2013 Probiotic Gut Effect Prevents The Chronic Psychological Stress Induced Brain Abnormality in Mice

You might also like

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

Neurogastroenterology & Motility

Neurogastroenterol Motil (2014) 26, 510–520 doi: 10.1111/nmo.12295

Probiotic gut effect prevents the chronic psychological


stress-induced brain activity abnormality in mice

A. AIT-BELGNAOUI ,*,† A. COLOM ,* V. BRANISTE ,* L. RAMALHO ,* A. MARROT ,*,† C. CARTIER ,* E. HOUDEAU ,*


V. THEODOROU * & T. TOMPKINS †

*Neuro-Gastroenterologie et Nutrition team, TOXALIM, UMR 1331-INRA/INP/UPS, Toulouse, France


†Lallemand Health Solutions Inc., Montreal, QC, Canada

Key Messages

• This study provides the conclusive proof effect of probiotic formulation (Lactobacillus helveticus R0052 and
Bifidobacterium longum R0175) on chronic stress-induced brain activity dysregulation.
• This probiotic formulation treatment may contribute to its ability to ameliorate chronic stress-induced
abnormal brain plasticity, reduction in neurogenesis, and increase in stress hormone.
• These findings highlight the important role of bacteria in the bidirectional communication of the gut- brain
axis and suggest that probiotic treatment may prove to be a useful therapeutic alternative in stress-related
disorders such as anxiety and depression.

Abstract ability and tight junction (TJ) proteins levels were also
Background A probiotic formulation (Lactobacillus determinated. Key Results We observed that a pretreat-
helveticus R0052 and Bifidobacterium longum R0175 ment with the probiotic formulation attenuated HPA
combination, Probio’Stickâ) displays anxiolytic-like axis and ANS activities in response to WAS, and
activity and reduces apoptosis in the lymbic system in reduced cFos expression in different brain areas but
animal models of depression. Based on the hypothesis Lactobacillus salivarius (a negative control) treatment
that modulation of gut microbiota by this probiotic was ineffective on these parameters. Moreover, probi-
formulation has beneficial effects on brain activity in otic pretreatment prevented the WAS-induced decrease
stress conditions, we report a set of probiotic-evoked hippocampal neurogenesis and expression changes in
physiological, cellular, and molecular events in the hypothalamic genes involved in synaptic plasticity.
brain of Probio’Stickâ pretreated mice submitted to These central effects were associated with restoration of
chronic psychological stress. Methods Water avoidance TJ barrier integrity in stressed mice. Conclusions &
stress (WAS) was applied or not (sham). Hypothalamic- Inferences These data suggest that chronic stress-
pituitary-adrenal (HPA) axis and autonomic nervous induced abnormal brain plasticity and reduction in
system (ANS) responses to the chronic stress were neurogenesis can be prevented by a pretreatment with
assessed through plasma corticosterone and catechol- the Probio’Stickâ formulation, suggesting that probiot-
amine measurements. Specific markers for neuronal ics modulate neuroregulatory factors and various sig-
activity, neurogenesis, and synaptic plasticity were naling pathways in the central nervous system involved
used to assess brain activity. In addition, gut perme- in stress response.

Keywords brain-gut axis, chronic psychological


Address for Correspondence stress, intestinal barrier, probiotics.
Dr Afifa Ait-Belgnaoui, Neuro-Gastroenterology & Nutrition
Team, TOXALIM, UMR 1331-INRA/INP/UPS, 180 chemin de
Tournefeuille, BP 3, 31027 Toulouse Cedex 9, France.
Tel: 33 561 28 55 61; fax: 33 561 28 51 45; INTRODUCTION
e-mail: afifa@toulouse.inra.fr
Received: 30 July 2013 Chronic stress and acute traumatic experiences during
Accepted for publication: 29 November 2013 life are prevalent psychosocial factors worldwide,

510 © 2013 John Wiley & Sons Ltd


Volume 26, Number 4, April 2014 Probiotic and Brain-gut axis

increasing susceptibility to functional gastrointestinal also in humans, where beneficial properties of the
(GI) disorders or exacerbating symptoms in inflamma- probiotic formulation were correlated with decreased
tory bowel diseases. Patients with irritable bowel levels of urinary free cortisol, suggesting attenuation of
syndrome (IBS) also show augmented sensitivity to HPA axis response to daily stressors.14 Reduction of
environmental stressors, often found comorbid with apoptosis in several brain areas or changes in memory
psychiatric disorders including major depression and and anxiety-like behaviors were also observed in
anxiety. Of note, these patients displayed abnormal animal models of depression after consumption of the
activities of the hypothalamic-pituitary-adrenal (HPA) same probiotic strains,15 or diet supplemented by
axis and autonomic nervous system (ANS) associated L. helveticus R0052 only.16
with exaggerated stress responses and cognitive disor- Based on works to date, our aim was to investigate in
ders.1–3 Recently, animal studies highlighted that mice the central effect of the probiotic combina-
stress or chronic GI inflammation can alter the tion with B. longum R0175 and L. helveticus R0052
composition of gut microbiota, which overall suggest (Probio’Stickâ) on anxiety-like markers induced by
a link between change in enteric bacteria composition, chronic psychological stress. The hypothesis was that
and stress-induced altered GI function4 or anxiety the probiotic treatment may target abnormal brain
behavior.5 activity, enhancement of HPA and ANS activities,
Accumulating data indicate that enteric bacteria and intestinal barrier disruption in chronic stress
communicate with the central nervous system (CNS). conditions.
It is proposed that the microbiota could be intimately
and constitutively involved in the modulation of both
CNS and peripheral nerve functions.6,7 For example, MATERIALS & METHODS
the postnatal bacterial colonization of the gut programs
HPA axis function during development, and germ-free Animal and bacteria preparation
mice display higher exploratory and lower anxiety-like Male 6–8 week old C57Bl6 mice (Janvier SA, Le Genest St Isle,
behavior compared to conventional animals.8,9 Diaz France) weighing 21–23 g were used. The animals were housed
Heijtz et al. extended the complex set of interactions individually under a constant temperature (21  1 °C) in a
pathogen-free animal facility, and maintained on a 12 h light/
between intestinal microbiota and the CNS, in dem-
dark cycle. Food (UAR pellets, Epinay, France) and water were
onstrating a modulatory role in central synaptogenesis available ad libitum. All protocols were approved by the local
supporting normal cognitive function.10 Although the institutional animal (the Toulouse - Midi Pyr en
ees Ethical
causal link between CNS and microbiota remains committee) care and use committee in compliance with the
European laws on the protection of animals (2010/63/EU). The
unknown, overall data support the notion that specific
commercial probiotic given was a combination of freeze-dried
changes in microbiota composition may help to ame- bacteria: L. helveticus R0052 and B. longum R0175 (Pro-
liorate psychological behavior and quality of life in IBS bio’Stickâ, provided by Lallemand Heath Solutions Inc., Blagnac,
patients. According to the studies in laboratory rodents France). L. salivarius HA113 (Lallemand Heath Solutions Inc.)
was used as control negative bacteria and grown at 37 °C in MRS
underlying the benefit of probiotics on stress-evoked
broth (VWR International, Fontenay-sous-Bois, France). After 17 h
GI disorders, it is now of importance to validate of incubation, cultures were harvested by centrifugation at 4500 g
specific probiotic formulations as therapeutic alterna- for 10 min. Bacterial solution was prepared every day in 0.9%
tives in the management of anxiety and stress NaCl and administrated orally at a concentration of 109 CFU/day/
mice during 2 weeks.
responses linked to IBS symptoms. Indeed, some
probiotics have the potential to reduce mRNA expres-
sion of gamma-aminobutyric acid (GABA) receptors Water avoidance stress
associated with anxiety,11 or suppress mitogen stimu-
All stress sessions were performed at the same period of the day
lation-induced increase in plasma tryptophan impli- (between 10:00 am and 12:00 pm) to minimize the influence of
cated in depressive-like behaviors.12 Furthermore, the circadian rhythms. The test apparatus consisted of a plexiglass
modulation of intestinal microbiota by probiotics may tank (42 cm length 9 42 cm width 9 19 cm height) with a block
(3 9 3 9 8 cm height) affixed in the center of the floor. The tank
also prevent stress-associated gut leakiness, due to a
was filled with fresh room temperature water (22 °C) to within
probiotic-mediated attenuation of HPA response in 1 cm of the top of the block. The animals were placed on the
rats exposed to acute psychological stress.13 Recently, block for a period of 1 h daily for four consecutive days
studies assessing the effect of a combination of corresponding to WAS. Sham consisted of placing the mice
similarly for 1 h daily for 4 days on the same platform in a
two probiotics (Bifidobacterium longum R0175 and
waterless container. This well characterized test represents a
Lactobacillus helveticus R0052) not only high- potent psychological stressor with large elevations of ACTH and
lighted their ability to reduce anxiety in rats, but corticosterone within 30 min.17

© 2013 John Wiley & Sons Ltd 511


A. Ait-Belgnaoui et al. Neurogastroenterology and Motility

Experimental design Watson’s atlas.18 For quantitative assessment, the number of


immuno-positive cells was counted bilaterally in 8–10 sections of
Three series of experiments were conducted to assess the effect of selected nuclei using Lucia G4.8 software (Nikon France) in
probiotic formulation on WAS or sham. Eight groups of 8–10 male different brain areas.
mice received orally during 2 weeks either saline or probiotic
formulation (109 CFU/mouse/day). At the end of this period,
animals were submitted to WAS 1 h/day during four consecutive Plasma corticosterone, adrenaline, and
days or sham. In the first series of experiments, four groups of noradrenaline concentration
animals were used. After the chronic stress, blood was collected for
corticosterone, noradrenaline, and adrenaline plasmatic levels were After chronic stress, blood was withdrawn from the facial vein in
determined using Elisa kit. Mice were sacrificed and transcriptional heparinised tube, centrifuged, and plasma was recovered and
analyses of neuronal gene plasticity were investigated on the stored at 80 °C until assay. Corticosterone concentrations were
hypothalamus. Colonic samples were also dissected for the determined by a solid phase sandwich enzyme linked immuno-
evaluation of the TJs protein expression levels. In the second series sorbent assay (ELISA), specific for mice (Immunodiagnostic
of experiments, another four groups of animals were used and after system, Pouilly-en-Auxois, France). Noradrenaline and adrenaline
the chronic stress, the brain of animals was removed and the free were assessed by 2-CAT Research ELISA kit (LDN, Nordhorm,
floating sections were immunostained for cFos and doublecortine Germany) according to manufacturer’s instruction.
positive cells determination. And the in vivo intestinal perme-
ability was assessed in the last series of experiments.
The same experiments described below were conducted with PCR array focused on HPA axis plasticity
another probiotic L. salivarius HA113 to verify the probiotic
specificity on brain activity in response to chronic stress. Mice were decapitated and in aseptic conditions, brains were
removed from the skull and cooled on ice for 20 s followed by
coronal section. The hypothalamus was quickly dissected out
Tissue processing and staining from a coronal midbrain section that included the anterior to
posterior hypothalamus, placed in sterile tubes, and frozen on
Mice were deeply anesthetized using a mixture of xylazine- dry ice. Each sample was individually homogenized in a bead
ketamine (10 mg/kg and 100 mg/kg i.p., respectively). Transcar- beater for 2 min in a sterile microcentrifuge tube containing
dial perfusion was performed with 50 mL isotonic saline (0.9% 1 mL QIAzol lysing reagent (Qiagen, GmbH, Hilden, Germany).
NaCl) followed by 60–100 mL of 4% paraformaldehyde (PAF) After the addition of 160 lL of chloroform, the samples were
fixation. Brain were dissected and removed, postfixed at 4 °C in vortexed for 15 s and left to stand for 2–3 min at room
4% PAF in phosphate-buffered saline (PBS) for 4 h and cryo- temperature. Finally, the samples were centrifuged at 12 000 g
protected overnight in 30% sucrose at 4 °C. Frozen serial for 15 min at 4 °C and the supernatants collected. Total RNA
sections (40 lm-thick) were collected in PBS and then rinsed was reverse-transcribed with a high-capacity cDNA reverse
twice. For immunostainning, the brain slices were incubated at transcription kit (Applied Biosystems, Saint-Aubin, France).
room temperature in a blocking solution of 1% normal goat Real time PCR was performed using the SYBR green IQ mix
serum in PBS with 0.25% Triton X-100 for 30 min and then (Bio-Rad, Hercules, CA, USA). Fluorescence was monitored and
incubated overnight at 4 °C with rabbit polyclonal cFos anti- analyzed in a CFX 96 detection system instrument (Thermal-
body diluted in blocking solution (1 : 10 000; Ab-5, AbCys, Cycler C1000; Bio-Rad). All data were normalized to b-actin for
Paris, France) for 24 h at 4 °C. The incubated sections were each sample, all samples were run in duplicate and were
washed twice and incubated with biotinylated goat antirabbit analyzed using LingRegPCR.19
secondary antibody, diluted 1 : 1000 in blocking solution, and
then incubated with the avidin–biotin complex (Vectastain Elite
kit; Vector Laboratories, Paris, France). Peroxidase activity was
revealed using diaminobenzidine as chromogene (DAB substrate In vivo intestinal permeability measurement
kit; Vector Laboratories). The sections were then mounted on
Evaluation of colonic paracellular permeability was performed
gelatin-coated slides, dehydrated, and coverslipped with Prolong
using 51Cr-EDTA (Perkin Elmer Life Sciences, Paris, France) as a
Antifade kit (Life Technologies SAS, Saint Aubin, France). The
marker of paracellular permeation of apical TJ complexes. 51Cr-
presence of cFos immunoreactivity was detected as a dark
EDTA (50 000 cpm/mice), diluted in 200 lL of saline was
brown reaction product in cell nuclei under a light microscope
administrated orally after WAS or sham. The animals were then
(90i Nikon; Nikon France, Champigny-sur-Marne, France). For
placed in metabolic cages, and feces and urine were collected
immunofluorescence, sections were permeabilized in PBS-Tri-
separately for 6 h. The radioactivity in the urine was measured on
ton X-100 and incubated at RT in a blocking solution of 3%
a gamma counter (Cobra II, Packard Meriden, Connecticut, USA).
normal donkey serum in PBS with 0.3% Triton X-100 for
Permeability to 51Cr-EDTA was expressed as the percentage of the
30 min. The following antibodies and final dilution were used.
total radioactivity administered.
Primary antibodies: goat polyclonal anti-a-Doublecortin, 1 : 250
(DCX C-18; Santa Cruz Laboratories, Santa Cruz, CA, USA)
antibodies; Fluorescent conjugated secondary antibodies (Alexa
488 and 594; Molecular Probes) were also used. Images were
Protein extraction and Western blot
acquired with a 90i Nikon microscope (Nikon France).
Colons were homogenized in RIPA buffer (1% Igepal, 0.5%
deoxycholic acid, and 0.1% SDS in Tris buffered saline solution
19; pH 7.4) with protease inhibitors (Roche Diagnostics,
Cell counts and statistics Meylan, France), and centrifuged at 10 000 g for 10 min
(4 °C). Protein concentrations were assessed using BC Assay
The anatomical localization of immuno-positive cells on the
Uptima kit (Interchim, Montlucßon, France), and equal amounts
planes of sections was standardized according to the Paxinos and

512 © 2013 John Wiley & Sons Ltd


Volume 26, Number 4, April 2014 Probiotic and Brain-gut axis

of protein were separated by SDS/PAGE and transferred onto adrenaline and noradrenaline levels was also observed
nitrocellulose membranes. Membranes were blocked in Odyssey in WAS mice (p < 0.05 vs sham; Fig. 1). There is no
blocker and then incubated with the polyclonal rabbit antijunc-
tion adhesion molecule (JAM-A; 1/500) or antioccludin (1/500; effect of probiotic formulation on plasmatic levels of
Invitrogen, Vergy Pontoise, France) or 3-phosphate dehydroge- corticosterone and catecholamines in basal conditions,
nase (GAPDH 1/5000; Cell signaling Technology, Danvers, MA, while the probiotic treatment prevented WAS-induced
USA) overnight at 4 °C. After washing, the membranes were
increase in these plasma markers of chronic stress
incubated with fluorescent CF773 antirabbit 1 : 10 000 antibody
(Biotium, Hayward, CA, USA) with gentle rocking and protected (p < 0.05; Fig. 1).
from light. Finally, they were scanned on infrared imaging Regarding L. salivarius pretreatment, we have
system Odyssey (Li-Cor, Lincoln, NE, USA). Band intensity was observed that this probiotic has no effect on
analyzed by densitometry using the software ImajeJ (http://
higher plasmatic levels of corticosterone, adrenaline,
rsbweb.nih.gov/ij). Occludin and JAMA expression was assessed
relative to GAPDH for each sample analyzed. and noradrenaline induced by WAS (Fig. 1). So, we
used this strain as a negative control in other
experiment.
Statistical analysis
All data were expressed as means  SEM. Analyses were
performed by running Prism 4 software (GraphPad, San Diego, Effect of probiotic (Probio’Stickâ) treatment on
CA, USA). Statistical comparisons between two groups were chronic stress-induced neuronal activation in
performed using one-way ANOVA. Differences were considered
significant when post Student’s t-test (LSD) p < 0.05. Real
brain
time RT-PCR values were analyzed by a two-sided Student’s t-test.
Compared to basal levels (sham), chronic stress (WAS)
increased cFos positive nuclei number in the following
RESULTS brain areas: hypothalamic paraventricular nucleus
(PVN), amygdaloid nucleus (AN), lateral parabrachial
Effect of probiotic (Probio’Stickâ) treatment on nucleus, locus coeruleus, nucleus tractus solitary, and
neuroendocrine and catecholamine response to CA-1/3 of hippocampus (Table 1). In dentate gyrus
chronic stress (DG) of the hippocampus, the number of cFos positive
nuclei was significantly lower than in sham groups
Compared to sham animals, corticosterone plasmatic (Table 1). A two-week pretreatment with probiotic
levels sharply increased in response to chronic stress significantly (p < 0.05) lessened the increase in cFos
(184.5  20.2 vs 598.0  95.1 ng/mL in WAS mice, positive neurons induced by WAS in the PVN, AN, and
p < 0.05; Fig. 1). A significant increase in plasma CA-3 compared to sham. In contrast, the probiotic

Figure 1 Effect of oral pretreament by


Probio’Stickâ or vehicle (saline) on plasma
corticosterone, adrenaline, and
noradrenaline concentration in mice
submitted to chronic stress. (a and b)
p < 0.05, respectively, were significantly
different from sham + vehicle and WAS +
vehicle.

© 2013 John Wiley & Sons Ltd 513


A. Ait-Belgnaoui et al. Neurogastroenterology and Motility

pretreatment significantly increased the number of Differential expression of genes involved in


cFos positive nuclei in WAS mice compared to vehicle synaptic plasticity in the hypothalamus after
groups (p < 0.05; Table 1). Probio’Stickâ treatment in stressed mice

Table 1 Number of Fos-immunoreactive nuclei per region in the brain In WAS conditions, genes involved in the neurotrophic
of mice submitted to WAS or sham after or not probiotic treatment signals displayed a lower expression level 40.7% for
BDNF (brain-derived neutrophic factor), a neutrophic
Sham WAS
factor, compared with control mice (sham). Similar
Vehicle Probiotic Vehicle Probiotic difference in gene expression was also observed for
serpin, Plau (urokinase-type plasminogen activator)
Forebrain
LPB 2 1 11 23  2 25  1 and Mmp9 (matrix metalloprotease; involved in tissue
LC 7 3 32 24  4 21  2 remodeling). The same gene expression profiling of the
NTS 5 4 34 83  11 62  15 cytoskeleton (Gap43: neuronal growth associated pro-
Hypothalamus
PVN 20 1 15  3 150  13 54  7* tein 43) and cell adhesion markers (tenascin-C, Reln:
Amygdala reelin and neural cell adhesion molecule) was observed
BLA 17 1 23  1 120  11 113  12 in stressed mice. Regarding the expression of genes
CeA 15 3 19  1 112  8 93  12
MeA 19 4 20  1 160  10 43  2* involved in microglia activation and synaptogenesis,
Hippocampus mRNA levels of Gfap (glial fibrillary acid protein), Vim
CA1 2 0 11 14  4 15  3 (vimentine), Syt4 (synaptotagmin), and Snap25 (synap-
CA3 1 0 31 20  4 7  2*
DG 5 3 41 23  0 46  2* tic-associated protein 25 kDa) were significantly
higher in WAS mice compared to sham (p < 0.05;
Values are given as the mean total number of Fos-IR nuclei per region Fig. 2).
of interest, and asterisk values are significant higher than correspond-
ing value in WAS treated with vehicle (one-way ANOVA, p < 0.05). When compared to WAS mice receiving the vehicle,
LBP, lateral parabrachial nucleus; LC, locus coeruleus; NTS, nucleus the probiotic treatment in stressed mice significantly
tractus solitary; PVN, paraventricular hypothalamic nucleus; BLA, baso- increased the expression of neurotrophic factor
lateral amygdaloid nucleus; CeA, central amygdaloid nucleus; MeA,
medial amygdaloid nucleus; CA1, hippocampal pyramidal cells, aera CA1; (BDNF) and decreased the expression of cytoskeleton
CA3, hippocampal pyramidal cells, aera CA3; DG, dentate gyrus. organization and microglial activation markers

Figure 2 Absolute percentage of expression of differences in each significant gene for the WAS (W) and WAS + probiotic (WP) compared with the
sham + vehicle. Values are means; n = 8 per group. *p < 0.05 and #p < 0.05 were considered significant from the Sham and WAS groups.

514 © 2013 John Wiley & Sons Ltd


Volume 26, Number 4, April 2014 Probiotic and Brain-gut axis

(Gap43, Gfap, Vim), synaptogenesis (Syt4, Snap25) and compared to sham (87.5  5.2 versus 51.2  3;
cell adhesion markers (Reln, Sema [semaphorine]; p < 0.05). This decrease appeared more pronounced in
Fig. 2). the subgranular zone of the granular cell layer in the
DG (Fig. 3). Probiotic pretreatment before and during
WAS completely prevented the WAS-induced decrease
Effect of probiotic (Probio’Stickâ) treatment on
in neuronal growth (Fig. 3).
neurogenesis in stressed mice
The impact of probiotic formulation on neurogenesis
Probiotic (Probio’Stickâ) prevents gut barrier
was determined by quantifying in the DG of hippo-
impairment in stress mice
campus the doublecortin (DCX)-positive cells, which is
one of earliest marker of neuronal density. In non- Probiotics treatment did not affect intestinal permeabil-
treated animals, a significant reduction in DG neuro- ity nor the expression of the TJ proteins occludin and
genesis occurred in response to WAS, with a 41.5% JAM-A in basal conditions (i.e., without chronic stress)—
decrease of DCX-positive cells in WAS-vehicle group two transmembrane proteins that seal intercellular

Figure 3 Quantification of neurogenesis by


detection of Doublecortin-expressing cells
after probiotic treatment (Probio’Stickâ) in
stressed mice. (A) Histogram representing
the number of DCX-expressing cells (SEM)
in the dentate gyrus of mice hippocampus
submitted to sham and WAS after vehicle or
probiotic treatment (Probio’Stickâ).
Statistical differences between WAS and
sham or WAS + probiotic are shown,
respectively, as ap < 0.05 and bp < 0.05. (B)
Confocal images of Doublecortin-expressing
in the dentate gyrus of sham and WAS
treated or not by probiotic (Probio’Stickâ).
White arrows point to labeled cells in the
inner granule cell layer in the dentate gyrus.

© 2013 John Wiley & Sons Ltd 515


A. Ait-Belgnaoui et al. Neurogastroenterology and Motility

A esis induced by chronic stress. A transcriptional


analysis of the hypothalamus also suggested a benefi-
cial role of pretreatment with this probiotic in modu-
lating neuronal networks that coordinate synaptic
plasticity. We also showed that probiotic pretreatment
reduces the increase in intestinal permeability induced
by chronic stress, which is correlated by a prevention
of the stress-induced degradation in colonic mucosa of
TJ proteins.
It is well established that chronic stress exposure in
animals physically altered the structure and function
B of brain regions involved in the HPA and ANS response
to stress.21 Chronic stress is often used to mimic
human depressive illness in animals and to elicit an
increase in noradrenaline in the hippocampus known
to be associated with deficits in learning and memory
tasks.22 Water avoidance stress is a well-described and
validated model for chronic psychological stress and
WAS animals displayed a more anxious and timid
behavior.23 In this study, the treatment of stressed
mice with the probiotic attenuated the HPA axis and
ANS responses to chronic stress, in diminishing cor-
ticosterone, noradrenaline and adrenaline circulating
levels. In contrast, L. salivarius treatment had no
effect on neuroendocrine response to chronic stress.
The lack of stress hormone response suggests that the
regulation of HPA and ANS response to chronic stress
Figure 4 Effect of oral pretreatment by Probio’Stickâ of vehicle is bacterial strain dependant. Thus, the normalization
(saline) on (A) hyperpermeability and (B) both colonic mucosal
occludin and JAM-A expression in mice submitted to chronic stress. of these stress hormones may be considered as an
a,b
p < 0.05, respectively, were significantly different from sham + important criterion in the assessment of antidepres-
vehicle and WAS + vehicle. sant activity of probiotics. In support of this, a recent
clinical study has shown that the same probiotic given
space at apical tight junctions.20 Water avoidance stress daily over 30 days significantly reduced psychological
procedure significantly (p < 0.05) increased intestinal distress in healthy volunteers, associated with a
permeability to 51Cr-EDTA in comparison to non- decrease in urinary free cortisol levels.24 In rats,
stressed mice (sham; Fig. 4), and this effect was abolished Desbonnet et al., have shown a reduced noradrenaline
after probiotic (Probio’Stickâ) pretreatment (Fig. 4A). concentration in the hippocampus following chronic
A reduction in the expression of occludin and JAM-A in administration of bifidobacteria treatment for
the colonic mucosa of WAS mice was also observed 14 days.12 The administration of tricyclic antidepres-
(Fig. 4A). Probiotic (Probio’Stickâ) pretreatment signif- sants to animals induced also a decrease in noradren-
icantly (p < 0.05) restored occludin and JAM-A expres- aline levels in the hippocampus,25 suggesting that
sion to basal levels (i.e., sham+vehicule; Fig. 4B). bifidobacteria may affect the central neurochemical
under stress conditions by mechanisms comparable to
that observed in with chronic antidepressant drugs.
DISCUSSION
Germ-free mice exhibit a higher turnover rate of the
This study indicates that a 2-week treatment with the anxiety inducers, like noradrenaline, dopamine, and
probiotic formulation Probio’Stickâ (L. helveticus serotonin in the striatum,10 which can influence the
R0052 and B. longum R0175) attenuated hypothal- development of behavior.9 It is probably that these
amo-pituitary-adrenal (HPA) axis and ANS response to central neurochemical changes in the brain of stressed
chronic stress as reflected by a decrease in plasmatic mice were correlated with a marked neuroplasticity of
levels of corticosterone, adrenaline, and noradrenaline neurotransmitters in the PVN of the hypothalamus
in stressed mice. Moreover, the probiotic prevented that predicts enhanced HPA axis excitability in
changes in central neuronal activation and neurogen- response to chronic stress.26 Interestingly, recent

516 © 2013 John Wiley & Sons Ltd


Volume 26, Number 4, April 2014 Probiotic and Brain-gut axis

studies have shown the ability of a probiotic treatment of the balance between neuroprotection and neurotox-
to decrease hypothalamic corticotropin-releasing hor- icity.34 At a certain threshold of sensitivity, stress
mone (CRF) expression in stressed rats13 or to modu- exposure can evoke a neuroimmune response35 and
late GABA neurotransmission implicated in anxiety microglial remodeling36 able to modulate synaptic
behavior in a cerebral area dependent manner.11 These function, impairing the normal establishment of neu-
data suggest that the probiotics can affect neuronal rons connections. Our recent study reported the ability
circuits implicated in the HPA axis response to stress of probiotic pretreatment to counteract the hypotha-
and we examined gene expression changes involved in lamic neuroinflammation in stressed rats by a mech-
the neuronal plasticity in the hypothalamus. We report anism depending upon prevention of gut leakiness.13
herein the overall profiles of probiotic-triggered gene Thus, one way hypothesized in the current study that
expression with distinctive or common functions in probiotic treatment may attenuate the neuronal and
the regulation of neuronal networks. Transcriptional glial disorganization-induced by chronic psychological
analysis has shown that the neurotrophic factor BDNF stress, hence normalizes neuronal networks.
is upregulated after probiotic treatment in stressed Neuronal plasticity in the brain is a normal process
mice, a change could be link to the ability of throughout life, both in the formation of new synaptic
glucocorticoids to regulate the HPA axis. Indeed, it connections, new neurons and glial cells. However,
has been shown that impairment in glucocorticoid stress can inhibit this plasticity and lead to cerebral
receptor function in the PVN led to upregulation of the atrophy. In psychiatric disorders, such changes that
hypothalamic levels of BDNF, and disinhibition of the occurred in sensitive brain areas, which may play a role
HPA axis.27 Bercik et al. have demonstrated that a in memory function and emotional states. In the
transient perturbation of the gut microbiota increased hippocampus, a remarkable capacity for structural
hippocampal BDNF and this change was correlated reorganization has been well described.37 Preexisting
with an altered exploratory behavior.28 Moreover, it is neural circuit undergoes modifications in dendritic
interesting to note in this study that other genes complexity and synapse number, and entirely novel
known to participate in neurotransmission and synap- neural connections are formed through a process of
tic plasticity appeared also modulated in stressed mice neurogenesis. Stress exerts a significant brake in this
after probiotic treatment. For instance, the hypotha- process as a potent negative regulator of neurogenesis.
lamic expression of Syt4 and Snap25, a regulator of a Different chronic stress conditions suppress the rate of
synaptic vesicule exocytosis in neurons, was decreased adult DG proliferation in the hippocampus, and
after probiotic treatment in stressed mice. Previous decreases the size of newborn cell clusters.38 In the
studies emphasized their expression in the brain hippocampus and the prefrontal cortex, chronic stress
associated with associative and spatial memory29,30 can induce plasticity of stress circuitry through retrac-
and long-term potentiation,31 respectively. A decrease tion of apical dendrites, reduction in spine density in
in Snap25 and Syt4 knockout mouse are responsible for the CA-3 region of the hippocampus,39 suppression of
presynaptic terminal damage in ischemic brain32, and neurogenesis of DG granule neurons40 and a decrease in
mice displays lower anxiety and less depression-like glucocorticoid receptor expression.41 Chronic stress
behavior.29 On this basis, it is suggested in this study also promotes changes in the PVN of the hypothalamus,
that the probiotic treatment in stressed mice regulates including increase in CRF, reduction in glucocorticoid
synaptic functions in the neuronal networks, hence receptor42 and altered expression of numerous neuro-
protects against stress-induced aberrant synaptic transmitter receptor subunits.43,44 Finally, neurochem-
changes in the brain. Another group of genes, such as ical changes are seen in numerous stress regulatory
Mmp9, Gap43, Reln, Sema, Gfap and Vim highly pathways which are projected in the PVN and increases
implicated in tissue remodeling, cytoskeleton organi- secretagogue synthesis in the PVN. These neurochem-
zation, cell adhesion, and axonal elongation were also ical changes suggest that chronic stress enhances the
modulated in stressed mice receiving probiotic treat- excitability of HPA to novel stress and decreases HPA
ment. The regulation of stress activation and recovery negative feedback at the level of the PVN and hippo-
involves tight coordination between neuronal and glial campus. In this study, we found that chronic stress in
networks.33 In addition to their role of support and mice suppressed neurogenesis in the hippocampus
nutrition of neurons, glial cells defend CNS from stress reflected by a decrease in doublecortin (DCX)-immu-
insult through an up-regulation of the local inflamma- noreactive cells in the granule precursor cells of the DG,
tory processes. However, excessive or prolonged glial and this number of DCX-positive cells was restored in
activation enhances a chronic damage that eventually the probiotic-treated mice. These data suggest that
propagates neuroinflammation suggesting the delicacy probiotic is able to enhance survival and differentiation

© 2013 John Wiley & Sons Ltd 517


A. Ait-Belgnaoui et al. Neurogastroenterology and Motility

of cells into neurons. In the hippocampus, the same increase in intestinal paracellular permeability
probiotic in an animal model of depression reduces induced by chronic stress correlated by the prevention
apoptosis propensity through an Akt activity-depen- of the degradation in colonic mucosa expression of
dent mechanism.15 PI3K/Akt is a cell signaling path- tights junction’s proteins (occludin and JAMA). The
way that plays a key role in cellular homeostasis prevention of intestinal barrier integrity by this probi-
through its role in regulation of apoptosis, cell growth, otic formulation has been also shown in model of
and cell cycle.45 This signal transduction pathway postmyocardial infarction depression. It has been
seems to be involved in the changes of synaptic demonstrated that probiotic formulation (B. longum
plasticity in depression states.46 This data support our and L. helveticus) prevents stress-induced memory
findings and suggests that the combination of two deficit in mice through a reduction in a cytokine-
probiotics promotes neurogenesis in the DG of the dependant mechanism.53 Thus, targeting the microbi-
hippocampus in stressed mice, by ameliorating the ota-gut-brain axis to modulate behavior by probiotic
functional recovery of such synaptic plasticity. We have seems to be relevant for the regulation of the stress
also shown that this probiotic treatment enhanced response.
neuronal activation changes in several areas in the In conclusion, our study shows that a 2-week
brain, and more particularly in the hippocampus which treatment by the probiotic formulation Probio’Stickâ
can be correlated to axonal and dendrite neuronal (B. longum and L. helveticus) attenuated HPA axis and
prolongment.47 More investigation was needed for ANS activity in response to stress, and prevented the
exploring the mechanistic relationship of this probiotic stress-induced reduction in neurogenesis in the hippo-
formulation action toward cognitive improvement. An campus. The attenuation of HPA axis by the probiotic
assumed mechanism of action of probiotics on the formulation protects neuronal plasticity at the hypo-
interaction between the brain and the gut could be thalamic level, consequently maintains brain activity
explained by the fact for example that B. longum is able against stress-mediated brain circuitry insult. We can
to modulate enteric neuron excitability48 involving the hypothesize that the probiotic promotion of neurogen-
vagal pathways in the gut-brain communication.49 esis in the hippocampus restored the HPA axis nega-
While Perez-Burgos et al. have shown that other bacte- tive feedback.
ria such as L. salivarius did not alter firing rate of vagal
fibers.50 Consequently, these studies suggest firstly that
FUNDING
vagal afferent signaling pathways are required by
B. longum effect in the normalization of axiety-like No external funding was used for this study.
behavior. And secondly, these pathways are bacterial
strain specific in the brain-gut communication.
DISCLOSURE
It is clear that probiotic can modulate various
aspects of microbiota-gut-brain axis.51 Interestingly, None of the authors have conflicts of interest to declare.
both bacteria modulate enteric neuron excitability,
suggesting that enteric to vagus signaling is an impor-
AUTHOR CONTRIBUTION
tant means of communication along the microbiota-
gut-brain axis.49,52 An indirect role for microbiota in AAB designed the research study, carried out the study, data
stress response was recently demonstrated in an analysis and interpretation, and wrote the manuscript; VB
provided intellectual support to the study and contributed to
animal model of stress-induced intestinal barrier critical revised manuscript; AC, CC, and LR responsible for
impairment. Indeed, a prevention of gut leakiness by material support and contribute to the data interpretation:
intestinal microbiota modulation with a probiotic provide help in immunohistology and transcriptional analysis;
leads to attenuation of the HPA axis response to EH and VT provide critical revision of the manuscript; TT
obtained funding, contributed to data critical interpretation and
stress.13 In the same way, we have shown that manuscript writing.
probiotic formulation pretreatment reduces the

REFERENCES axis in irritable bowel syndrome. Neu- tion in irritable bowel syndrome:
rogastroenterol Motil 2009; 21: 149–59. plasma cytokines as a potential bio-
1 Chang L, Sundaresh S, Elliott J, Anton 2 Dinan TG, Quigley EM, Ahmed SM, marker? Gastroenterology 2006; 130:
PA, Baldi P, Licudine A, Mayer M, Scully P, O’Brien S, O’Mahony L, 304–11.
Vuong T et al. Dysregulation of the O’Mahony S, Shanahan F et al. Hypo- 3 Eriksson EM, Andren KI, Eriksson HT,
hypothalamic-pituitary-adrenal (HPA) thalamic-pituitary-gut axis dysregula- Kurlberg GK. Irritable bowel syn-

518 © 2013 John Wiley & Sons Ltd


Volume 26, Number 4, April 2014 Probiotic and Brain-gut axis

drome subtypes differ in body aware- L, Houdeau E, Fioramonti J et al. cholinergic and noradrenergic sys-
ness, psychological symptoms and Prevention of gut leakiness by a pro- tems in behavioral recovery following
biochemical stress markers. World J biotic treatment leads to attenuated oxotremorine treatment to chroni-
Gastroenterol 2008; 14: 4889–96. HPA response to an acute psycholog- cally stressed rats. Neuroscience
4 O’Mahony CM, Clarke G, Gibney S, ical stress in rats. Psychoneuroendo- 2006; 143: 679–88.
Dinan TG, Cryan JF. Strain crinology 2012; 37: 1885–95. 23 Bradesi S, Schwetz I, Ennes HS, Lamy
differences in the neurochemical 14 Messaoudi M, Lalonde R, Violle N, CM, Ohning G, Fanselow M, Potho-
response to chronic restraint stress Javelot H, Desor D, Nejdi A, Bisson ulakis C, McRoberts JA et al.
in the rat: relevance to depression. JF, Rougeot C et al. Assessment of Repeated exposure to water avoid-
Pharmacol Biochem Behav 2011; 97: psychotropic-like properties of a pro- ance stress in rats: a new model for
690–9. biotic formulation (Lactobacillus sustained visceral hyperalgesia. Am J
5 Bercik P, Verdu EF, Foster JA, Macri J, helveticus R0052 and Bifidobacteri- Physiol Gastrointest Liver Physiol
Potter M, Huang X, Malinowski P, um longum R0175) in rats and human 2005; 289: G42–53.
Jackson W et al. Chronic gastrointes- subjects. Br J Nutr 2011; 105: 755–64. 24 Messaoudi M, Violle N, Bisson JF,
tinal inflammation induces anxiety- 15 Girard SA, Bah TM, Kaloustian S, Desor D, Javelot H, Rougeot C. Ben-
like behavior and alters central Lada-Moldovan L, Rondeau I, Tomp- eficial psychological effects of a pro-
nervous system biochemistry in mice. kins TA, Godbout R, Rousseau G. biotic formulation (Lactobacillus
Gastroenterology 2010; 139: 2102–12, Lactobacillus helveticus and Bifido- helveticus R0052 and Bifidobacteri-
e2101. bacterium longum taken in combina- um longum R0175) in healthy human
6 Collins SM, Bercik P. The relation- tion reduce the apoptosis propensity volunteers. Gut Microbes 2011; 2:
ship between intestinal microbiota in the limbic system after myocardial 256–61.
and the central nervous system in infarction in a rat model. Br J Nutr 25 Chung MY, Kim DG, Yoo KJ, Hong
normal gastrointestinal function and 2009; 102: 1420–5. SS. Regional differences in the levels
disease. Gastroenterology 2009; 136: 16 Ohland CL, Kish L, Bell H, Thiesen of biogenic amines and their metab-
2003–14. A, Hotte N, Pankiv E, Madsen KL. olites in rat brain after tricyclic anti-
7 Grenham S, Clarke G, Cryan JF, Effects of Lactobacillus helveticus on depressant treatments. Yonsei Med J
Dinan TG. Brain-gut-microbe com- murine behavior are dependent on 1993; 34: 266–77.
munication in health and disease. diet and genotype and correlate with 26 Flak JN, Ostrander MM, Tasker JG,
Front Physiol 2011; 2: 94. alterations in the gut microbiome. Herman JP. Chronic stress-induced
8 Sudo N, Chida Y, Aiba Y, Sonoda J, Psychoneuroendocrinology 2013; 38: neurotransmitter plasticity in the
Oyama N, Yu XN, Kubo C, Koga Y. 1738–47. PVN. J Comp Neurol 2009; 517:
Postnatal microbial colonization pro- 17 Million M, Tache Y, Anton P. 156–65.
grams the hypothalamic-pituitary- Susceptibility of Lewis and Fischer 27 Jeanneteau FD, Lambert WM, Ismaili
adrenal system for stress response in rats to stress-induced worsening of N, Bath KG, Lee FS, Garabedian MJ,
mice. J Physiol 2004; 558: 263–75. TNB-colitis: protective role of brain Chao MV. BDNF and glucocorticoids
9 Neufeld KM, Kang N, Bienenstock J, CRF. Am J Physiol 1999; 276: G1027– regulate corticotrophin-releasing hor-
Foster JA. Reduced anxiety-like 36. mone (CRH) homeostasis in the
behavior and central neurochemical 18 Paxinos G, Watson C. The Rat Brain hypothalamus. Proc Natl Acad Sci
change in germ-free mice. Neurogas- in Stereotaxic Coordinates, 2nd edn. USA 2012; 109: 1305–10.
troenterol Motil 2011; 23: 255–64, New York: Academic Press, 1986. 28 Bercik P, Denou E, Collins J, Jackson
e119. 19 Ruijter JM, Ramakers C, Hoogaars W, Lu J, Jury J, Deng Y, Blennerhassett
10 Diaz Heijtz R, Wang S, Anuar F, Qian WM, Karlen Y, Bakker O, van den P et al. The intestinal microbiota
Y, Bj€orkholm B, Samuelsson A, Hib- Hoff MJ, Moorman AF. Amplification affect central levels of brain-derived
berd ML, Forssberg H et al. Normal efficiency: linking baseline and bias neurotropic factor and behavior in
gut microbiota modulates brain in the analysis of quantitative PCR mice. Gastroenterology 2011; 141:
development and behavior. Proc Natl data. Nucleic Acids Res 2009; 37: e45. 599–609, 609 e591–593.
Acad Sci USA 2011; 108: 3047–52. 20 Braniste V, Leveque M, Buisson-Bre- 29 Ferguson GD, Herschman HR, Storm
11 Bravo JA, Forsythe P, Chew MV, nac C, Bueno L, Fioramonti J, Hou- DR. Reduced anxiety and depression-
Escaravage E, Savignac HM, Dinan deau E. Oestradiol decreases colonic like behavior in synaptotagmin IV
TG, Bienenstock J, Cryan JF. Inges- permeability through oestrogen (-/-) mice. Neuropharmacology 2004;
tion of Lactobacillus strain regulates receptor beta-mediated up-regulation 47: 604–11.
emotional behavior and central of occludin and junctional adhesion 30 Liu YF, Chen HI, Wu CL, Kuo YM,
GABA receptor expression in a mouse molecule-A in epithelial cells. J Phys- Yu L, Huang AM, Wu FS, Chuang JI
via the vagus nerve. Proc Natl Acad iol 2009; 587: 3317–28. et al. Differential effects of treadmill
Sci USA 2011; 108: 16050–5. 21 Barha CK, Brummelte S, Lieblich SE, running and wheel running on
12 Desbonnet L, Garrett L, Clarke G, Galea LA. Chronic restraint stress in spatial or aversive learning and
Bienenstock J, Dinan TG. The probi- adolescence differentially influences memory: roles of amygdalar brain-
otic Bifidobacteria infantis: an assess- hypothalamic-pituitary-adrenal axis derived neurotrophic factor and syn-
ment of potential antidepressant function and adult hippocampal neu- aptotagmin I. J Physiol 2009; 587:
properties in the rat. J Psychiatr Res rogenesis in male and female rats. 3221–31.
2008; 43: 164–74. Hippocampus 2011; 21: 1216–27. 31 Roberts LA, Morris BJ, O’Shaugh-
13 Ait-Belgnaoui A, Durand H, Cartier 22 Srikumar BN, Raju TR, Shankaran- nessy CT. Involvement of two iso-
C, Chaumaz G, Eutamene H, Ferrier arayana Rao BS. The involvement of forms of SNAP-25 in the expression

© 2013 John Wiley & Sons Ltd 519


A. Ait-Belgnaoui et al. Neurogastroenterology and Motility

of long-term potentiation in the rat spine morphology in the rat medial 47 Chen S, Hillman DE. Transient c-fos
hippocampus. NeuroReport 1998; 9: prefrontal cortex. J Comp Neurol expression and dendritic spine plas-
33–6. 2008; 507: 1141–50. ticity in hippocampal granule cells.
32 Ishimaru H, Casamenti F, Ueda K, 40 Gould E, McEwen BS, Tanapat P, Brain Res 1992; 577: 169–74.
Maruyama Y, Pepeu G. Changes in Galea LA, Fuchs E. Neurogenesis in 48 Khoshdel A, Verdu EF, Kunze W,
presynaptic proteins, SNAP-25 and the dentate gyrus of the adult tree McLean P, Bergonzelli G, Huizinga
synaptophysin, in the hippocampal shrew is regulated by psychosocial JD. Bifidobacterium longum
CA1 area in ischemic gerbils. Brain stress and NMDA receptor activa- NCC3001 inhibits AH neuron excit-
Res 2001; 903: 94–101. tion. J Neurosci 1997; 17: 2492–8. ability. Neurogastroenterol Motil
33 Theodosis DT, Poulain DA, Oliet SH. 41 Magarinos AM, McEwen BS. Stress- 2013; 25: e478–84.
Activity-dependent structural and induced atrophy of apical dendrites of 49 Bercik P, Park AJ, Sinclair D, Khosh-
functional plasticity of astrocyte-neu- hippocampal CA3c neurons: involve- del A, Lu J, Huang X, Deng Y, Blen-
ron interactions. Physiol Rev 2008; ment of glucocorticoid secretion and nerhassett PA et al. The anxiolytic
88: 983–1008. excitatory amino acid receptors. Neu- effect of Bifidobacterium longum
34 Bailey SL, Carpentier PA, McMahon roscience 1995; 69: 89–98. NCC3001 involves vagal pathways
EJ, Begolka WS, Miller SD. Innate and 42 Makino S, Smith MA, Gold PW. for gut-brain communication. Neuro-
adaptive immune responses of the Increased expression of corticotro- gastroenterol Motil 2011; 23: 1132–9.
central nervous system. Crit Rev pin-releasing hormone and vasopres- 50 Perez-Burgos A, Wang B, Mao YK,
Immunol 2006; 26: 149–88. sin messenger ribonucleic acid Mistry B, McVeyNeufeld KA, Bie-
35 Hueston CM, Barnum CJ, Eberle JA, (mRNA) in the hypothalamic parav- nenstock J, Kunze W. Psychoactive
Ferraioli FJ, Buck HM, Deak T. entricular nucleus during repeated bacteria Lactobacillus rhamnosus (JB-
Stress-dependent changes in neuroin- stress: association with reduction in 1) elicits rapid frequency facilitation
flammatory markers observed after glucocorticoid receptor mRNA levels. in vagal afferents. Am J Physiol Gas-
common laboratory stressors are not Endocrinology 1995; 136: 3299– trointest Liver Physiol 2013; 304:
seen following acute social defeat of 309. G211–20.
the Sprague Dawley rat. Physiol 43 Cullinan WE. GABA(A) receptor sub- 51 Gareau MG, Sherman PM, Walker
Behav 2011; 104: 187–98. unit expression within hypophysio- WA. Probiotics and the gut microbi-
36 Tynan RJ, Beynon SB, Hinwood M, tropic CRH neurons: a dual ota in intestinal health and disease.
Johnson SJ, Nilsson M, Woods JJ, hybridization histochemical study. J Nat Rev Gastroenterol Hepatol 2010;
Walker FR. Chronic stress-induced Comp Neurol 2000; 419: 344–51. 7: 503–14.
disruption of the astrocyte network 44 Ziegler DR, Cullinan WE, Herman JP. 52 Kunze WA, Mao YK, Wang B, Huiz-
is driven by structural atrophy and Organization and regulation of parav- inga JD, Ma X, Forsythe P, Bienen-
not loss of astrocytes. Acta Neuropa- entricular nucleus glutamate signal- stock J. Lactobacillus reuteri
thol 2013; 126: 75–91. ing systems: N-methyl-D-aspartate enhances excitability of colonic AH
37 Shors TJ, Foy MR, Levine S, Thomp- receptors. J Comp Neurol 2005; 484: neurons by inhibiting calcium-depen-
son RF. Unpredictable and uncontrol- 43–56. dent potassium channel opening. J
lable stress impairs neuronal 45 Datta SR, Dudek H, Tao X, Masters S, Cell Mol Med 2009; 13: 2261–70.
plasticity in the rat hippocampus. Fu H, Gotoh Y, Greenberg ME. Akt 53 Arseneault-Breard J, Rondeau I,
Brain Res Bull 1990; 24: 663–7. phosphorylation of BAD couples sur- Gilbert K, Girard SA, Tompkins TA,
38 Sapolsky RM. Is impaired neurogene- vival signals to the cell-intrinsic Godbout R, Rousseau G. Combina-
sis relevant to the affective symptoms death machinery. Cell 1997; 91: tion of Lactobacillus helveticus
of depression? Biol Psychiatry 2004; 231–41. R0052 and Bifidobacterium longum
56: 137–9. 46 Marsden WN. Synaptic plasticity in R0175 reduces post-myocardial
39 Radley JJ, Rocher AB, Rodriguez A, depression: molecular, cellular and infarction depression symptoms and
Ehlenberger DB, Dammann M, McE- functional correlates. Prog Neuropsy- restores intestinal permeability in a
wen BS, Morrison JH, Wearne SL chopharmacol Biol Psychiatry 2013; rat model. Br J Nutr 2012; 107:
et al. Repeated stress alters dendritic 43: 168–84. 1793–9.

520 © 2013 John Wiley & Sons Ltd

You might also like