenatally like behaviors and changes PSD-95 levels in the forebrain. Neurosci
stressed offspring. Behav Brain Res 257: 1–7. Lett 514: 27–30.

You might also like