You are on page 1of 8

Psychoneuroendocrinology 68 (2016) 202–209

Contents lists available at ScienceDirect

Psychoneuroendocrinology
journal homepage: www.elsevier.com/locate/psyneuen

A diet high in fat and sugar reverses anxiety-like behaviour induced


by limited nesting in male rats: Impacts on hippocampal markers
Jayanthi Maniam, Christopher P. Antoniadis, Vivian Le, Margaret J. Morris ∗
Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, New South Wales, Australia

a r t i c l e i n f o a b s t r a c t

Article history: Stress exposure during early development is known to produce long-term mental health deficits. Stress
Received 2 December 2015 promotes poor lifestyle choices such as poor diet. Early life adversity and diets high in fat and sugar
Received in revised form 1 February 2016 (HFHS) are known to affect anxiety and memory. However additive effects of HFHS and stress during
Accepted 9 March 2016
early development are less explored. Here, we examined whether early life stress (ELS) simulated by
limited nesting (LN) induces anxiety-like behaviour and cognitive deficits that are modulated by HFHS
Keywords:
diet. We examined key hippocampal markers involved in anxiety and cognition, testing the hypothesis
Early life stress
that post-weaning HFHS following ELS would ameliorate anxiety-like behaviour but worsen memory
Anxiety-like behaviour
Akt3
and associated hippocampal changes. Sprague-Dawley rats were exposed to LN, postnatal days 2–9, and
Cognition at weaning, male siblings were given unlimited access to chow or HFHS resulting in (Con-Chow, Con-
Glucocorticoid receptor HFHS, LN-Chow, LN-HFHS, n = 11–15/group). Anxiety-like behaviour was assessed by Elevated Plus Maze
Glycogen synthase kinase (EPM) at 10 weeks and spatial and object recognition tested at 11 weeks of age. Rats were culled at 13
High-fat and sugar diet weeks. Hippocampal mRNA expression was measured using TaqMan® Array Micro Fluidic cards (Life
Hippocampus Technologies). As expected HFHS diet increased body weight; LN and control rats had similar weights
Limited nesting at 13 weeks, energy intake was also similar across groups. LN-Chow rats showed increased anxiety-like
behaviour relative to control rats, but this was reversed by HFHS diet. Spatial and object recognition mem-
ory were unaltered by LN exposure or consumption of HFHS diet. Hippocampal glucocorticoid receptor
(GR) protein was not affected by LN exposure in chow rats, but was increased by 45% in HFHS rats rel-
ative to controls. Hippocampal genes involved in plasticity and mood regulation, GSK␣ and GSK␤ were
affected, with reductions in GSK␤ under both diet conditions, and reduced GSK␣ only in LN-HFHS versus
Con-HFHS. Interestingly, HFHS diet and LN exposure independently reduced expression of Akt3 mRNA,
a key gene involved post-natal brain development. In summary, while an energy rich diet ameliorated
anxiety-like behaviour induced by LN exposure, it significantly altered key genes that are essential for
hippocampal development.
© 2016 Elsevier Ltd. All rights reserved.

1. Introduction utilised animal models, predominantly in rats, to simulate ELS in


order to investigate its effects. A variety of methods have been
Early life stress (ELS) is prevalent in today’s society, being mani- employed to induce ELS, including maternal separation and depri-
fested in a number of different ways such as child abuse, childhood vation (Maniam et al., 2014). A more recently developed model,
trauma and neglect. ELS has been shown to have significant adverse limited nesting (LN), involving dams being supplied with inade-
effects lasting into adulthood, altering the brain and behavioural quate nesting materials, has been proven to be robust in mimicking
aspects such as mood and anxiety, as well as increasing the risk the behavioural patterns of maternal neglect in humans (Ivy et al.,
of psychiatric disorders (Bremne and Vermetten, 2001; Heim and 2008; Prusator and Greenwood-Van Meerveld, 2015).
Nemeroff, 1999; Sanchez et al., 2001). These effects on emotions The modern diet is rich in foods high in both fats and sugars,
have been demonstrated to be a result of changes to the hippocam- and people commonly crave these palatable ‘comfort foods’ when
pus and amygdala (Aust et al., 2014). Further research efforts have under stress. With diet known to be an important factor for its
effects on the brain and behaviour, the combined effects of ELS
along with a poor quality diet high in fat and sugar needs to be
considered as a possible outcome for those who undergo ELS. The
∗ Corresponding author. detrimental effects of a high fat diet on metabolism are numerous
E-mail address: m.morris@unsw.edu.au (M.J. Morris).

http://dx.doi.org/10.1016/j.psyneuen.2016.03.007
0306-4530/© 2016 Elsevier Ltd. All rights reserved.
J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209 203

and well-known (Nakamura and Terauchi, 2013), but its effects, were returned to normal bedding at PND 10. The control group had
and the mechanisms underpinning these effects, on the brain and normal bedding throughout.
mental health are less clearly understood.
A high fat diet has been linked to adverse effects on cognition, 2.2. Post-weaning diet
learning and memory, although there are a number of proposed
potential underlying mechanisms (Cordner and Tamashiro, 2015; At PND 21, the pups were weaned and housed 3–4 rats per cage.
Freeman et al., 2014). Diets high in fats and sugars have been linked Male offspring were assigned to either standard laboratory chow
to stress and chronic stressful conditions have previously been (11 kJ/g, energy 12% fat, 21% protein, 65% carbohydrate, Gordon’s
shown to increase the intake of palatable foods (Teegarden and Specialty Stockfeeds, NSW, Australia) or standard laboratory chow
Bale, 2008). This bidirectional association has been demonstrated and palatable HFHS, SF03-020 (20 kJ/g, energy 43% fat, 17% pro-
in response to both acute and chronic stress in rodents (Maniam and tein, 40% sucrose supplied by Specialty Feeds, Glen Forrest, WA,
Morris, 2012). However, limited work has examined the effects of Australia). Chow, water, and HFHS were available ad libitum. This
ELS on post-weaning high fat diet intake. While conflicting data has generated four groups: Con-Chow; Con-HFHS; LN-Chow; and LN-
been obtained regarding overall energy intake following ELS, it has HFHS, comprising, n = 10–15/group. Rats were weighed at weaning
been demonstrated that adult female rats had an increased prefer- and at regular intervals throughout the study. Food intake over 24 h
ence for palatable “comfort foods” high in sucrose and fats when was recorded by weighing food and bottles before presentation to
exposed to ELS by limiting access to nesting materials (Machado the rats and again after 24 h. Energy intake per cage was measured
et al., 2013). Additional negative effects of an unhealthy diet on the and average intake was calculated assuming equal intake for each
brain and mental health that have also been established include rat.
modifications to the inflammatory response, leading to a worse
outcome following stroke (Maysami et al., 2015). Of particular 2.3. Elevated plus maze (EPM)
interest to our study, it has also been shown to induce neurochem-
ical changes in the hippocampus (Krishna et al., 2015). A high fat The rats undertook the EPM as previously described (Maniam
diet has been demonstrated to ameliorate anxiety-like behaviour and Morris, 2010a, b) during the light phase at 10 weeks of age.
in rats that had previously been exposed to ELS (induced by mater- Opaque perspex was used to construct a plus-shaped maze con-
nal separation) (Maniam and Morris, 2010a, b), but its effects on sisting of 2 opposite open arms (50 cm × 10 cm) and 2 closed arms
hippocampal gene expression are unclear. (50 cm × 10 cm with 40 cm walls). Once the rat was placed in the
The aims of this paper were to firstly investigate whether HFHS arena the experimenter left the room. Behaviour was analysed
consumption may moderate anxiety-like behaviour and cognitive using footage obtained from directly above the arena using a video-
impairment induced by ELS in the form of LN, and secondly to camera. Macropod’s OD log software was used to score the data,
explore whether the diet has any additional effects on hippocam- both by the experimenter and a second observer unaware of the
pal markers involved in pathways known to be affected by ELS. allocation of the groups. The time and entries of the rats in each
Therefore, we hypothesized that ELS (via limited nesting) would of the open and closed arms was recorded over a 10-min period.
induce anxiety-like behaviour and cognitive deficits in offspring, Data are presented as percentage time in open arms, percentage
and given that foods rich in sugar and fat are reported to reduce entries into open arms and number of entries into open arms. We
stress levels in humans (Adam and Epel, 2007) and rodents (la also recorded number of rearing events.
Fleur et al., 2005), that voluntary intake of HFHS would ame-
liorate ELS induced anxiety-like behaviour and cognitive deficit. 2.4. Novel objects and place recognition task
Further, we hypothesized that chronic consumption of HFHS would
worsen hippocampal markers that regulate pathways (neuroplas- This task was conducted as previously described (Beilharz et al.,
ticity, neurogenesis, stress response, inflammatory, mitochondrial 2014). Rats were given 10 min on 2 consecutive days to acclimatize
biogenesis) that are known to regulate a range of behaviours con- themselves to the empty arena (a black acrylic open field arena
sistently associated with ELS. (60 cm × 60 cm × 60 cm)), and behavioural assessment started on
the following day. The rats from each diet group were split into 2
equal groups, and on the first day one half undertook the object task
2. Materials and methods whilst the remainder were assessed on the place recognition task.
The groups then switched tasks on the following day of testing.
2.1. Animals At the commencement of every test, 2 identical objects were
placed into two of the middle four sections. The novel objects were
All animal procedures were approved by the Animal Care and commercial products that varied in size and texture (e.g., mugs,
Ethics Committee of UNSW Australia. Male and female Sprague- tins). The familiarisation phase of the test involved positioning the
Dawley rats (Animal Resource Centre, Perth, WA, Australia) were rat in the middle of the arena and giving it 5 min to explore. At
maintained in a temperature controlled (23 ◦ C) colony room on a the conclusion of this phase, the rat was removed for 5 min (the
12 h light/dark cycle (lights on at 0700) with ad libitum access to retention phase). For the test phase of the object task, the objects
standard laboratory chow and water. Mating was carried out in were then set up in the same formation as during the familiarisa-
house to generate litters. After mating females were housed singly tion phase, except one of the objects was replaced with a novel one.
and littered normally. In the test phase of the place recognition task, the same objects that
Litters comprising 9–15 pups were included and standardized had been used for the familiarisation phase were put back into the
to 12 pups per litter on postnatal day (PND) 1 using pups from arena, except one of them was placed in one of the corner quad-
dams littering on the same day to minimize alterations in mater- rants instead of its original position. These test phases were 3 min
nal behaviour and pup nutrition. Litters were housed with the dam in duration. Each rat was only allowed exposure to the same object
in polypropylene cages (20 cm × 32 cm × 19 cm) on wood shavings for one trial, and testing took place at the same time of day for
with a metal lid. From post-natal days 2–9 cages of the ELS group each repeated trial. The order and locations of the objects were
had a metal grid floor inserted that allowed waste to be excluded, counterbalanced across trials and between the rats.
and given only half a sheet of paper towel as nesting material. Behaviour was analysed using footage obtained from directly
Both groups were left undisturbed between PND 2–9, then ELS rats above the arena using a video-camera. Exploration was defined
204 J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209

as the rat extending its neck to position its head within 2 cm of 2.6. Western blot
the object followed by moving its vibrissae around the object.
Macropod’s OD log software was used to score the data, both by Whole tissue lysates from powdered hippocampus were pre-
the experimenter and a second observer unaware of the alloca- pared using a Precellys 24 homogenizer (Sapphire Bioscience,
tion of the groups. The Exploration Ratio was used to represent Sydney, NSW, Australia) in radioimmunoprecipitation assay buffer
the data—this was calculated by dividing the time spent explor- (RIPA) buffer containing 65 mM Tris (pH 7.4), 150 mM NaCl, 5 mM
ing the novel object by the total time spent exploring both objects EDTA, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% sodium
(tnovel /(tnovel + told )). dodecyl sulfate (SDS), 10% glycerol, 1 ␮g/ml aprotinin, 1 ␮g/ml
leupeptin, 10 mM sodium fluoride, 1 mM sodium orthovanadate
(Na3 VO4 ) and 1 mM phenylmethanesulfonylfluoride or phenyl-
methylsulfonyl fluoride (PMSF) according to methods described
2.5. Reverse-transcription PCR (RT-PCR) elsewhere (Hoy et al., 2007). The hippocampus homogenate was
then incubated for 2 h at 4 ◦ C and then centrifuged at 10 000g
RNA was extracted using Tri-reagent (Sigma-Aldrich, Sydney, for 15 min to remove insoluble material. Protein concentration
NSW, Australia) and treated with DNase I (Invitrogen, Mel- was determined by Bradford protein assay (Bio-Rad) using bovine
bourne, VIC, Australia) to remove any contaminating genomic serum albumin (BSA) as standard. Proteins were resolved by
DNA and stored at −80 ◦ C. RNA concentration was determined sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-
using a Biospec-nano spectrophotometer (Shimadzu, Sydney, NSW, PAGE) electrophoresis on a 4–15% polyacrylamide gel (BioRad) and
Australia). RNA was reverse transcribed to cDNA using Omnis- transferred to a polyvinylidene difluoride (PVDF) membrane. Non-
cript Reverse Transcription kit (Qiagen, Melbourne, VIC, Australia) specific binding sites were blocked in Tris-buffered saline with 4%
and stored at −20 ◦ C. We performed RT-PCR using TaqMan® BSA and 0.1% Tween-20 for 1 h at room temperature. Membranes
Array Micro Fluidic Cards (Life Technologies) which were pre- were incubated on a roller at 4 ◦ C overnight with glucocorticoid
customised with Taqman probes (Applied Biosystems, Melbourne, receptor antibody (Cell Signaling; 1:1000) followed by anti-rabbit
VIC, Australia) for nuclear factor of kappa light polypeptide gene IgG horse radish peroxidase (HRP)-linked antibody (Cell Signal-
enhancer in B-cells 1(Nfkb1-Rn01399583 m1), toll-like receptor 3 ing; 1:10 000); and anti-alpha-tubulin (Millipore; 1:1000) followed
(Tlr3-Rn01488472 g1), toll-like receptor 4 (Tlr4-Rn00569848 m1), by anti-mouse IgG HRP-linked antibody (Cell Signaling; 1:10 000).
tumor necrosis factor (Tnf-Rn99999017 m1), S100 calcium After washing in Tris-buffered saline with 0.1% Tween-20, mem-
binding protein B (S100b-Rn04219408 m1), nuclear respira- branes were visualised with enhanced chemiluminescence (ECL,
tory factor 1(Nrf1-Rn01455958 m1), peroxisome proliferator- Millipore) with images captured using Image Quant LAS-4000
activated receptor gamma, coactivator 1 alpha (Ppargc1a- (GE Healthcare, Sydney, NSW, Australia). Protein expression lev-
Rn00580241 m1), sirtuin 3 (Sirt3-Rn01501410 m1), superoxide els were quantified by densitometry using ImageJ 1.47v software
dismutase 1, soluble (Sod1-Rn00566938 m1), v-akt murine (NIH, Maryland, USA).
thymoma viral oncogene homolog 1 (Akt1-Rn00583646 m1),
v-akt murine thymoma viral oncogene homolog 2 (Akt2-
Rn00567290 m1), v-akt murine thymoma viral oncogene homolog 3. Results
3 (protein kinase B, gamma) (Akt3-Rn00442194 m1), brain-
derived neurotrophic factor (Bdnf-Rn01484924 m1), neuritin 3.1. The effects of LN exposure and HFHS diet on body weight and
1(Nrn1-Rn00584304 m1), cAMP responsive element binding food intake
protein 1(Creb1-Rn00578829 g1), reelin (Reln-Rn00589609 m1),
caspase 3 (Casp3-Rn00563902 m1), glycogen synthase kinase Body weight was measured at termination (Con-Chow:
3 alpha (Gsk3a-Rn00569232 m1), glycogen synthase kinase 3 357.8 ± 7.83 g LN-Chow: 330.11 ± 8.62 g, Con-HFHS: 478.1 ± 17.3 g,
beta (Gsk3b-Rn00583429 m1), glycogen synthase kinase 3 alpha LN-HFHS: 425.0 ± 14.0 g). In both chow and HFHS fed groups, the
(Gsk3a-Rn00569232 m1), gamma-aminobutyric acid (GABA-A) body weight of LN rats were not significantly different from con-
receptor, subunit alpha 2 (Gabra2-Rn01413643 m1), homer trol rats as evidenced by no significant interaction between LN
homolog 1 (Drosophila) (Homer1-Rn00581785 m1), nuclear and HFHS (F(1,53) = 1.104, p = 0.298). However, as expected a main
receptor subfamily 3, group C, member 1/glucocorticoid recep- effect of HFHS diet was observed (F(1,53) = 79.26, p < 0.0001). HFHS
tor (Nr3c1-Rn00561369 m1), 5-hydroxytryptamine (serotonin) fed rats were heavier than their chow counterparts.
receptor 1A (Htr1a-Rn00561409 s1), 5-hydroxytryptamine (sero- In line with THE body weight data, LN exposure had no effect on
tonin) receptor 2A (Htr2a-Rn00568473 m1), mitogen activated energy intake in the chow [Con-Chow (2536 ± 97 kJ/rat), LN-Chow
protein kinase 1 (Mapk1-Rn00587719 m1), mitogen-activated (2448 ± 68 kJ/rat)], or HFHS fed rats [Con-HFHS (3290 ± 107 kJ/rat),
protein kinase 8 (Mapk8-Rn01453358 m1), mitogen activated LN-HFHS (3013 ± 88 kJ/rat)] as evidenced by no significant interac-
protein kinase 3 (Mapk3-Rn00820922 g1), mitogen activated pro- tion between LN and HFHS diet [LN × HFHS, F(1, 11) = 1.10, p > 0.05].
tein kinase 14 (Mapk14-Rn00578842 m1), signal transducer and However, as expected, HFHS increased energy intake across groups
activator of transcription 6 (Stat6-Rn01505881 m1), signal trans- as evidenced by a main effect of HFHS (F(1, 11) = 53.49, p < 0.0001).
ducer and activator of transcription 3 (Stat3-Rn00562562 m1),
signal transducer and activator of transcription 5A (Stat5a- 3.2. The effects of LN exposure and HFHS diet on anxiety-like
Rn00567011 m1). behaviour
We used 1.5 ␮g of cDNA. 100 ml of cDNA/Mastermix (Advanced
Fast Mastermix, Life Technologies) mixture was pipetted into the Anxiety-like behaviour was assessed at 10 weeks of age using
micro fluid cards, after centrifugation the cards were sealed and EPM. Increased percentage of open arm entries or time spent in
RT-PCR was performed. The geometric mean of two housekeepers, open arm is indicative of reduced anxiety-like behaviour (Korte
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activa- and De Boer, 2003; Rodgers and Dalvi, 1997). Here, in those con-
tion protein (Ywhaz) and hypoxanthine phosphoribosyltransferase suming chow, LN rats made approximately 20% less open arm
1 (Hprt1) was used as reference. Analysis was performed using the entries compared to control rats (post-hoc p < 0.05 LN versus con-
CT method and data expressed relative to a calibrator sample trol, Fig. 1A). A significant interaction was observed between LN
of all groups. and diet; LN × Diet, F(1,29) = 4.21, p = 0.04. Interestingly, provision
J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209 205

Fig. 1. Anxiety-like behaviour assessed by elevated plus maze at 10 weeks of age. (A) Percentage open arm entries, (B) percentage time in open, (C) number of open arm
entries, (D) number of rearing events. Results are expressed as mean ± S.E.M; n = 7–10, two-way ANOVA followed by LSD. Post-hoc analysis was performed when a significant
interaction between diet and LN was present.
†<0.05 versus control rats consuming the same diet (LN effect).
**p < 0.01 versus rats consuming chow (diet effect)

of a palatable HFHS diet to LN rats almost doubled the percentage 3.4. The effects of LN exposure and HFHS diet on hippocampal GR
of open arm entries versus chow fed LN rats (p < 0.05, Fig. 1A). protein levels
While the percentage of time spent in open arms (Fig. 1B)
appears to show a similar trend as the percentage of open The hippocampal GR plays a key role in HPA axis activity reg-
arm entries, we observed no statistically significant interaction ulation, and here we examined GR protein expression by western
between LN exposure and diet consumed (two-way ANOVA anal- blot. A significant interaction was observed between LN and diet;
ysis, (F(1,29) = 1.34, p > 0.05), see Fig. 1B). In addition, there were LN × Diet, F(1,43) = 5.14, p = 0.03. LN exposure had no effect on hip-
no significant differences in two-way ANOVA analysis of general pocampal GR protein expression in the chow fed groups. However,
activity, as evidenced by the number of entries into open arms in those consuming HFHS, LN exposure increased hippocampal GR
(p > 0.05, see Fig. 1C) and rearing (p > 0.05, see Fig. 1D). While LN- mRNA expression by about 45 percent relative to control rats (post
HFHS appears to have reduced rearing activity, two-way ANOVA hoc p < 0.05, Fig. 3). Further, LN-HFHS rats had an approximate
analysis showed no significant interaction between LN exposure 50% increase in hippocampal GR versus their chow counterparts
and diet (F (1, 29) = 0.1578, P = 0.67). (LN-HFHS versus LN-chow p < 0.01, Fig. 3).

3.5. The effects of LN exposure and HFHS diet on hippocampal


genes

3.3. The effects of LN exposure and HFHS diet on spatial and A targeted array of hippocampal genes associated with neuro-
object recognition memory plasticity, neurogenesis, stress response, serotonin, inflammatory,
mitochondrial biogenesis including MAP kinases and STATs were
Tests of spatial and object recognition memory were conducted examined. All of these processes are critical in brain function and
using novel object and place recognition tasks. We observed no behavioural regulation of anxiety, depression, stress response and
statistical differences across groups in terms of times of exploration, cognition. Several genes showed a main effect of LN exposure. An
on object (F(1,35) = 3.16, p > 0.05, Fig. 2A) and place (F(1,35) = 0.09, interaction was observed between LN and diet for GSK isoforms
p > 0.05, Fig. 2C) testing. Neither LN exposure nor HFHS diet had (LN × HSHS interaction, GSKB␣ F(1,28) = 5.14, p = 0.031 and GSK3␤
any effect on performance of the object recognition task (LN × Diet F(1,27) = 4.74, p = 0.039). In those consuming chow, LN exposure
F(1,35) = 1.001, p = 0.32, Fig. 2), however, there was a main effect significantly reduced hippocampal GSK␤ mRNA expression by
of LN exposure (F(1,35) = 4.23, p = 0.047). The average exploration about 30% (p < 0.01, Table 1), but had no effect on GSKB␣ mRNA
ratio in LN rats consuming chow appears to be slightly higher than expression. In those rats consuming HFHS, LN exposure markedly
control rats consuming the same diet. reduced the expression of both GSKB␣ and GSK3␤ compared to
In terms of the place recognition task, while there appears to control rats (p < 0.01, Table 1). Akt3 was affected by both LN expo-
be a trend for HFHS effects in the control condition, the inter- sure and HFHS (LN × HFHS interaction F(1,28) = 18.83, p = 0.0002).
action between LN exposure and diet did not reach significance LN exposure alone significantly reduced the expression of Akt3
(F(1,35) = 3.15, p = 0.08). mRNA by about 34% in the chow fed rats (p < 0.01, Table 1), and had
206 J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209

Fig. 2. Object and place recognition memory was assessed with novel object and place tasks at 11 weeks. Object task exploration time (A) and ratio (B) and place task
exploration time (C) and ratio (D) are expressed as mean ± SEM, with n = 9–10 rats per group. Data were analysed by two-way ANOVA followed by LSD.

our previous observations using maternal separation as a model of


ELS (Maniam and Morris, 2010a, b). Here we observed markedly
upregulated GR protein levels in response to the combination of
LN and HFHS exposure, which may be associated with the rever-
sal of anxiety-like behaviour by the diet. Surprisingly, contrary to
the findings of others, in our hands, object and spatial recognition
memory were not affected by LN exposure or chronic consumption
of HFHS diet. Interestingly, of the array of hippocampal genes that
are involved in pathways that regulate anxiety, depression and cog-
nition measured, only GSKB␣, GSKB␤ and Akt3 were significantly
affected by LN exposure.
Given the percentages of entries and time spent in each arm con-
stitute an index of anxiety (Korte and De Boer, 2003; Rodgers and
Dalvi, 1997), here in our hands, we concluded that LN-Chow rats
exhibited increased anxiety-like behaviour when compared to Con-
Chow rats. Others using elevated plus maze and different batteries
of anxiety-like behaviour tests such as open-field, light-dark box
Fig. 3. Glucocorticoid receptor protein measured at 13 weeks of age. Data are
and novelty-induced hypophagia have consistently demonstrated
expressed as mean ± S.E.M; two-way ANOVA followed by LSD, n = 11–12/group. that LN exposure increases anxiety-like behaviour in pups. Thus
Post-hoc analysis performed when a significant interaction between diet and LN an LN induced anxiety-like behaviour phenotype was observed in
present. mice (Malter Cohen et al., 2013; Wang et al., 2012) and rats (Dalle
† p < 0.05 versus control rats consuming the same diet (LN effect).
Molle et al., 2012; Machado et al., 2013). This is suggestive of LN
** p < 0.01 versus rats consuming chow (diet effect)
being a robust model of ELS across species. It is important to note
that, the main effect of diet was significant in both measures of time
no effect of LN in the HFHS fed rats. Interestingly, HFHS diet alone spent in open arm and percentage open arm entries which suggests
produced similar effects as LN exposure on Akt3 mRNA expression that diet has a robust effect on anxiety-like behaviour across control
in control rats, where consumption of HFHS reduced the expres- and LN groups.
sion of Akt3 mRNA by about 20% (p < 0.01, Table 1). There was no Changes in maternal behaviour have been consistently shown
additive effect when LN and HFHS diet were combined. to impact offsprings’ neurodevelopment including hypothalamic-
pituitary-adrenal axis activity which governs stress response
4. Discussion activity (Caldji et al., 2000; Ziabreva et al., 2003, 2000). Work from
our lab and others has previously demonstrated that ELS in the form
Our finding that post-weaning HFHS ameliorated the increased of maternal separation significantly reduces the quality of maternal
anxiety-like behaviour induced by ELS in the form of LN is the first care provided by the dams to their pups and this was associated
demonstration of a post-weaning diet interaction with ELS in this with behavioural outcomes such as increased stress responsive-
paradigm. Several of the findings of this study are consistent with ness, anxiety- and depression-like behaviours (Macri et al., 2008;
J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209 207

Table 1
The effects of LN exposure and post-weaning HFHS diet on the expression of hippocampal genes. Results are expressed as mean ± S.E.M; data were analysed by two-way
ANOVA followed by LSD, n = 7–9 rats/group. mRNA expression was measured using the microfluidic card platform (Life Technologies).

Significance

Con-Chow LN-Chow Con-HFHS LN-HFHS Interaction Diet LN

Inflammatory genes
nfkb1 0.94 ± 0.07 0.82 ± 0.09 1.00 ± 0.12 0.74 ± 0.05 ns ns 0.035
Tlr3 1.15 ± 0.30 1.09 ± 0.27 1.28 ± 0.37 0.92 ± 0.12 ns ns ns
Tlr4 1.09 ± 0.20 0.98 ± 0.08 0.97 ± 0.10 0.97 ± 0.17 ns ns ns
TNF 1.49 ± 0.44 2.22 ± 0.37 1.86 ± 0.30 1.57 ± 0.30 ns ns ns
s100b 1.02 ± 0.08 0.76 ± 0.07 1.06 ± 0.10 0.90 ± 0.04 ns ns 0.009

Mitochondrial biogenesis and energy metabolism


Nrf1 1.01 ± 0.05 0.76 ± 0.08 1.01 ± 0.10 0.62 ± 0.05 ns ns p < 0.0001
ppargc1a 1.02 ± 0.07 1.13 ± 0.17 1.15 ± 0.12 0.80 ± 0.06 ns ns ns
SIRT3 1.02 ± 0.07 0.99 ± 0.09 1.18 ± 0.12 0.99 ± 0.06 ns ns ns
SOD1 1.03 ± 0.09 0.77 ± 0.05 1.16 ± 0.07 0.72 ± 0.05 ns ns p < 0.0001
Akt1 1.00 ± 0.03 1.07 ± 0.12 0.98 ± 0.04 1.02 ± 0.04 ns ns ns
Akt2 1.01 ± 0.06 0.89 ± 0.06 1.17 ± 0.13 0.90 ± 0.03 ns ns p = 0.012
Akt3 1.01 ± 0.06 0.66 ± 0.04† 0.80 ± 0.03* 0.82 ± 0.04 0.0002 ns 0.001

Neurogenesis and plasticity


BDNF 1.02 ± 0.07 1.01 ± 0.06 1.33 ± 0.14 0.98 ± 0.07 ns ns ns
nrn1 0.90 ± 0.05 0.86 ± 0.09 1.01 ± 0.08 0.69 ± 0.05 ns ns 0.019
creb1 1.02 ± 0.08 1.06 ± 0.09 1.17 ± 0.11 0.96 ± 0.04 ns ns ns
reln 1.04 ± 0.11 0.80 ± 0.04 1.14 ± 0.12 0.65 ± 0.05 ns ns 0.0002
Casp3 1.03 ± 0.09 0.79 ± 0.08 0.83 ± 0.09 0.71 ± 0.07 ns ns 0.044
GSK3␤ 0.95 ± 0.07 0.87 ± 0.07† 1.19 ± 0.08 0.78 ± 0.09† 0.039 ns 0.004
GSK3␣ 1.02 ± 0.06 0.92 ± 0.06 1.22 ± 0.07 0.86 ± 0.04† 0.031 ns 0.001
Gabra2 1.02 ± 0.06 1.10 ± 0.09 1.16 ± 0.13 1.12 ± 0.06 ns ns ns

Stress response
GR 1.03 ± 0.09 0.94 ± 0.06 1.02 ± 0.06 0.80 ± 0.07 ns ns p = 0.043
Homer1 1.01 ± 0.06 0.97 ± 0.07 1.01 ± 0.06 0.79 ± 0.04 ns ns 0.028

Serotonin
Htr1a 1.01 ± 0.05 1.05 ± 0.06 1.14 ± 0.09 1.05 ± 0.06 ns ns ns
Htr2b 1.05 ± 0.11 0.90 ± 0.08 0.98 ± 0.07 0.70 ± 0.08 ns ns 0.021

MAP kinases
Mapk1 1.02 ± 0.08 1.04 ± 0.17 1.36 ± 0.30 0.96 ± 0.13 ns ns ns
Mapk3 1.02 ± 0.07 0.85 ± 0.10 1.01 ± 0.06 0.83 ± 0.05 ns ns 0.022
Mapk14 1.02 ± 0.07 0.73 ± 0.05 1.05 ± 0.04 0.69 ± 0.03 ns ns p < 0.0001
Mapk8 1.01 ± 0.06 0.79 ± 0.06 0.95 ± 0.09 0.71 ± 0.05 ns ns 0.001

Signal transducer and activator of transcription


STAT6 0.96 ± 0.03 0.79 ± 0.06 1.01 ± 0.06 0.80 ± 0.06 ns ns 0.002
STAT3 1.01 ± 0.04 0.969 ± 0.08 1.03 ± 0.07 0.79 ± 0.04 ns ns 0.032
STAT5a 1.03 ± 0.10 0.965 ± 0.08 1.19 ± 0.10 0.95 ± 0.10 ns ns ns

p < 0.01 versus control rats consuming the same diet (LN effect).
*
p < 0.01 versus rats consuming chow (diet effect).

Maniam and Morris, 2010a, b; Meaney et al., 1991, 1988). There- behaviour across the LN and diet groups, the overall exploration
fore, one possible reason for the changes in anxiety-like behaviour time was not different in the novel object tasks, so we do not think
induced by the LN model in this study could be alterations in mater- this influenced the outcomes of the cognitive measures.
nal behaviour. The lab that developed the LN material as a model While we observed an increase in anxiety-like behaviour in LN
of ELS has systematically demonstrated that dams exposed to LN rats, GR protein was not affected. At the level of mRNA expres-
regularly leave the pups in the nesting area and so the duration of sion, we observed a main effect of LN exposure but no significant
each nurturing behaviour (licking, grooming and feeding) is short interaction with HFHS diet. We previously showed that ELS induced
and variable (Ivy et al., 2008). This altered maternal behaviour by maternal separation reduced the expression of hippocampal GR
has been proposed to be linked with altered stress response as mRNA (Maniam and Morris, 2010a, b) and a similar effect was
evidenced by changes in the hypothalamo-pituitary-adrenal axis observed in female rats exposed to LN versus controls (Maniam
activity (Avishai-Eliner et al., 2001; Brunson et al., 2005). & Morris unpublished data). In the current study, we provide the
In terms of cognitive measurements, we observed no effects first evidence that chronic consumption of HFHS altered hippocam-
of ELS in the form of LN on spatial and object recognition tasks. pal GR protein levels in those exposed to LN relative to their chow
However, other studies using similar ELS paradigms showed LN counterparts. The marked increase in GR protein levels in LN rats
exposure produced spatial memory deficits (Ivy et al., 2010; consuming HFHS diet versus LN rats consuming chow appears to
Naninck et al., 2015). The order of behavioural tests conducted be in line with the reversal of anxiety-like behaviour. In support,
may have had an influence on the cognitive measures in our study. we previously showed that cafeteria style diet normalised the ELS
Even though the cognitive task was conducted one week after the induced reduction in hippocampal GR mRNA expression (Maniam
anxiety-like behaviour test, we cannot exclude the possibility that and Morris, 2010a, b). There are several other studies that simu-
this test may have impacted the cognitive measures. Future work lated ELS with LN exposure (Cui et al., 2006; Naninck et al., 2015),
may need to consider increasing the gap between anxiety and cog- however, no data on hippocampal GR were reported. In summary,
nitive testing and counterbalancing the order of the anxiety and across two different ELS paradigms, we showed that HFHS diet can
cognitive tests. Although we observed differences in anxiety-like interact with ELS on hippocampal GR.
208 J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209

We targeted several genes that mediate important pathways independently affect expression of hippocampal genes that are vital
known to be affected by early life adversity such as plasticity & for mood and brain development. There is evidence in human stud-
neurogenesis, stress response, serotonin, inflammatory and down- ies that people prefer diets that are rich in sugar and fat as a stress
stream mediators such as MAP kinases and STATs. However, only 3 coping strategy (Adam and Epel, 2007). However, given that such
genes were significantly affected by LN and diet interaction. Firstly, diet produces deleterious effects at a molecular level, it is impor-
we observed a marked decrease in hippocampal GSK3a and GSK3b tant for the public to be educated on the adverse effects of diets rich
expression of LN rats in those consuming HFHS diet relative to in fat and sugar. Moreover, we simulated childhood trauma via LN
control rats. Serine/threonine protein kinase synthase is known exposure which is thought to mimic conditions of human mater-
to mediate many brain functions including mood and neuroge- nal neglect (Ivy et al., 2008) and so if similar processes are at play
nesis. Antipsychotics and antidepressants such as fluoxetine are in the human, other interventions to offset the adverse effects on
shown to directly or indirectly block the action of GSK3␣/␤, and brain and behaviour are crucial.
selective inhibition of GSK3 produces antidepressant behaviour in
the forced swim test (Ackermann et al., 2010). Transgenic mice Conflict of interest
expressing elevated GSK3 activity display memory deficits, reduced
brain size and alterations in mood behaviour and social interac- The authors declare that the research was conducted in the
tions (Hernandez et al., 2002; Polter et al., 2010; Prickaerts et al., absence of any commercial or financial relationships that could be
2006; Spittaels et al., 2002). Several other studies have outlined construed as a potential conflict of interest.
the role of GSK3 in mediating the effects of antidepressant drugs
(Duman and Aghajanian, 2014; Li et al., 2014). Antidepressant drugs
Role of the funding source
have also been shown to increase neurogenesis through recruit-
ment of new neurons (Surget et al., 2011). Here, a reduction of
Funding for this study was provided by Diabetes Australia
expression in hippocampal GSK3␣/␤ mRNA was observed in chow
Research Trust (DART). The DART had no further role in the study
and HFHS fed rats exposed to ELS, but their memory task perfor-
design, in the collection, analysis and interpretation of data; in
mance was not affected by LN exposure or by the combination with
writing of the report; and in the decision to submit the paper for
HFHS diet. However, the reduced expression of GSK3␣/␤ mRNA
publication.
by the HFHS diet in LN rats is supportive of the amelioration of
anxiety-like behaviour in LN-HFHS rats. It is important to note that
altering mRNA expression may not reflect the pathological condi- Author contributions
tion in which GSK3 activity is dysregulated in the application of
mood stabilisers and antidepressants (Polter et al., 2010, 2012). Designed the experiments: JM and MJM. Performed the exper-
However, this is the first evidence of GSK3 mRNA alterations fol- iments: JM, CPA, and VL. Analysed the data: JM, CPA and VL. All
lowing ELS, particularly when combined with post-weaning HFHS authors contributed to the writing of the manuscript.
diet, and provides impetus for future work to further examine the
role of GSK3 activity under conditions of early life insults and later Acknowledgment
diet manipulation.
Another gene that was affected by LN exposure and chronic con- This work was supported by project funding from the Diabetes
sumption of HFHS is Akt3. Hippocampal expression of Akt3 mRNA Australia Research Trust awarded to MJM.
was reduced by about 40% by LN exposure in chow fed rats only
and the HFHS diet only reduced its expression in control rats; hence References
providing evidence that an unhealthy diet produces similar effects
to LN exposure on the expression of Akt3 mRNA. There appears Ackermann, T.F., Kempe, D.S., Lang, F., Lang, U.E., 2010. Hyperactivity and
enhanced curiosity of mice expressing PKB/SGK-resistant glycogen synthase
to be limited work examining the impact of HFHS on Akt related
kinase-3 (GSK-3). Cell. Physiol. Biochem. 25, 775–786.
pathways in the brain. Akt plays an important role in brain devel- Adam, T.C., Epel, E.S., 2007. Stress, eating, and the reward system. Physiol. Behav.
opment, synaptic plasticity and neurodegeneration (Easton et al., 91, 449–458.
Aust, S., Stasch, J., Jentschke, S., Alkan Hartwig, E., Koelsch, S., Heuser, I., Bajbouj,
2005; Tschopp et al., 2005; Wang et al., 2003). Among all the iso-
M., 2014. Differential effects of early life stress on hippocampus and amygdala
forms, Akt3 is predominantly expressed in the brain and represents volume as a function of emotional abilities. Hippocampus 24, 1094–1101.
50% of the total Akt in the cerebral cortex and hippocampus (Easton Avishai-Eliner, S., Gilles, E.E., Eghbal-Ahmadi, M., Bar-El, Y., Baram, T.Z., 2001.
et al., 2005). Moreover, Akt3 is involved in postnatal brain devel- Altered regulation of gene and protein expression of
hypothalamic-pituitary-adrenal axis components in an immature rat model of
opment (Tschopp et al., 2005). Of concern is our finding that the chronic stress. J. Neuroendocrinol. 13, 799–807.
chronic consumption of HFHS led to a similar degree of reduc- Beilharz, J.E., Maniam, J., Morris, M.J., 2014. Short exposure to a diet rich in both fat
tion in Akt3 mRNA as LN exposure. If a similar process occurs in and sugar or sugar alone impairs place, but not object recognition memory in
rats. Brain Behav. Immun. 37, 134–141.
the human, the consumption of HFHS may increase the risk for Bremne, J.D., Vermetten, E., 2001. Stress and development: behavioral and
psychopathologies. biological consequences. Dev. Psychopathol. 13, 473–489.
In summary, LN exposure produced long-lasting effects in adult Brunson, K.L., Kramar, E., Lin, B., Chen, Y., Colgin, L.L., Yanagihara, T.K., Lynch, G.,
Baram, T.Z., 2005. Mechanisms of late-onset cognitive decline after early-life
behaviour as evidenced by increased anxiety-like behaviour in stress. J. Neurosci. 25, 9328–9338.
those consuming chow and LN was associated with a marked reduc- Caldji, C., Diorio, J., Meaney, M.J., 2000. Variations in maternal care in infancy
tion in genes associated with plasticity and mood regulation which regulate the development of stress reactivity. Biol. Psychiatry 48, 1164–1174.
Cordner, Z.A., Tamashiro, K.L., 2015. Effects of high-fat diet exposure on learning &
are also a main target of antidepressant drugs. A diet rich in fat and
memory. Physiol. Behav. 152, 363–371.
sugar ameliorated the increased anxiety-like behaviour induced by Cui, M., Yang, Y., Yang, J., Zhang, J., Han, H., Ma, W., Li, H., Mao, R., Xu, L., Hao, W.,
LN exposure suggesting it may have “therapeutic value” (Machado Cao, J., 2006. Enriched environment experience overcomes the memory
deficits and depressive-like behavior induced by early life stress. Neurosci.
et al., 2013) but this was at the expense of adverse effects on the
Lett. 404, 208–212.
expression of hippocampal genes related to mood regulation and Dalle Molle, R., Portella, A.K., Goldani, M.Z., Kapczinski, F.P., Leistner-Segal, S.,
neurogenesis; GSK␣ and GSK3␤. Moreover, we observed the HFHS Salum, G.A., Manfro, G.G., Silveira, P.P., 2012. Associations between parenting
diet produced equivalent adverse effects on hippocampal Akt3 gene behavior and anxiety in a rodent model and a clinical sample: relationship to
peripheral BDNF levels. Transl. Psychiatry 2, e195.
that may have implications for brain development. Hence, we pro- Duman, R.S., Aghajanian, G.K., 2014. Neurobiology of rapid acting antidepressants:
vide evidence for the first time that LN exposure and HFHS diet role of BDNF and GSK-3beta. Neuropsychopharmacology 39, 233.
J. Maniam et al. / Psychoneuroendocrinology 68 (2016) 202–209 209

Easton, R.M., Cho, H., Roovers, K., Shineman, D.W., Mizrahi, M., Forman, M.S., Lee, Meaney, M.J., Aitken, D.H., Bhatnagar, S., Sapolsky, R.M., 1991. Postnatal handling
V.M., Szabolcs, M., de Jong, R., Oltersdorf, T., Ludwig, T., Efstratiadis, A., attenuates certain neuroendocrine, anatomical, and cognitive dysfunctions
Birnbaum, M.J., 2005. Role for Akt3/protein kinase Bgamma in attainment of associated with aging in female rats. Neurobiol. Aging 12, 31–38.
normal brain size. Mol. Cell. Biol. 25, 1869–1878. Nakamura, A., Terauchi, Y., 2013. Lessons from mouse models of high-fat
Freeman, L.R., Haley-Zitlin, V., Rosenberger, D.S., Granholm, A.C., 2014. Damaging diet-induced NAFLD. Int. J. Mol. Sci. 14, 21240–21257.
effects of a high-fat diet to the brain and cognition: a review of proposed Naninck, E.F., Hoeijmakers, L., Kakava-Georgiadou, N., Meesters, A., Lazic, S.E.,
mechanisms. Nutr. Neurosci. 17, 241–251. Lucassen, P.J., Korosi, A., 2015. Chronic early life stress alters developmental
Heim, C., Nemeroff, C.B., 1999. The impact of early adverse experiences on brain and adult neurogenesis and impairs cognitive function in mice. Hippocampus
systems involved in the pathophysiology of anxiety and affective disorders. 25, 309–328.
Biol. Psychiatry 46, 1509–1522. Polter, A., Beurel, E., Yang, S., Garner, R., Song, L., Miller, C.A., Sweatt, J.D.,
Hernandez, F., Borrell, J., Guaza, C., Avila, J., Lucas, J.J., 2002. Spatial learning deficit McMahon, L., Bartolucci, A.A., Li, X., Jope, R.S., 2010. Deficiency in the inhibitory
in transgenic mice that conditionally over-express GSK-3beta in the brain but serine-phosphorylation of glycogen synthase kinase-3 increases sensitivity to
do not form tau filaments. J. Neurochem. 83, 1529–1533. mood disturbances. Neuropsychopharmacology 35, 1761–1774.
Hoy, A.J., Bruce, C.R., Cederberg, A., Turner, N., James, D.E., Cooney, G.J., Kraegen, Polter, A.M., Yang, S., Jope, R.S., Li, X., 2012. Functional significance of glycogen
E.W., 2007. Glucose infusion causes insulin resistance in skeletal muscle of rats synthase kinase-3 regulation by serotonin. Cell. Signal. 24, 265–271.
without changes in Akt and AS160 phosphorylation. Am. J. Physiol. Endocrinol. Prickaerts, J., Moechars, D., Cryns, K., Lenaerts, I., van Craenendonck, H., Goris, I.,
Metab. 293, E1358–1364. Daneels, G., Bouwknecht, J.A., Steckler, T., 2006. Transgenic mice
Ivy, A.S., Brunson, K.L., Sandman, C., Baram, T.Z., 2008. Dysfunctional nurturing overexpressing glycogen synthase kinase 3beta: a putative model of
behavior in rat dams with limited access to nesting material: a clinically hyperactivity and mania. J. Neurosci. 26, 9022–9029.
relevant model for early-life stress. Neuroscience 154, 1132–1142. Prusator, D.K., Greenwood-Van Meerveld, B., 2015. Gender specific effects of
Ivy, A.S., Rex, C.S., Chen, Y., Dube, C., Maras, P.M., Grigoriadis, D.E., Gall, C.M., Lynch, neonatal limited nesting on viscerosomatic sensitivity and anxiety-like
G., Baram, T.Z., 2010. Hippocampal dysfunction and cognitive impairments behavior in adult rats. Neurogastroenterol. Motil. 27, 72–81.
provoked by chronic early-life stress involve excessive activation of CRH Rodgers, R.J., Dalvi, A., 1997. Anxiety, defence and the elevated plus-maze.
receptors. J. Neurosci. 30, 13005–13015. Neurosci. Biobehav. Rev. 21, 801–810.
Korte, S.M., De Boer, S.F., 2003. A robust animal model of state anxiety: Sanchez, M.M., Ladd, C.O., Plotsky, P.M., 2001. Early adverse experience as a
fear-potentiated behaviour in the elevated plus-maze. Eur. J. Pharmacol. 463, developmental risk factor for later psychopathology: evidence from rodent
163–175. and primate models. Dev. Psychopathol. 13, 419–449.
Krishna, S., Keralapurath, M.M., Lin, Z., Wagner, J.J., de La Serre, C.B., Harn, D.A., Spittaels, K., Van den Haute, C., Van Dorpe, J., Terwel, D., Vandezande, K., Lasrado,
Filipov, N.M., 2015. Neurochemical and electrophysiological deficits in the R., Bruynseels, K., Irizarry, M., Verhoye, M., Van Lint, J., Vandenheede, J.R.,
ventral hippocampus and selective behavioral alterations caused by high-fat Ashton, D., Mercken, M., Loos, R., Hyman, B., Van der Linden, A., Geerts, H., Van
diet in female C57BL/6 mice. Neuroscience 297, 170–181. Leuven, F., 2002. Neonatal neuronal overexpression of glycogen synthase
Li, G., Liu, T., Kong, X., Wang, L., Jin, X., 2014. Hippocampal glycogen synthase kinase-3 beta reduces brain size in transgenic mice. Neuroscience 113,
kinase 3beta is critical for the antidepressant effect of cyclin-dependent kinase 797–808.
5 inhibitor in rats. J. Mol. Neurosci. 54, 92–99. Surget, A., Tanti, A., Leonardo, E.D., Laugeray, A., Rainer, Q., Touma, C., Palme, R.,
Machado, T.D., Dalle Molle, R., Laureano, D.P., Portella, A.K., Werlang, I.C., Benetti Griebel, G., Ibarguen-Vargas, Y., Hen, R., Belzung, C., 2011. Antidepressants
Cda, S., Noschang, C., Silveira, P.P., 2013. Early life stress is associated with recruit new neurons to improve stress response regulation. Mol. Psychiatry 16,
anxiety, increased stress responsivity and preference for comfort foods in adult 1177–1188.
female rats. Stress 16, 549–556. Teegarden, S.L., Bale, T.L., 2008. Effects of stress on dietary preference and intake
Macri, S., Chiarotti, F., Wurbel, H., 2008. Maternal separation and maternal care act are dependent on access and stress sensitivity. Physiol. Behav. 93, 713–723.
independently on the development of HPA responses in male rats. Behav. Brain Tschopp, O., Yang, Z.Z., Brodbeck, D., Dummler, B.A., Hemmings-Mieszczak, M.,
Res. 191, 227–234. Watanabe, T., Michaelis, T., Frahm, J., Hemmings, B.A., 2005. Essential role of
Malter Cohen, M., Jing, D., Yang, R.R., Tottenham, N., Lee, F.S., Casey, B.J., 2013. protein kinase B gamma (PKB gamma/Akt3) in postnatal brain development
Early-life stress has persistent effects on amygdala function and development but not in glucose homeostasis. Development 132, 2943–2954.
in mice and humans. Proc. Natl. Acad. Sci. U. S. A. 110, 18274–18278. Wang, Q., Liu, L., Pei, L., Ju, W., Ahmadian, G., Lu, J., Wang, Y., Liu, F., Wang, Y.T.,
Maniam, J., Morris, M.J., 2010a. Palatable cafeteria diet ameliorates anxiety and 2003. Control of synaptic strength, a novel function of Akt. Neuron 38,
depression-like symptoms following an adverse early environment. 915–928.
Psychoneuroendocrinology 35, 717–728. Wang, X.D., Labermaier, C., Holsboer, F., Wurst, W., Deussing, J.M., Muller, M.B.,
Maniam, J., Morris, M.J., 2010b. Voluntary exercise and palatable high-fat diet both Schmidt, M.V., 2012. Early-life stress-induced anxiety-related behavior in
improve behavioural profile and stress responses in male rats exposed to early adult mice partially requires forebrain corticotropin-releasing hormone
life stress: role of hippocampus. Psychoneuroendocrinology 35, 1553–1564. receptor 1. Eur. J. Neurosci. 36, 2360–2367.
Maniam, J., Morris, M.J., 2012. The link between stress and feeding behaviour. Ziabreva, I., Schnabel, R., Braun, K., 2000. Parental deprivation induces
Neuropharmacology 63, 97–110. N-methyl-d-aspartate-receptor upregulation in limbic brain areas of Octodon
Maniam, J., Antoniadis, C., Morris, M.J., 2014. Early-life stress, HPA axis adaptation, degus: protective role of the maternal call. Neural Plast. 7, 233–244.
and mechanisms contributing to later health outcomes. Front. Endocrinol. 5, Ziabreva, I., Poeggel, G., Schnabel, R., Braun, K., 2003. Separation-induced receptor
73. changes in the hippocampus and amygdala of Octodon degus: influence of
Maysami, S., Haley, M.J., Gorenkova, N., Krishnan, S., McColl, B.W., Lawrence, C.B., maternal vocalizations. J. Neurosci. 23, 5329–5336.
2015. Prolonged diet-induced obesity in mice modifies the inflammatory la Fleur, S.E., Houshyar, H., Roy, M., Dallman, M.F., 2005. Choice of lard, but not
response and leads to worse outcome after stroke. J. Neuroinflammation 12, total lard calories, damps adrenocorticotropin responses to restraint.
140. Endocrinology 146, 2193–2199.
Meaney, M.J., Aitken, D.H., van Berkel, C., Bhatnagar, S., Sapolsky, R.M., 1988. Effect
of neonatal handling on age-related impairments associated with the
hippocampus. Science 239, 766–768.

You might also like