ArticlePDF Available

Effects of a probiotic add-on treatment on fronto-limbic brain structure, function, and perfusion in depression: Secondary neuroimaging findings of a randomized controlled trial

Authors:

Abstract and Figures

Background Probiotics are suggested to improve depressive symptoms via the microbiota-gut-brain axis. We have recently shown a beneficial clinical effect of probiotic supplementation in patients with depression. Their underlying neural mechanisms remain unknown. Methods A multimodal neuroimaging approach including diffusion tensor imaging, resting-state functional MRI, and arterial spin labeling was used to investigate the effects of a four-weeks probiotic supplementation on fronto-limbic brain structure, function, and perfusion and whether these effects were related to symptom changes. Results Thirty-two patients completed both imaging assessments (18 placebo and 14 probiotics group). Probiotics maintained mean diffusivity in the left uncinate fasciculus, stabilized it in the right uncinate fasciculus, and altered resting-state functional connectivity (rsFC) between limbic structures and the temporal pole to a cluster in the precuneus. Moreover, a cluster in the left superior parietal lobule showed altered rsFC to the subcallosal cortex, the left orbitofrontal cortex, and limbic structures after probiotics. In the probiotics group, structural and functional changes were partly related to decreases in depressive symptoms. Limitations This study has a rather small sample size. An additional follow-up MRI session would be interesting for seeing clearer changes in the relevant brain regions as clinical effects were strongest in the follow-up. Conclusion Probiotic supplementation is suggested to prevent neuronal degeneration along the uncinate fasciculus and alter fronto-limbic rsFC, effects that are partly related to the improvement of depressive symptoms. Elucidating the neuronal mechanisms underlying probiotics' clinical effects on depression provide potential targets for the development of more precise probiotic treatments.
Content may be subject to copyright.
Journal of Aective Disorders 324 (2023) 529–538
Available online 4 January 2023
0165-0327/© 2023 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Research paper
Effects of a probiotic add-on treatment on fronto-limbic brain structure,
function, and perfusion in depression: Secondary neuroimaging ndings of
a randomized controlled trial
Gulnara Yamanbaeva
a
,
1
, Anna-Chiara Schaub
a
,
1
, Else Schneider
a
, Nina Schweinfurth
a
,
Cedric Kettelhack
a
, Jessica P.K. Doll
a
, Laura M¨
ahlmann
a
, Serge Brand
a
, Christoph Beglinger
b
,
Stefan Borgwardt
c
, Undine E. Lang
a
, Andr´
e Schmidt
a
,
*
a
University of Basel, Department of Psychiatry (UPK), Basel, Switzerland
b
Department of Research, St. Clara Hospital, Basel, Switzerland
c
Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
ARTICLE INFO
Keywords:
Multi-strain probiotics
Depression
Microbiota-gut-brain axis
Uncinate fasciculus
Fronto-limbic network
Multimodal neuroimaging
ABSTRACT
Background: Probiotics are suggested to improve depressive symptoms via the microbiota-gut-brain axis. We have
recently shown a benecial clinical effect of probiotic supplementation in patients with depression. Their un-
derlying neural mechanisms remain unknown.
Methods: A multimodal neuroimaging approach including diffusion tensor imaging, resting-state functional MRI,
and arterial spin labeling was used to investigate the effects of a four-weeks probiotic supplementation on fronto-
limbic brain structure, function, and perfusion and whether these effects were related to symptom changes.
Results: Thirty-two patients completed both imaging assessments (18 placebo and 14 probiotics group). Pro-
biotics maintained mean diffusivity in the left uncinate fasciculus, stabilized it in the right uncinate fasciculus,
and altered resting-state functional connectivity (rsFC) between limbic structures and the temporal pole to a
cluster in the precuneus. Moreover, a cluster in the left superior parietal lobule showed altered rsFC to the
subcallosal cortex, the left orbitofrontal cortex, and limbic structures after probiotics. In the probiotics group,
structural and functional changes were partly related to decreases in depressive symptoms.
Limitations: This study has a rather small sample size. An additional follow-up MRI session would be interesting
for seeing clearer changes in the relevant brain regions as clinical effects were strongest in the follow-up.
Conclusion: Probiotic supplementation is suggested to prevent neuronal degeneration along the uncinate
fasciculus and alter fronto-limbic rsFC, effects that are partly related to the improvement of depressive symp-
toms. Elucidating the neural mechanisms underlying probioticsclinical effects on depression provide potential
targets for the development of more precise probiotic treatments.
1. Introduction
There is accumulating evidence showing the relevance of the gut
microbiota on the brain and its link to psychiatric disorders such as
depression (Cryan and Dinan, 2012). This connection is described as the
microbiota-gut-brain (MGB) axis and preclinical studies provided rst
insights on the specic underlying mechanisms in depression (Bravo
et al., 2011; Kelly et al., 2016; Zheng et al., 2016). Deciphering MGB
interactions may allow to detect novel biological targets for the
development of more efcient and tailored antidepressant treatments
(Cryan et al., 2019; Evrensel and Tarhan, 2021). Probiotics, which are
living bacteria that have benecial effects on the host (Markowiak and
´
Sli˙
zewska, 2017), are suggested to alleviate depressive symptoms by
modifying the MGB axis via the microbiome. Preclinical and clinical
studies found rst supporting results of benecial effects of probiotics on
depressive symptoms (Akkasheh et al., 2016; Goh et al., 2019; Huang
et al., 2016; Johnson et al., 2021; Ng et al., 2018; Yong et al., 2020). In a
recent study of our group, we found an antidepressant effect of a
* Corresponding author at: University of Basel, Department of Psychiatry (UPK), Wilhelm Klein-Strasse 27, 4002 Basel, Switzerland.
E-mail address: andre.schmidt@unibas.ch (A. Schmidt).
1
Shared rst authors.
Contents lists available at ScienceDirect
Journal of Affective Disorders
journal homepage: www.elsevier.com/locate/jad
https://doi.org/10.1016/j.jad.2022.12.142
Received 29 July 2022; Received in revised form 21 December 2022; Accepted 25 December 2022
Journal of Aective Disorders 324 (2023) 529–538
530
probiotic supplement in a sample of patients with current depressive
episodes (Schaub et al., 2022). Notably, this clinical improvement was
accompanied by increased abundance of the genus Lactobacillus.
Only few MRI studies have investigated effects of probiotics on brain
markers. A rst study, which could not nd any benecial behavioral
changes after a four-week probiotic supplementation in healthy partic-
ipants, showed reduced responses to emotional faces in a network
including the insula, periaqueductal gray and somatosensory cortex
(Tillisch et al., 2013). These results were supported by resting-state
functional connectivity (rsFC) analysis showing altered midbrain con-
nectivity after probiotics in healthy women. Further effects of probiotics
on brain functioning in healthy subjects were investigated in an analysis
on resting-state networks, showing decreased connectivity after a four-
week probiotic intervention in the default mode network, visual
network and a middle/superior frontal network compared to placebo
(Bagga et al., 2019). Conversely, increased rsFC was present in a salience
network including the cingulate cortex and precuneus. Notably, these
effects were not shown in brain structures measured with diffusion
tensor imaging (DTI), where only insignicant increases of fractional
anisotropy (FA) in the cingulum and the precuneus were observed.
Generally, on the neural level, patients with depression express ab-
normalities in both brain structure and function within a fronto-limbic
network (Drevets et al., 2008a; Fitzgerald et al., 2008; Liao et al.,
2013; van Velzen et al., 2020). One relevant associative white matter
tract in depression is the uncinate fasciculus (UF), which connects parts
of the limbic system including the temporal pole, subcallosal cortex,
cingulate gyrus, and amygdala (Catani et al., 2013; Morgane et al.,
2005) with inferior regions of the frontal lobe such as the orbitofrontal
cortex (OFC). Microstructural integrity abnormalities in the UF play a
role in the pathophysiology of depression (van Velzen et al., 2020) as
lower FA (LeWinn et al., 2014) and higher mean diffusivity (MD) (van
Velzen et al., 2020) have been reported along the UF in depressed pa-
tients compared to healthy controls. Relations between structural and
functional abnormalities in regions related to the UF in depression have
been described, showing reduced white matter integrity, expressed as
reduced FA along the UF, and increased task-based functional connec-
tivity between the subgenual anterior cingulate cortex (ACC) and the
medial-temporal lobe, changes that were related to the severity of
depressive symptoms (de Kwaasteniet et al., 2013). Also, increased ac-
tivity in limbic regions such as the amygdala, hippocampus, ventral
striatum (Anand et al., 2005; Mayberg et al., 1999), and the insula (Sliz
and Hayley, 2012), have been described compared to healthy controls as
well as decreased activity in various cortical areas, including sub-
divisions of the ACC, OFC, and dorsolateral prefrontal cortex (Mayberg
et al., 1999). However, divergent results on OFC connectivity depending
on its subregions have been reported, showing increased rsFC in the
lateral OFC connecting to the precuneus, posterior cingulate cortex and
angular gyrus in depressed patients, and decreased rsFC from the medial
OFC to the medial temporal lobe (Rolls et al., 2020). The subgenual ACC
has shown decreased gray matter volume and elevated metabolic ac-
tivity in patients with depression using positron emission tomography
(PET), which appeared to be reversed by antidepressant therapies
(Drevets et al., 2008b). Furthermore, regional cerebral blood ow (CBF)
was reduced in the ACC and bilateral parahippocampal areas and
increased in frontoparietal and striatal regions in depressed patients
using arterial spin labeling (ASL) (Vasic et al., 2015). In chronic
depressed patients, however, a hyperperfusion in the bilateral subgenual
ACC, left dorsomedial prefrontal cortex, left ACC and left subcortical
areas was found (Duhameau et al., 2010). Antidepressant treatment
with selective serotonin reuptake inhibitors and amesergide was related
to increased CBF in the left and mid ACC, left superior temporal gyrus,
and OFC as measured with single photon emission computed tomogra-
phy (SPECT) (Vlassenko et al., 2004). Another study found normaliza-
tions of reduced CBF in parieto/cerebellar regions after six-weeks
antidepressant therapy and decreased CBF in frontal regions after two
years (Kohn et al., 2008).
Here, we used a multimodal neuroimaging approach to investigate
whether probiotic supplementation over four weeks in depressed pa-
tients altered fronto-limbic network structure, function, and perfusion
by using DTI, resting-state functional MRI (fMRI), and ASL. Given that
probiotic supplementation ameliorated depressive symptoms in the
same sample (Schaub et al., 2022), we tested whether neural effects
were related to the benecial clinical response. We hypothesized that
probiotics would increase the structural integrity of the UF as expressed
by increasing FA and decreasing MD compared to placebo. We further
expected effects of probiotics on rsFC and CBF in regions along the UF,
specically increases in frontal areas and decreases in limbic regions
such as the amygdala. In addition, we predicted that the increased
structural integrity of the UF, as well as function and perfusion changes
in fronto-limbic regions are related to decreases of depressive symptoms.
2. Methods
This is a secondary analysis of a randomized controlled trial (RCT,
identier NCT02957591). Clinical and gut microbiota analyses have
previously been published (Schaub et al., 2022). Here we present nd-
ings of secondary outcomes such as DTI, resting-state fMRI and ASL.
2.1. Study sample
Patients with depressive episodes were recruited at the University
Hospital of Psychiatry (UPK), Basel, Switzerland and gave written
informed consent before participating in the study. The study was
approved by the local ethics committee (Ethikkommission Nordwest-
und Zentralschweiz). All patients were within inpatient care and
received treatment as usual (TAU) for depression. Detailed inclusion and
exclusion criteria are presented in the Supplementary material. The
initial sample size of 30 patients per group was calculated for the main
clinical outcome measure of this RCT as reported elsewhere (Schaub
et al., 2022). Out of 50 patients with baseline MRI data, 35 completed
the MRI assessment after the study intervention. As three patients were
<65 % compliant (Cramer and Rosenheck, 1998), they were excluded
from the imaging analysis resulting in a nal sample of 32 patients for
resting-state fMRI and ASL analyses (Supplementary Fig. 1). Analysis of
DTI data was conducted with 31 patients since one patient aborted the
MRI session before the DTI sequence. Medication was tracked over the
study period and summarized as dened daily dose (DDD) for antide-
pressants and antipsychotics separately. An overview of the study
sample characteristics is presented in Table 1.
Table 1
Study sample characteristics.
Probiotics
N =14
Placebo
N =18
Between-group
comparison
Demographics
Age (years), mean (SD) 38.36
(10.87)
36.83
(10.15)
t(27.08) =0.40, p
=.69
Sex, male/female 4/10 10/8
χ
2
(1) =1.36, p =
.24
Antidepressants (DDD), mean
(SD)
1.92 (1.45) 1.78
(0.93)
t(20.93) =0.31, p
=.76
Antipsychotics (DDD), mean
(SD)
0.13 (0.21) 0.20
(0.27)
W =120, p =.83
a
HAM-D
Baseline, sum score, mean (SD) 19.54
(4.89)
16.83
(4.37)
W =171.5, p =
.09
Change score: baseline -
postintervention, mean (SD)
10.68
(4.62)
8.33
(5.71)
W =163.5, p =
.16
Change score: baseline - follow-
up, mean (SD)
13.96
(4.46)
8.26
(4.40)
W =185.5, p <
.01**
Notes. DDD =dened daily dose, HAM-D =Hamilton Depression Rating Scale.
a
Non-parametric test since not normally distributed.
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
531
2.2. Study design and intervention
In this double-blind RCT, subjects were allocated to two different
study groups and received either a probiotic supplementation or a pla-
cebo for 31 days in addition to TAU. The probiotics consisted of eight
different strains such as Streptococcus thermophilus NCIMB 30438, Bi-
dobacterium breve NCIMB 30441, Bidobacterium longum NCIMB 30435
(Re-classied as B. lactis), Bidobacterium infantis NCIMB 30436 (Re-
classied as B. lactis), Lactobacillus acidophilus NCIMB 30442, Lactoba-
cillus plantarum NCIMB 30437, Lactobacillus paracasei NCIMB 30439,
Lactobacillus delbrueckii subsp. Bulgaricus NCIMB 30440 (Re-classied as
L. helveticus). The daily dose contained 900 billion CFU/day that could
be mixed with any cold, noncarbonated drink. In the control group,
participants took a placebo, which contained maltose but no bacteria
and was indistinguishable in color, shape, size, smell, and taste from the
probiotic supplement. Further details on the intervention are reported in
the Supplementary material. Patients received a standardized diet con-
taining stable amounts of bers and starch during inpatient setting.
Depressive symptoms were measured with the 17-items Hamilton
Depression Rating Scale (HAM-D) (Hamilton, 1967). Clinical and MRI
assessments were conducted before and after the four-weeks interven-
tion. Clinical data was also assessed at a four-week follow-up after the
end of the intervention.
2.3. MRI acquisition
DTI, resting-state fMRI and ASL data were acquired with a 3T
Siemens Magnetom Prisma Scanner before and after the probiotic
intervention The diffusion-weighted images were sampled with a RF-
pulsed gradient-spin-echo sequence during resting-state activity (inter-
leaved, FoV: 256 mm
2
, TR =7500 ms, TE =71 ms, slice thickness: 2
mm, 2 ×2 ×2 mm voxel size, EPI factor: 128, b-value =800 s/mm
2
including b0 images, i.e. a non-DWIs, the b =0 s/mm
2
). The protocol
comprised 64 diffusion directions. RsFC was assessed with an 8.4 min
echo-planar T2* weighted imaging sequence (interleaved, FoV: 256
mm
2
, TR =3000 ms, TE =28 ms, 3 ×3 ×3 mm voxel size). Patients
were instructed to relax but stay awake and not to think of anything
specic during the acquisition. A structural sequence was used as
anatomical reference (MPRAGE, TR =2000 ms, TE =3.37 ms, 1 ×1 ×1
mm voxel size). ASL technique was used to estimate the CBF at resting-
state (Chen et al., 2015). Data were sampled with a RF-pulsed fast
gradient echo sequence (interleaved, FoV: 256 mm
2
, TR =3000 ms, TE
=12 ms, slice thickness: 4 mm, voxel size: 4 ×4 ×4 mm, EPI factor: 64,
20 transversal (axial) slices, PICORE Q2T perfusion mode, inversion
time: 1800 ms, bolus duration: 700 ms).
2.4. Structural properties analysis
DTI data was preprocessed using a standalone version of ExploreDTI
(Version 8.3, 2009) (Jeurissen et al., 2019; Tournier et al., 2011). In
brief, we rst performed the data reconstruction (Supplementary
methods). The preprocessing included subject motion and eddy-current-
induced distortions correction. Virtual dissection of the three-
dimensional UF tract was done as described by Wakana et al. (Wakana
et al., 2007). In brief, the ‘AND gates (Supplementary methods) were
located on a coronal slice at the position where the temporal and frontal
lobes are divided, allowing a selection of only those bers from the
temporal lobe, which projected to the frontal lobe. To omit ber pro-
jections that are not considered to belong to the UF, additional ‘NOT
gates were placed when necessary (Fig. 1A). After preprocessing and
virtual dissection, FA and MD were calculated using ExploreDTI (Sup-
plementary methods).
2.4.1. Factorial ANOVA analysis
The UF tracts were dissected for all patients and parameters of the
ber tract structures were calculated. An entire set of the calculated tract
parameters (including FA and MD) was exported and further analyzed in
the software package STATISTICA (StatSoft, Inc., Tulsa, OK.: STATIS-
TICA, version 8). FA and MD parameters of the left and right UF at
baseline and post-intervention were entered in a factorial ANOVA
analysis to detect main effects of hemisphere, time and group, and their
interactions. In case of signicant main effects or interactions, post-hoc
Fig. 1. Uncinate fasciculus (UF) tract DTI trac-
tography. (A) Virtual dissection of UF. Green lines
at the coronal slice represents the ‘ANDgates for
the virtual dissection. Virtually dissected left and
right UF shown in sagittal view. (B) Fractional
anisotropy (FA) of UF. The factorial ANOVA
analysis showed a signicant main effect of group
(F =7.433, p =.007) and hemisphere (F =7.288,
p =.008). (C) Mean diffusivity (MD) of UF. The
factorial ANOVA analysis showed a signicant
hemispheric effect (F =4.988, p =.027) and
time*group interaction (F =6.303, p =.013).
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
532
testing using Tukey HSD test was performed.
2.5. Functional connectivity analysis
Resting-state fMRI data was analyzed using CONN toolbox (v19.c,
www.nitrc.org/projects/conn) (Whiteld-Gabrieli and Nieto-Castanon,
2012), which is an open-source Matlab/SPM-based software. A default
preprocessing pipeline for volume-based analyses was run including
functional realignment and unwrapping, slice-time correction, outlier
identication, segmentation and indirect normalization to MNI-space
and functional smoothing (6 mm kernel). Susceptibility distortion
correction using B0 eld maps with short and long real and imaginary
images was applied during the functional alignment and unwrapping
step. Data were denoised using linear regression of potential con-
founding effects in the BOLD signal (subject motion parameters, scrub-
bing (Power et al., 2014), noise components from white matter and
cerebrospinal areas and session effects). Furthermore, linear detrending
and temporal band-pass ltering (0.0080.09 Hz) were applied.
2.5.1. Seed-to-voxel analysis
We investigated rsFC with a seed-to-voxel approach in which rsFC
maps are created for selected seeds. The study design with two groups
(probiotics and placebo) and two time points (baseline and post-
intervention) resulted in an 2 ×2 mixed ANOVA with interaction to
compare changes in rsFC between the two study groups. ROIs or rather
seeds-of-interest were chosen based on previous results linking the
medial temporal lobe and prefrontal regions to depression (Helm et al.,
2018). As ROIs, the subcallosal cortex (also referred to as subgenual
ACC), hippocampus and amygdala as well as further regions surround-
ing the UF such as the temporal pole, insula and OFC were included
(Supplementary Fig. 2). All ROIs were dened according to the Harvard-
Oxford cortical and subcortical structural atlas (Desikan et al., 2006).
First, all seeds were taken simultaneously in the model to avoid prob-
lems of multiple testing. Post-hoc tests were computed to determine
which selected seeds were responsible for the effects. Results were
corrected using false discovery rate (FDR) procedures and considered as
signicant with a p-FDR <0.05. When baseline differences in rsFC be-
tween groups occurred (two sample t-test), an ANCOVA with baseline
beta values as covariate, group as predictor and post-intervention beta
values as dependent variable was conducted in R. Beta values represent
Fisher-transformed correlations coefcients and were extracted from
CONN.
2.6. Blood perfusion analysis
ASL image preprocessing was performed using FSL6.0.0 (Smith et al.,
2004) and custom-written in Bash Unix shell (an interactive command
and scripting language that provides a command line user interface for
Unix-like operating systems, https://www.gnu.org/software/bash/)
routines, with help of FSL FEAT toolbox commands (Woolrich et al.,
2001). Preprocessing included motion correction, calibration of rst
volume against remaining volume as tag-control pairs, relative CBF
maps calculation, T1 structural scan reorientation and brain extraction
(Smith, 2002), calibrated volume image registration to T1, absolute CBF
calculation, thresholding, registration of thresholded images to T1 and
masking it, registration and normalization to MNI-space.
2.6.1. CBF ROI analysis
In line with the resting-state fMRI analysis, a CBF ROI analysis was
performed for the following regions: subcallosal cortex, hippocampus,
amygdala, insula, OFC and temporal pole (Supplementary Fig. 2). Ab-
solute CBF values were extracted and mean CBF for each patient was
calculated across each ROI separately (in ml/100 g/min). Mean CBF
estimates of each ROI were analyzed in STATISTICA (StatSoft, Inc.,
Tulsa, OK.: STATISTICA, version 8) using a factorial ANOVA analysis to
detect main effects of group, hemisphere and time, as well as their
interactions. In case of signicance, post-hoc Tukey HSD tests were
performed (Supplementary methods).
2.7. Correlation analysis
To investigate relationships between neuroimaging results and
depressive symptoms, parameters of all three imaging modalities were
linked to depressive symptoms using non-parametric Spearman rank
correlation. HAM-D change scores (baseline post-intervention) were
correlated with FA and MD changes in the UF, beta values of seeds with
signicant interactions in rsFC and region-wise mean CBF estimates
within the probiotic and placebo group.
3. Results
3.1. Participants
Sample characteristics such as age, sex and medication were equally
distributed across the placebo and probiotic group (Table 1) and were
therefore not included in the analyses. Depressive symptoms decreased
in both study groups but changes were not statistically different between
the two groups from baseline to post-intervention. In the longer period
from the baseline to follow-up assessment, however, patients in the
probiotics group had a higher symptom decrease than patients in the
placebo group (Table 1).
3.2. Structural properties
3.2.1. Fractional anisotropy
The analysis of FA showed signicant main effects of group (F =
7.433, p =.007) and hemisphere (F =7.288, p =.008). Post-hoc testing
showed signicantly higher FA values in the probiotics (mean FA ±
SEM =0.428 ±0.003, p =.008) compared to the placebo group (0.417
±0.003) and signicantly higher FA in the left (0.427 ±0.003, p =
.007) than right UF (0.416 ±0.003) over both timepoints (Fig. 1B). No
signicant hemisphere*group (F =0.326, p =.569), time*hemisphere
(F =0.433, p =.512), time*group (F =1.324, p =.252), and time*-
hemisphere*group interactions (F =0.004, p =.947) were found.
3.2.2. Mean diffusivity
The analysis of MD revealed a signicant hemispheric effect (F =
4.988, p =.027) and time*group interaction (F =6.303, p =.013). No
signicant effects of time (F =2.253, p =.136), group (F =1.313, p =
.254) or time*hemisphere*group interaction (F =0.057, p =.812) were
found. Subsequent post-hoc testing showed signicantly higher MD in
the right UF (0.838E3 ±0.004E3, p =.022) compared to the left UF
(0.828E3 ±0.003E3) over both groups and timepoints. Further-
more, in the placebo group, right UF MD values were signicantly
increased at the post-intervention (0.845E3 ±0.005E3, p =.018)
compared to the baseline assessment (0.826E3 ±0.004E3), whereas
MD at post-intervention remained stable in the probiotics group (0.828
±0.004E3, p =.904) compared to baseline (0.832E3 ±0.005E3)
(Fig. 1C).
3.3. Functional connectivity
The analysis including all seeds of interest showed a signicant
interaction in two different clusters located in the precuneus (x = +16 y
= 52 z = +18, 89 voxels, p-FDR =0.0068, Fig. 2) and the left superior
parietal lobule extending to the left posterior supramarginal gyrus (x =
44 y = 42 z = +50, 61 voxels, p-FDR =0.0266). Post-hoc tests of the
precuneus cluster revealed signicant time*group interactions in the
subcallosal cortex, bilateral amygdala and hippocampus, and left tem-
poral pole (Fig. 2AF). In these regions, the probiotics group showed
rsFC increases, whereas the placebo group showed rsFC decreases.
Baseline rsFC was only identical in both study groups in the left temporal
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
533
pole. Subsequent models correcting for baseline differences in all other
regions showed that the group effect was signicant in the right (F(1) =
4.24, p <.05) and left hippocampus (F(1) =6.1, p <.05), and left
amygdala (F(1) =10.31, p <.01), but not in the subcallosal cortex (F(1)
=1.63, p =.21) and right amygdala (F(1) =3.46, p =.07).
The cluster in the left superior parietal lobule (Fig. 3) had signicant
time*group interactions in rsFC originating from the subcallosal cortex,
left OFC, right amygdala and left hippocampus. Besides an increase of
rsFC in the left OFC (Fig. 3B), rsFC decreased in all other regions in the
probiotics group compared to placebo (Fig. 3A, C, D). Baseline rsFC was
not identical in the study groups in the subcallosal cortex rsFC but when
controlling for differences in baseline rsFC values, the group effect was
signicant (F(1) =8.09, p <.01).
3.4. Blood perfusion
The factorial ANOVA analysis showed no signicant group (F =
2.654, p =.109) and time (F =0.171, p =.681) effects, and no
group*time interaction (F =0.034, p =.854) in the subcallosal cortex.
There was also no signicant time (F =3.087, p =.081) and hemispheric
(F =1.051, p =.307) effect in the hippocampus, while the group effect
was signicant (F =5.415, p =.023). The placebo group demonstrated
signicantly higher mean CBF in the hippocampus (18.856 ±0.380 ml/
100 g/min, p =.003) compared to the probiotics group (17.099 ±
0.444 ml/100 g/min) across both time points. A signicant time effect
was found in the amygdala (F =7.198, p =.007); at the post-
intervention the mean CBF was signicantly higher (mean CBF =
14.642 ±0.421 ml/100 g/min, p =.007) compared to baseline (13.070
Fig. 2. Changes in resting-state functional connectivity (rsFC) over time for six connections with signicant interactions from (A) subcallosal cortex, (B) temporal
pole left, (C) hippocampus right, (D) hippocampus left, (E) amygdala right, (F) amygdala left to the cluster in the precuneus. The connection from subcallosal cortex
and right amygdala were not signicant after baseline correction. *Baseline differences between groups p <.05.
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
534
±0.374 ml/100 g/min) (Supplementary Fig. 3). No other effects and
interactions were signicant in the amygdala, left and right insula, left
and right OFC, left and right temporal pole.
3.5. Relations to depressive symptoms
In the left UF within the probiotics group, changes in FA correlated
negatively with HAM-D changes (rho = 0.662, p =.010) (Fig. 4A),
indicating greater HAM-D decreases with greater FA increases. On the
other hand, changes in MD in the left UF correlated positively with
HAM-D changes (rho =0.701, p =.005) (Fig. 4B), indicating greater
HAM-D decreases with greater MD decreases. Within the probiotics
group, FA changes correlated negatively with MD changes in the left UF
(rho = 0.587, p =.027) (Fig. 4C) and in the right UF (rho = 0.626, p
=.017) (Fig. 4D). No signicant correlations were found in the placebo
group.
Changes in rsFC from the right amygdala to the cluster in the supe-
rior parietal lobule correlated positively with HAM-D changes in the
probiotics group (rho =0.57, p =.032, Fig. 4E), indicating greater
symptom decreases with greater rsFC decreases. However, after cor-
recting for multiple testing, the correlation was not signicant anymore
(p-Bonferroni =0.12). Other correlations in the probiotics group and all
correlations in the placebo group were low and did not reach signi-
cance (Supplementary Table 1).
Changes of absolute CBF did not correlate with HAM-D changes in
any assessed region (Supplementary Table 2).
4. Discussion
A multi-modal neuroimaging approach was used to study the effects
of a probiotic add-on supplement on fronto-limbic network structure,
function, and blood perfusion in depressed patients. As rst major result,
we found an increased MD in the right UF within the placebo group with
no signicant change within the probiotics group, i,e, MD remained
stable. This stabilization of MD within the probiotic group positively
correlated with changes in depressive symptoms. Secondly, we found
changes over time between the study groups in rsFC between specic
fronto-limbic seed regions and a cluster in the precuneus and another in
the left superior parietal lobule. Decreases in right amygdala-superior
parietal lobule rsFC correlated positively with decreases in depressive
symptoms in the probiotics group.
Since lower FA and higher MD are attributed to a loss of micro-
structural integrity and neurodegeneration (Thomalla et al., 2004; van
Velzen et al., 2020), probiotics might prevent further neurodegeneration
given the fact that in the right UF the MD was signicantly increased in
the placebo group whereas in the probiotics group it remained stable, i.
e. the greater MD is the worse the neuronal integrity is preserved.
Notably, this effect was linked to the improvement of depressive
symptoms. In support, albeit not signicant, the left UF MD in the pro-
biotics group even showed a decrease, while it further increased in the
placebo group. FA has been attributed to the nerve ber arrangements,
axonal integrity, and the degree of axonal myelination (Basser and
Jones, 2002), whereas MD is considered to indicate atrophy and broad
cellular damages (Hutchinson et al., 2018; Immonen et al., 2009; Lai-
tinen et al., 2015; Pierpaoli et al., 1996; Putten et al., 2005). Interest-
ingly, decreased FA and increased MD are associated with neuronal
degeneration (Thomalla et al., 2004). Taken the abovementioned facts
together, our MD ndings suggest that the benecial clinical effect of
probiotic supplementation in depression (Schaub et al., 2022) may
partially be mediated by counteracting neuronal degeneration along the
Fig. 3. Changes in resting-state functional connectivity (rsFC) over time for four connections with signicant interactions from (A) subcallosal cortex, (B) orbito-
frontal cortex right, (C) hippocampus left (D) amygdala right to the cluster in the superior parietal lobule. *Baseline differences between groups p <.05.
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
535
UF.
FA and MD are typically inversely correlated since myelination,
which facilitates directionality (i.e. increasing FA), also mediates a re-
striction to the overall movement (i.e. decreasing MD) (Green et al.,
2010; Niogi and Mukherjee, 2010). In our study, the signicant negative
correlation between FA and MD changes in both left and right UF within
the probiotics group indicates a subtle increase in the FA that is asso-
ciated with the reduction of depressive symptoms at least in the left UF
even though the time*group interaction in the FA was not signicant.
Taking into account that none of these correlations were found in the
placebo group, we propose that probiotics might not only prevent the
worsening of microstructural properties of the UF but also induce the
recovery of them revealed in the strong inverse correlation between FA
and MD and their association with the improvement of the depressive
symptoms.
Time*group interactions in rsFC from specied fronto-limbic regions
linked to the UF showed signicant clusters in the precuneus and the
superior parietal lobule, indicating that rsFC increased in connections to
the precuneus and decreased in connections to the superior parietal
lobule after probiotics intake. Decreased rsFC between the precuneus
and the subcallosal cortex has previously been shown in medication-
naive rst-episode adolescents with depression (Connolly et al., 2013).
Thus, the increase in this connection we found in the probiotics group
compared to placebo, might indicate a normalization of this connection
due to the probiotics. However, our result was not signicant after
controlling for baseline differences between the study groups.
Previously, increased rsFC has been found from the precuneus to the
hippocampus, as well as to different frontal regions and the middle
temporal gyrus in medication-naïve patients with rst-episode depres-
sion (Peng et al., 2015). This study also showed negative associations
between symptom severity and rsFC of the precuneustemporo/parietal
junction as well as a positive association between symptom severity and
the precuneusdorsomedial prefrontal cortex rsFC. In contrast to our
hypothesis, we could not nd any correlation between precuneus rsFC
and symptom changes over time. Generally, the precuneus is structur-
ally and functionally relevant in depression, showing volume loss
(Grieve et al., 2013) and altered rsFC as part of the default-mode
network (DMN) (Greicius et al., 2007; Kaiser et al., 2015; Li et al.,
2018). Previous results showed decreasing rsFC between lateral OFC and
precuneus in patients receiving medication while an increased rsFC has
been observed in unmedicated depressed patients (Cheng et al., 2018).
We could not nd a signicant time*group interaction effect in this OFC-
precuneus connection which might be explained with a general anti-
depressant effect of the medication that is independent from the pro-
biotics and was not of interest in this analysis.
In our second cluster located in the left superior parietal lobule, four
Fig. 4. Correlations of changes in depressive symp-
toms (HAM-D) and changes in neuroimaging mea-
surements. (A) Correlation between changes in
depressive symptoms and changes in fractional
anisotropy (FA) of the left uncinated fasciculus (UF)
in the probiotics group. (B) Correlation between
changes in depressive symptoms and changes in mean
diffusivity (MD) of the left UF in the probiotics group.
(C) Correlation between changes in FA and MD of the
left UF within the probiotics group. (D) Correlation
between changes in FA and MD of the right UF within
the probiotics group. (E) Correlation between changes
in depressive symptoms and changes in resting-state
functional connectivity (rsFC) from right amygdala
to the cluster in the left superior parietal lobule.
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
536
of our seeds showed signicant interactions including the subcallosal
cortex, left OFC, right amygdala and left hippocampus. The superior
parietal lobule is involved in cognition including visually guided actions
in spatial processing (Sack, 2009) and it is also supposed to rearrange
information as part of the working memory (Koenigs et al., 2009).
However, the role of the superior parietal lobule in depression is not well
elucidated yet; its involvement has mostly been investigated as part of
the DMN that shows a hyperconnectivity in depression (Kaiser et al.,
2015). First-episode, drug-naive depressed patients exhibited a signi-
cantly lower magnetization transfer ratio (a quantitative measure of the
macromolecular structural integrity of brain tissue, to identify bio-
physical alterations) in the left superior parietal lobule compared to
healthy controls, which may be associated with the attentional and
cognitive dysfunction in depressed patients (Chen et al., 2016; Zhang
et al., 2018). The increasing rsFC from the superior parietal lobule to the
left OFC might be linked with cognitive functioning which has been
shown previously to be improved after probiotics treatment in depres-
sion (Kim et al., 2021). Furthermore, the OFC plays a special role in
decision making and emotions (Rolls et al., 2020; Wallis, 2007). In
contrast, the connectivity from subcortical regions (amygdala, hippo-
campus) and the subcallosal cortex to the cluster in the superior parietal
lobule decreases after the probiotics. The detected positive correlation in
the right amygdala-superior parietal lobule connectivity indicates that
greater rsFC decreases are associated with greater treatment response in
the probiotics but not in the placebo group. The decrease in this
connection of cognition-emotion relevant structures might be linked to
normal affect/reduced depressive symptoms. As altered rsFC from
amygdala to various brain regions has previously been shown in anti-
depressant treatments, it has been suggested that amygdala rsFC might
be a potential biomarker in depression (Connolly et al., 2017). Future
research could deepen the understanding of the probioticseffect on this
identied connection by additionally investigating working memory
functioning.
In depression, there is increased blood perfusion in the subgenual
ACC and lateral OFC, which normalizes in remission (Drevets, 2007).
Abnormalities in CBF of depressed patients were found in the dorsolat-
eral prefrontal cortex, rostral and ventral ACC, amygdala, and basal
ganglia (Bonne et al., 2003; Drevets, 2000; Mayberg et al., 1999): most
of the studies report reduced perfusion in these regions (Klemm et al.,
1996; Mayberg et al., 1994; Videbech, 2000), although contradicting
results have also been described reporting an increase in activity (Abou-
Saleh et al., 1999; Maes et al., 1993). In our study, we found a signicant
CBF increase over time in the amygdala in both groups, but the CBF
changes in the amygdala did not correlate with HAM-D changes in both
study groups. Taking into account the absent time*group interaction,
these ndings suggest that probiotics did not affect the blood perfusion
in the amygdala, or that the four-weeks probiotics intervention is not
sufcient long to see an effect. Moreover, the prevalent impact of the
antidepressant medication on the levels of the blood perfusion in the
assessed regions might be a reason for the absence of a reduced diffu-
sion. Nevertheless, it was reported that even a short-term antidepressant
therapy in depressed patients normalizes elevated CBF levels in the
amygdala (Godlewska et al., 2012; Smith and Cavanagh, 2005; Solleveld
et al., 2020) which is not consistent with the amygdala CBF elevation in
our patients who took antidepressant medication. However, there is a
certain contrariety in ndings related to the level of perfusion in limbic/
paralimbic regions including the amygdala (Drevets, 2002; Mayberg,
2003). More consistent single-photon emission computed tomography
in patients with MDD showed decreased perfusion (hypoperfusion) in
the dorsolateral prefrontal cortex, rostral and ventral anterior cingulate
cortex, basal ganglia, and amygdala in depressed patients (Smith and
Cavanagh, 2005). Also, the abnormal elevation in CBF (hyperperfusion)
in the amygdala has been associated with the severity of depression
(Drevets et al., 1992). Thus, the CBF elevation in our patients which was
not associated with changes in depressive symptoms nor with the pro-
biotic treatment must be interpreted carefully. In other words, to
determine whether after the antidepressants and probiotics the dynamic
of CBF level in the amygdala is positive (i.e. normalized) or negative, we
have to know whether initially there was a hyper- or hypoperfusion.
Further studies of this aspect with the larger study sample and
involvement of the healthy control would be required.
There are some limitations in this study that should be addressed in
future research. First, our sample size was moderate due to the multi-
modal complexity of the study design implying the participation of the
patients in two neuroimaging sessions and an intervention period of four
weeks, which produced a drop-out rate of >20 %. Also, some study
participants were excluded due to low compliance. Second, to see more
prominent changes, it could be advantageous to have a third MRI session
after eight weeks of intervention as the clinical effect was strongest at
the follow-up assessment (Schaub et al., 2022). The protective effect of
probiotics on the UF microstructural properties with their possible
consequent recovery could be more apparent then. Nevertheless, the
probiotic-induced prevention of further neurodegeneration along the UF
within the relatively short four-weeks intervention period and its rela-
tion to the improvement of depressive symptoms is intriguing.
This post-hoc analysis of an RCT explored the underlying neural
mechanisms of a probiotic add-on intervention in depressed patients.
The results suggest that the probiotics affect brain structure and function
in the fronto-limbic network and these effects are partly associated with
the decrease in depressive symptoms. Understanding the neural and
further biological mechanisms of probiotic supplementation along the
MGB axis on depressive symptoms in patients across the affective
spectrum may help to foster stratied treatment guidance by parsing
heterogeneity in the treatment responses. Large-scale studies may
further elucidate the precise mechanisms of probiotic supplementation
along the MGB axis including neural, immune, metabolic, and endocrine
pathways.
Conict of interest
The authors report no biomedical nancial interests or potential
conicts of interest.
Acknowledgments
The study was supported by the Gertrud Thalmann Foundation of the
University Psychiatric Clinics (UPK) Basel (SBo, UEL), the K¨
ampf-
B¨
otschi Foundation (UEL), the research fund junior researchers from
University of Basel (Appln 3MS1041, AS), the research fund of the UPK
Basel (AS) and the Stiftung zur F¨
orderung der gastroenterologischen und
allgemeinen klinischen Forschung sowie der medizinischen Bildaus-
wertung (AS). MENDES S.A., Switzerland, supplied the investigational
medicinal product. The funders had no role in the design and conduct of
the study; collection, management, analysis, and interpretation of the
data; preparation, review, or approval of the manuscript; and decision to
submit the manuscript for publication.
AS had full access to all the data in the study and takes responsibility
for the integrity of the data and the accuracy of the data analysis. LM,
CB, SBo, AS, and UEL designed the study. GY, ACS, ES, NS, CK, JPKD,
and AS contributed to the acquisition, analysis, or interpretation of data.
Statistical analyses were performed by GY and ACS. Administrative,
technical, or material support was provided by LM, SBr, SBo, AS, and
UEL. GY, ACS, and AS drafted the manuscript. All authors critically
reviewed the article and approved the nal manuscript.
T1-weighted structural MRI data of 50 patients at baseline were
recently published in the paper Neural mapping of anhedonia across
psychiatric diagnoses: A transdiagnostic neuroimaging analysis
(Schaub et al., 2021).
The authors report no biomedical nancial interests or potential
conicts of interest.
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
537
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.jad.2022.12.142.
References
Abou-Saleh, M.T., Al Suhaili, A.R., Karim, L., Prais, V., Hamdi, E., 1999. Single photon
emission tomography with 99m tc-HMPAO in Arab patients with depression.
J. Affect. Disord. 55, 115123. https://doi.org/10.1016/S0165-0327(98)00209-2.
Akkasheh, G., Kashani-Poor, Z., Tajabadi-Ebrahimi, M., Jafari, P., Akbari, H.,
Taghizadeh, M., Memarzadeh, M.R., Asemi, Z., Esmaillzadeh, A., 2016. Clinical and
metabolic response to probiotic administration in patients with major depressive
disorder: a randomized, double-blind, placebo-controlled trial. Nutrition 32,
315320. https://doi.org/10.1016/j.nut.2015.09.003.
Anand, A., Li, Y., Wang, Y., Wu, J., Gao, S., Bukhari, L., Mathews, V.P., Kalnin, A.,
Lowe, M.J., 2005. Activity and connectivity of brain mood regulating circuit in
depression: a functional magnetic resonance study. Biol. Psychiatry 57, 10791088.
https://doi.org/10.1016/j.biopsych.2005.02.021.
Bagga, D., Aigner, C.S., Reichert, J.L., Cecchetto, C., Fischmeister, F.Ph.S., Holzer, P.,
Moissl-Eichinger, C., Sch¨
opf, V., 2019. Inuence of 4-week multi-strain probiotic
administration on resting-state functional connectivity in healthy volunteers. Eur. J.
Nutr. 58, 18211827. https://doi.org/10.1007/s00394-018-1732-z.
Basser, P.J., Jones, D.K., 2002. Diffusion-tensor MRI: theory, experimental design and
data analysis - a technical review. NMR Biomed. 15, 456467. https://doi.org/
10.1002/nbm.783.
Bonne, O., Louzoun, Y., Aharon, I., Krausz, Y., Karger, H., Lerer, B., Bocher, M.,
Freedman, N., Chisin, R., 2003. Cerebral blood ow in depressed patients: a
methodological comparison of statistical parametric mapping and region of interest
analyses. Psychiatry Res. Neuroimaging 122, 4957. https://doi.org/10.1016/
S0925-4927(02)00103-8.
Bravo, J.A., Forsythe, P., Chew, M.V., Escaravage, E., Savignac, H.M., Dinan, T.G.,
Bienenstock, J., Cryan, J.F., 2011. Ingestion of Lactobacillus strain regulates
emotional behavior and central GABA receptor expression in a mouse via the vagus
nerve. Proc. Natl. Acad. Sci. 108, 1605016055. https://doi.org/10.1073/
pnas.1102999108.
Catani, M., DellAcqua, F., Thiebaut de Schotten, M., 2013. A revised limbic system
model for memory, emotion and behaviour. Neurosci. Biobehav. Rev. 37,
17241737. https://doi.org/10.1016/j.neubiorev.2013.07.001.
Chen, J.J., Jann, K., Wang, D.J.J., 2015. Characterizing resting-state brain function using
arterial spin labeling. Brain Connect. 5, 527542. https://doi.org/10.1089/
brain.2015.0344.
Chen, Z., Peng, W., Sun, H., Kuang, W., Li, W., Jia, Z., Gong, Q., 2016. High-eld
magnetic resonance imaging of structural alterations in rst-episode, drug-naive
patients with major depressive disorder. Transl. Psychiatry 6. https://doi.org/
10.1038/tp.2016.209 e942-e942.
Cheng, W., Rolls, E.T., Qiu, J., Yang, D., Ruan, H., Wei, D., Zhao, L., Meng, J., Xie, P.,
Feng, J., 2018. Functional connectivity of the precuneus in unmedicated patients
with depression. Biol. Psychiatry 3, 10401049. https://doi.org/10.1016/j.
bpsc.2018.07.008.
Connolly, C.G., Wu, J., Ho, T.C., Hoeft, F., Wolkowitz, O., Eisendrath, S., Frank, G.,
Hendren, R., Max, J.E., Paulus, M.P., Tapert, S.F., Banerjee, D., Simmons, A.N.,
Yang, T.T., 2013. Resting-state functional connectivity of subgenual anterior
cingulate cortex in depressed adolescents. Biol. Psychiatry 74, 898907. https://doi.
org/10.1016/j.biopsych.2013.05.036.
Connolly, C.G., Ho, T.C., Blom, E.H., LeWinn, K.Z., Sacchet, M.D., Tymoyeva, O.,
Simmons, A.N., Yang, T.T., 2017. Resting-state functional connectivity of the
amygdala and longitudinal changes in depression severity in adolescent depression.
J. Affect. Disord. 207, 8694. https://doi.org/10.1016/j.jad.2016.09.026.
Cramer, J.A., Rosenheck, R., 1998. Compliance with medication regimens for mental and
physical disorders. PS 49, 196201. https://doi.org/10.1176/ps.49.2.196.
Cryan, J.F., Dinan, T.G., 2012. Mind-altering microorganisms: the impact of the gut
microbiota on brain and behaviour. Nat. Rev. Neurosci. 13, 701712. https://doi.
org/10.1038/nrn3346.
Cryan, J.F., ORiordan, K.J., Cowan, C.S.M., Sandhu, K.V., Bastiaanssen, T.F.S.,
Boehme, M., Codagnone, M.G., Cussotto, S., Fulling, C., Golubeva, A.V., Guzzetta, K.
E., Jaggar, M., Long-Smith, C.M., Lyte, J.M., Martin, J.A., Molinero-Perez, A.,
Moloney, G., Morelli, E., Morillas, E., OConnor, R., Cruz-Pereira, J.S., Peterson, V.L.,
Rea, K., Ritz, N.L., Sherwin, E., Spichak, S., Teichman, E.M., van de Wouw, M.,
Ventura-Silva, A.P., Wallace-Fitzsimons, S.E., Hyland, N., Clarke, G., Dinan, T.G.,
2019. The microbiota-gut-brain Axis. Physiol. Rev. 99, 18772013. https://doi.org/
10.1152/physrev.00018.2018.
de Kwaasteniet, B., Ruhe, E., Caan, M., Rive, M., Olabarriaga, S., Groefsema, M.,
Heesink, L., van Wingen, G., Denys, D., 2013. Relation between structural and
functional connectivity in major depressive disorder. Biol. Psychiatry 74, 4047.
https://doi.org/10.1016/j.biopsych.2012.12.024.
Desikan, R.S., S´
egonne, F., Fischl, B., Quinn, B.T., Dickerson, B.C., Blacker, D.,
Buckner, R.L., Dale, A.M., Maguire, R.P., Hyman, B.T., Albert, M.S., Killiany, R.J.,
2006. An automated labeling system for subdividing the human cerebral cortex on
MRI scans into gyral based regions of interest. NeuroImage 31, 968980. https://doi.
org/10.1016/j.neuroimage.2006.01.021.
Drevets, W.C., 2000. Neuroimaging studies of mood disorders. Biol. Psychiatry 48,
813829. https://doi.org/10.1016/S0006-3223(00)01020-9.
Drevets, W., 2002. Functional anatomical correlates of antidepressant drug treatment
assessed using PET measures of regional glucose metabolism. Eur.
Neuropsychopharmacol. 12, 527544. https://doi.org/10.1016/S0924-977X(02)
00102-5.
Drevets, W.C., 2007. Orbitofrontal cortex function and structure in depression. Ann. N. Y.
Acad. Sci. 1121, 499527. https://doi.org/10.1196/annals.1401.029.
Drevets, W., Videen, T., Price, J., Preskorn, S., Carmichael, S., Raichle, M., 1992.
A functional anatomical study of unipolar depression. J. Neurosci. 12, 36283641.
https://doi.org/10.1523/JNEUROSCI.12-09-03628.1992.
Drevets, W.C., Price, J.L., Furey, M.L., 2008a. Brain structural and functional
abnormalities in mood disorders: implications for neurocircuitry models of
depression. Brain Struct. Funct. 213, 93118. https://doi.org/10.1007/s00429-008-
0189-x.
Drevets, W.C., Savitz, J., Trimble, M., 2008b. The subgenual anterior cingulate cortex in
mood disorders. CNS Spectr. 13, 663681. https://doi.org/10.1017/
s1092852900013754.
Duhameau, B., Ferr´
e, J.-C., Jannin, P., Gauvrit, J.-Y., V´
erin, M., Millet, B., Drapier, D.,
2010. Chronic and treatment-resistant depression: a study using arterial spin
labeling perfusion MRI at 3Tesla. Psychiatry Res. Neuroimaging 182, 111116.
https://doi.org/10.1016/j.pscychresns.2010.01.009.
Evrensel, A., Tarhan, K.N., 2021. Emerging role of gut-microbiota-brain axis in
depression and therapeutic implication. Prog. Neuro-Psychopharmacol. Biol.
Psychiatry 106, 110138. https://doi.org/10.1016/j.pnpbp.2020.110138.
Fitzgerald, P.B., Laird, A.R., Maller, J., Daskalakis, Z.J., 2008. A meta-analytic study of
changes in brain activation in depression. Hum. Brain Mapp. 29, 683695. https://
doi.org/10.1002/hbm.20426.
Godlewska, B.R., Norbury, R., Selvaraj, S., Cowen, P.J., Harmer, C.J., 2012. Short-term
SSRI treatment normalises amygdala hyperactivity in depressed patients. Psychol.
Med. 42, 26092617. https://doi.org/10.1017/S0033291712000591.
Goh, K.K., Liu, Y.-W., Kuo, P.-H., Chung, Y.-C.E., Lu, M.-L., Chen, C.-H., 2019. Effect of
probiotics on depressive symptoms: a meta-analysis of human studies. Psychiatry
Res. 282, 112568 https://doi.org/10.1016/j.psychres.2019.112568.
Green, R., Koshimori, Y., Turner, G., 2010. Research digest: understanding the organic
basis of persistent complaints in mtbi: ndings from functional and structural
neuroimaging. Neuropsychol. Rehabil. 20, 471478. https://doi.org/10.1080/
09602011003693298.
Greicius, M.D., Flores, B.H., Menon, V., Glover, G.H., Solvason, H.B., Kenna, H., Reiss, A.
L., Schatzberg, A.F., 2007. Resting-state functional connectivity in major depression:
abnormally increased contributions from subgenual cingulate cortex and thalamus.
Biol. Psychiatry 62, 429437. https://doi.org/10.1016/j.biopsych.2006.09.020.
Grieve, S.M., Korgaonkar, M.S., Koslow, S.H., Gordon, E., Williams, L.M., 2013.
Widespread reductions in gray matter volume in depression. NeuroImage: Clin. 3,
332339. https://doi.org/10.1016/j.nicl.2013.08.016.
Hamilton, M., 1967. Development of a rating scale for primary depressive illness. Br. J.
Soc. Clin. Psychol. 6, 278296. https://doi.org/10.1111/j.2044-8260.1967.tb00530.
x.
Helm, K., Viol, K., Weiger, T.M., Tass, P.A., Grefkes, C., del Monte, D., Schiepek, G.,
2018. Neuronal connectivity in major depressive disorder: a systematic review. NDT
14, 27152737. https://doi.org/10.2147/NDT.S170989.
Huang, R., Wang, K., Hu, J., 2016. Effect of probiotics on depression: a systematic review
and meta-analysis of randomized controlled trials. Nutrients 8, 483. https://doi.org/
10.3390/nu8080483.
Hutchinson, E.B., Schwerin, S.C., Avram, A.V., Juliano, S.L., Pierpaoli, C., 2018.
Diffusion MRI and the detection of alterations following traumatic brain injury.
J. Neuro Res. 96, 612625. https://doi.org/10.1002/jnr.24065.
Immonen, R.J., Kharatishvili, I., Niskanen, J.-P., Gr¨
ohn, H., Pitk¨
anen, A., Gr¨
ohn, O.H.J.,
2009. Distinct MRI pattern in lesional and perilesional area after traumatic brain
injury in rat 11 months follow-up. Exp. Neurol. 215, 2940. https://doi.org/
10.1016/j.expneurol.2008.09.009.
Jeurissen, B., Descoteaux, M., Mori, S., Leemans, A., 2019. Diffusion MRI ber
tractography of the brain. NMR Biomed. 32, e3785 https://doi.org/10.1002/
nbm.3785.
Johnson, D., Thurairajasingam, S., Letchumanan, V., Chan, K.-G., Lee, L.-H., 2021.
Exploring the role and potential of probiotics in the eld of mental health: major
depressive disorder. Nutrients 13, 1728. https://doi.org/10.3390/nu13051728.
Kaiser, R.H., Andrews-Hanna, J.R., Wager, T.D., Pizzagalli, D.A., 2015. Large-scale
network dysfunction in major depressive disorder: a meta-analysis of resting-state
functional connectivity. JAMA Psychiatry 72, 603. https://doi.org/10.1001/
jamapsychiatry.2015.0071.
Kelly, J.R., Borre, Y., OBrien, C., Patterson, E., El Aidy, S., Deane, J., Kennedy, P.J.,
Beers, S., Scott, K., Moloney, G., Hoban, A.E., Scott, L., Fitzgerald, P., Ross, P.,
Stanton, C., Clarke, G., Cryan, J.F., Dinan, T.G., 2016. Transferring the blues:
depression-associated gut microbiota induces neurobehavioural changes in the rat.
J. Psychiatr. Res. 82, 109118. https://doi.org/10.1016/j.jpsychires.2016.07.019.
Kim, C.-S., Cha, L., Sim, M., Jung, S., Chun, W.Y., Baik, H.W., Shin, D.-M., 2021.
Probiotic supplementation improves cognitive function and mood with changes in
gut microbiota in community-dwelling older adults: a randomized, double-blind,
placebo-controlled, multicenter trial. J. Gerontol. A 76, 3240. https://doi.org/
10.1093/gerona/glaa090.
Klemm, E., Danos, P., Grünwald, F., Kasper, S., M¨
oller, H.-J., Biersack, H.-J., 1996.
Temporal lobe dysfunction and correlation of regional cerebral blood ow
abnormalities with psychopathology in schizophrenia and major depression a
study with single photon emission computed tomography. Psychiatry Res.
Neuroimaging 68, 110. https://doi.org/10.1016/S0925-4927(96)02837-5.
G. Yamanbaeva et al.
Journal of Aective Disorders 324 (2023) 529–538
538
Koenigs, M., Barbey, A.K., Postle, B.R., Grafman, J., 2009. Superior parietal cortex is
critical for the manipulation of information in working memory. J. Neurosci. 29,
1498014986. https://doi.org/10.1523/JNEUROSCI.3706-09.2009.
Kohn, Y., Freedman, N., Lester, H., Krausz, Y., Chisin, R., Lerer, B., Bonne, O., 2008.
Cerebral perfusion after a 2-year remission in major depression. Int. J.
Neuropsychopharm. 11 https://doi.org/10.1017/S1461145708008936.
Laitinen, T., Sierra, A., Bolkvadze, T., Pitk¨
anen, A., Gr¨
ohn, O., 2015. Diffusion tensor
imaging detects chronic microstructural changes in white and gray matter after
traumatic brain injury in rat. Front. Neurosci. 9 https://doi.org/10.3389/
fnins.2015.00128.
LeWinn, K.Z., Connolly, C.G., Wu, J., Drahos, M., Hoeft, F., Ho, T.C., Simmons, A.N.,
Yang, T.T., 2014. White matter correlates of adolescent depression: structural
evidence for frontolimbic disconnectivity. J. Am. Acad. Child Adolesc. Psychiatry 53
(899909). https://doi.org/10.1016/j.jaac.2014.04.021, 909.e1-7.
Li, G., Rossbach, K., Zhang, A., Liu, P., Zhang, K., 2018. Resting-state functional changes
in the precuneus within rst-episode drug-naive patients with MDD. NDT 14,
19911998. https://doi.org/10.2147/NDT.S168060.
Liao, Y., Huang, X., Wu, Q., Yang, C., Kuang, W., Du, M., Lui, S., Yue, Q., Chan, R.,
Kemp, G., Gong, Q., 2013. Is depression a disconnection syndrome? Meta- analysis of
diffusion tensor imaging studies in patients with MDD. J. Psychiatry Neurosci. 38,
4956. https://doi.org/10.1503/jpn.110180.
Maes, M., Dierckx, R., Meltzer, H.Y., Ingels, M., Schotte, C., Vandewoude, M.,
Calabrese, J., Cosyns, P., 1993. Regional cerebral blood ow in unipolar depression
measured with tc-99m-HMPAO single photon emission computed tomography:
negative ndings. Psychiatry Res. Neuroimaging 50, 7788. https://doi.org/
10.1016/0925-4927(93)90012-7.
Markowiak, P., ´
Sli˙
zewska, K., 2017. Effects of probiotics, prebiotics, and synbiotics on
human health. Nutrients 9, 1021. https://doi.org/10.3390/nu9091021.
Mayberg, H.S., 2003. Modulating dysfunctional limbic-cortical circuits in depression:
towards development of brain-based algorithms for diagnosis and optimised
treatment. Br. Med. Bull. 65, 193207. https://doi.org/10.1093/bmb/65.1.193.
Mayberg, H.S., Lewis, P.J., Regenold, W., Wagner, H.N., 1994. Paralimbic hypoperfusion
in unipolar depression. J. Nucl. Med. 35, 929934.
Mayberg, H.S., Liotti, M., Brannan, S.K., McGinnis, S., Mahurin, R.K., Jerabek, P.A.,
Silva, J.A., Tekell, J.L., Martin, C.C., Lancaster, J.L., Fox, P.T., 1999. Reciprocal
limbic-cortical function and negative mood: converging PET ndings in depression
and normal sadness. Am. J. Psychiatry 156, 675682. https://doi.org/10.1176/
ajp.156.5.675.
Morgane, P., Galler, J., Mokler, D., 2005. A review of systems and networks of the limbic
forebrain/limbic midbrain. Prog. Neurobiol. 75, 143160. https://doi.org/10.1016/
j.pneurobio.2005.01.001.
Ng, Q.X., Peters, C., Ho, C.Y.X., Lim, D.Y., Yeo, W.-S., 2018. A meta-analysis of the use of
probiotics to alleviate depressive symptoms. J. Affect. Disord. 228, 1319. https://
doi.org/10.1016/j.jad.2017.11.063.
Niogi, S.N., Mukherjee, P., 2010. Diffusion tensor imaging of mild traumatic brain injury.
J. Head Trauma Rehabil. 25, 241255. https://doi.org/10.1097/
HTR.0b013e3181e52c2a.
Peng, D., Liddle, E.B., Iwabuchi, S.J., Zhang, C., Wu, Z., Liu, J., Jiang, K., Xu, L.,
Liddle, P.F., Palaniyappan, L., Fang, Y., 2015. Dissociated large-scale functional
connectivity networks of the precuneus in medication-naïve rst-episode depression.
Psychiatry Res. Neuroimaging 232, 250256. https://doi.org/10.1016/j.
pscychresns.2015.03.003.
Pierpaoli, C., Jezzard, P., Basser, P.J., Barnett, A., Di Chiro, G., 1996. Diffusion tensor MR
imaging of the human brain. Radiology 201, 637648. https://doi.org/10.1148/
radiology.201.3.8939209.
Power, J.D., Mitra, A., Laumann, T.O., Snyder, A.Z., Schlaggar, B.L., Petersen, S.E., 2014.
Methods to detect, characterize, and remove motion artifact in resting state fMRI.
NeuroImage 84, 320341. https://doi.org/10.1016/j.neuroimage.2013.08.048.
Putten, H.P.V., Bouwhuis, M.G., Muizelaar, J.P., Lyeth, B.G., Berman, R.F., 2005.
Diffusion-weighted imaging of edema following traumatic brain injury in rats: effects
of secondary hypoxia. J. Neurotrauma 22, 857872. https://doi.org/10.1089/
neu.2005.22.857.
Rolls, E.T., Cheng, W., Feng, J., 2020. The orbitofrontal cortex: reward, emotion and
depression. Brain Commun. 2, fcaa196 https://doi.org/10.1093/braincomms/
fcaa196.
Sack, A.T., 2009. Parietal cortex and spatial cognition. Behav. Brain Res. 202, 153161.
https://doi.org/10.1016/j.bbr.2009.03.012.
Schaub, A.-C., Kirschner, M., Schweinfurth, N., M¨
ahlmann, L., Kettelhack, C., Engeli, E.
E., Doll, J.P.K., Borgwardt, S., Lang, U.E., Kaiser, S., Walter, M., Herdener, M.,
Wrege, J., Schmidt, A., 2021. Neural mapping of anhedonia across psychiatric
diagnoses: a transdiagnostic neuroimaging analysis. NeuroImage: Clin. 32, 102825
https://doi.org/10.1016/j.nicl.2021.102825.
Schaub, A.-C., Schneider, E., Vazquez-Castellanos, J.F., Schweinfurth, N., Kettelhack, C.,
Doll, J.P.K., Yamanbaeva, G., M¨
ahlmann, L., Brand, S., Beglinger, C., Borgwardt, S.,
Raes, J., Schmidt, A., Lang, U.E., 2022. Clinical, gut microbial and neural effects of a
probiotic add-on therapy in depressed patients: a randomized controlled trial.
Transl. Psychiatry 12, 227. https://doi.org/10.1038/s41398-022-01977-z.
Sliz, D., Hayley, S., 2012. Major depressive disorder and alterations in insular cortical
activity: a review of current functional magnetic imaging research. Front. Hum.
Neurosci. 6 https://doi.org/10.3389/fnhum.2012.00323.
Smith, S.M., 2002. Fast robust automated brain extraction. Hum. Brain Mapp. 17,
143155. https://doi.org/10.1002/hbm.10062.
Smith, D.J., Cavanagh, J.T.O., 2005. The use of single photon emission computed
tomography in depressive disorders. Nucl. Med. Commun. 26, 197203. https://doi.
org/10.1097/00006231-200503000-00004.
Smith, S.M., Jenkinson, M., Woolrich, M.W., Beckmann, C.F., Behrens, T.E.J., Johansen-
Berg, H., Bannister, P.R., De Luca, M., Drobnjak, I., Flitney, D.E., Niazy, R.K.,
Saunders, J., Vickers, J., Zhang, Y., De Stefano, N., Brady, J.M., Matthews, P.M.,
2004. Advances in functional and structural MR image analysis and implementation
as FSL. NeuroImage 23, S208S219. https://doi.org/10.1016/j.
neuroimage.2004.07.051.
Solleveld, M.M., Schrantee, A., Homberg, J.R., Lucassen, P.J., Reneman, L., 2020. The
inuence of age-of-onset of antidepressant use on the acute CBF response to a
citalopram challenge; a pharmacological MRI study. Psychiatry Res. Neuroimaging
303, 111126. https://doi.org/10.1016/j.pscychresns.2020.111126.
Thomalla, G., Glauche, V., Koch, M.A., Beaulieu, C., Weiller, C., R¨
other, J., 2004.
Diffusion tensor imaging detects early wallerian degeneration of the pyramidal tract
after ischemic stroke. NeuroImage 22, 17671774. https://doi.org/10.1016/j.
neuroimage.2004.03.041.
Tillisch, K., Labus, J., Kilpatrick, L., Jiang, Z., Stains, J., Ebrat, B., Guyonnet, D., Legrain-
Raspaud, S., Trotin, B., Naliboff, B., Mayer, E.A., 2013. Consumption of fermented
Milk product with probiotic modulates brain activity. Gastroenterology 144. https://
doi.org/10.1053/j.gastro.2013.02.043, 1394-1401.e4.
Tournier, J.-D., Mori, S., Leemans, A., 2011. Diffusion tensor imaging and beyond. Magn.
Reson. Med. 65, 15321556. https://doi.org/10.1002/mrm.22924.
van Velzen, L.S., Kelly, S., Isaev, D., Aleman, A., Aftanas, L.I., Bauer, J., Baune, B.T.,
Brak, I.V., Carballedo, A., Connolly, C.G., Couvy-Duchesne, B., Cullen, K.R.,
Danilenko, K.V., Dannlowski, U., Enneking, V., Filimonova, E., F¨
orster, K., Frodl, T.,
Gotlib, I.H., Groenewold, N.A., Grotegerd, D., Harris, M.A., Hatton, S.N., Hawkins, E.
L., Hickie, I.B., Ho, T.C., Jansen, A., Kircher, T., Klimes-Dougan, B., Kochunov, P.,
Krug, A., Lagopoulos, J., Lee, R., Lett, T.A., Li, M., MacMaster, F.P., Martin, N.G.,
McIntosh, A.M., McLellan, Q., Meinert, S., Nenadi´
c, I., Osipov, E., Penninx, B.W.J.H.,
Portella, M.J., Repple, J., Roos, A., Sacchet, M.D., S¨
amann, P.G., Schnell, K.,
Shen, X., Sim, K., Stein, D.J., van Tol, M.-J., Tomyshev, A.S., Tozzi, L., Veer, I.M.,
Vermeiren, R., Vives-Gilabert, Y., Walter, H., Walter, M., van der Wee, N.J.A., van
der Werff, S.J.A., Schreiner, M.W., Whalley, H.C., Wright, M.J., Yang, T.T., Zhu, A.,
Veltman, D.J., Thompson, P.M., Jahanshad, N., Schmaal, L., 2020. White matter
disturbances in major depressive disorder: a coordinated analysis across 20
international cohorts in the ENIGMA MDD working group. Mol. Psychiatry 25,
15111525. https://doi.org/10.1038/s41380-019-0477-2.
Vasic, N., Wolf, N.D., Gr¨
on, G., Sosic-Vasic, Z., Connemann, B.J., Sambataro, F., von
Strombeck, A., Lang, D., Otte, S., Dudek, M., Wolf, R.C., 2015. Baseline brain
perfusion and brain structure in patients with major depression: a multimodal
magnetic resonance imaging study. J. Psychiatry Neurosci. 40, 412421. https://doi.
org/10.1503/jpn.140246.
Videbech, P., 2000. PET measurements of brain glucose metabolism and blood ow in
major depressive disorder: a critical review: PET measurements in major depressive
disorder. Acta Psychiatr. Scand. 101, 1120. https://doi.org/10.1034/j.1600-
0447.2000.101001011.x.
Vlassenko, A., Sheline, Y.I., Fischer, K., Mintun, M.A., 2004. Cerebral perfusion response
to successful treatment of depression with different serotoninergic agents. JNP 16,
360363. https://doi.org/10.1176/jnp.16.3.360.
Wakana, S., Caprihan, A., Panzenboeck, M.M., Fallon, J.H., Perry, M., Gollub, R.L.,
Hua, K., Zhang, J., Jiang, H., Dubey, P., Blitz, A., van Zijl, P., Mori, S., 2007.
Reproducibility of quantitative tractography methods applied to cerebral white
matter. NeuroImage 36, 630644. https://doi.org/10.1016/j.
neuroimage.2007.02.049.
Wallis, J.D., 2007. Orbitofrontal cortex and its contribution to decision-making. Annu.
Rev. Neurosci. 30, 3156. https://doi.org/10.1146/annurev.
neuro.30.051606.094334.
Whiteld-Gabrieli, S., Nieto-Castanon, A., 2012. Conn : a functional connectivity toolbox
for correlated and anticorrelated brain networks. Brain Connect. 2, 125141.
https://doi.org/10.1089/brain.2012.0073.
Woolrich, M.W., Ripley, B.D., Brady, M., Smith, S.M., 2001. Temporal autocorrelation in
univariate linear modeling of FMRI data. NeuroImage 14, 13701386. https://doi.
org/10.1006/nimg.2001.0931.
Yong, S.J., Tong, T., Chew, J., Lim, W.L., 2020. Antidepressive mechanisms of probiotics
and their therapeutic potential. Front. Neurosci. 13, 1361. https://doi.org/10.3389/
fnins.2019.01361.
Zhang, F.-F., Peng, W., Sweeney, J.A., Jia, Z.-Y., Gong, Q.-Y., 2018. Brain structure
alterations in depression: psychoradiological evidence. CNS Neurosci. Ther. 24,
9941003. https://doi.org/10.1111/cns.12835.
Zheng, P., Zeng, B., Zhou, C., Liu, M., Fang, Z., Xu, X., Zeng, L., Chen, J., Fan, S., Du, X.,
Zhang, X., Yang, D., Yang, Y., Meng, H., Li, W., Melgiri, N.D., Licinio, J., Wei, H.,
Xie, P., 2016. Gut microbiome remodeling induces depressive-like behaviors through
a pathway mediated by the hosts metabolism. Mol. Psychiatry 21, 786796. https://
doi.org/10.1038/mp.2016.44.
G. Yamanbaeva et al.
... These improvements were observed as early as 4 weeks into the treatment and were sustained over time. Furthermore, these probiotic supplements were found to be well-tolerated, with high adherence rates and no serious adverse reactions reported [151][152][153][154][155][157][158][159][160][161][162]. ...
... Recent studies have also started to investigate the neural mechanisms underlying the effects of probiotics on depression. Preliminary neuroimaging findings suggest that probiotic supplementation may prevent neuronal degeneration and alter functional connectivity in fronto-limbic brain structures, which are often implicated in MDD [161]. ...
Article
Full-text available
Major depressive disorder (MDD) is a leading cause of disability worldwide. While traditional pharmacological treatments are effective for many cases, a significant proportion of patients do not achieve full remission or experience side effects. Nutritional interventions hold promise as an alternative or adjunctive approach, especially for treatment-resistant depression. This review examines the potential role of nutrition in managing MDD through addressing biological deficits and modulating pathways relevant to its pathophysiology. Specifically, it explores the ketogenic diet and gut microbiome modulation through various methods, including probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation. Numerous studies link dietary inadequacies to increased MDD risk and deficiencies in nutrients like omega-3 s, vitamins D and B, magnesium, and zinc. These deficiencies impact neurotransmitters, inflammation, and other biological factors in MDD. The gut-brain axis also regulates mood, stress response, and immunity, and disruptions are implicated in MDD. While medications aid acute symptoms, nutritional strategies may improve long-term outcomes by preventing relapse and promoting sustained remission. This comprehensive review aims to provide insights into nutrition’s multifaceted relationship with MDD and its potential for developing more effective integrated treatment approaches.
... These findings indicate that probiotics have the potential to elicit beneficial outcomes on the central nervous system (CNS). It is important to highlight that depression is linked not only to modifications in neurotransmitters but also variations in brain morphology [145]. ...
... Functional connectivity strength increases in the cerebellum and TP were also reported in depressive patients . Treatments of ECT and probiotics would increase the functional connectivity of TP that correlated with the reduction of depressive symptoms (Argyelan et al., 2016;Liu et al., 2015;Yamanbaeva et al., 2023). It was suggested that the antidepressant effect of treatments may be mediated by regulation of functional activity and connectivity (Argyelan et al., 2016). ...
Article
Full-text available
Background The efficacy of bright light therapy (BLT) in ameliorating depression has been validated. The present study is to investigate the changes of depressive symptoms, cognitive function and cerebellar functional connectivity (FC) following BLT in individuals with subthreshold depression (StD). Method Participants were randomly assigned to BLT group (N = 47) or placebo (N = 41) in this randomized controlled trial between March 2020 and June 2022. Depression severity and cognitive function were assessed, as well as resting-state functional MRI scan was conducted before and after 8-weeks treatment. Seed-based whole-brain static FC (sFC) and dynamic FC (dFC) analyses of the bilateral cerebellar subfields were conducted. Besides, a multivariate regression model examined whether baseline brain FC was associated with changes of depression severity and cognitive function during BLT treatment. Results After 8-week BLT treatment, individuals with StD showed improved depressive symptoms and attention/vigilance cognitive function. BLT also increased sFC between the right cerebellar lobule IX and left temporal pole, and decreased sFC within the cerebellum, and dFC between the right cerebellar lobule IX and left medial prefrontal cortex. Moreover, the fusion of sFC and dFC at baseline could predict the improvement of attention/vigilance in response to BLT. Conclusions The current study identified that BLT improved depressive symptoms and attention/vigilance, as well as changed cerebellum-DMN connectivity, especially in the cerebellar-frontotemporal and cerebellar internal FC. In addition, the fusion features of sFC and dFC at pre-treatment could serve as an imaging biomarker for the improvement of attention/vigilance cognitive function after BLT in StD.
... In patients with anxiety and depression, the probiotic combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduced symptoms of the disease, improved problem-solving ability, and reduced urinary-free cortisol levels [23]. In addition to the improvement in cognitive function, imaging tests have shown anatomical and functional changes in different brain areas involved in the pathophysiology of affective disorders following probiotic consumption [24]. However, most of the results are derived from individuals with multiple pathologies so it is crucial to assess whether such findings can be transferred to the healthy population, with a special focus on the elderly [25]. ...
Article
Full-text available
As the population ages, cognitive decline becomes more common. Strategies targeting the gut-brain axis using probiotics are emerging to achieve improvements in neuropsychiatric and neurological disorders. However, the beneficial role of probiotics on brain function in healthy older adults remains unclear. Our aim was to evaluate a multi-species probiotic formulation as a therapeutic approach to reduce emotional and cognitive decline associated with aging in healthy adults. A randomized double-blind placebo-controlled crossover trial was conducted. The study involved a 10-week intervention where participants consumed the assigned probiotic product daily, followed by a 4-week washout period before the second condition started. Cognitive function was assessed using the Mini-Mental State Examination (MMSE) and the Psychological Experiments Construction Language Test Battery. At the emotional level, the Beck Depression Inventory (BDI) and the State-Trait Anxiety Inventory (STAI) were used. Thirty-three participants, recruited between July 2020 and April 2022, ingested a multispecies probiotic (Lactobacillus rhamnosus and Bifidobacterium lactis). After the intervention, noticeable enhancements were observed in cognitive function (mean difference 1.90, 95% CI 1.09 to 2.70, p < 0.005), memory (mean difference 4.60, 95% CI 2.91 to 6.29, p < 0.005) by MMSE and digit task, and depressive symptoms (mean difference 4.09, 95% CI 1.70 to 6.48, p < 0.005) by BDI. Furthermore, there were significant improvements observed in planning and problem-solving skills, selective attention, cognitive flexibility, impulsivity, and inhibitory ability. Probiotics administration improved cognitive and emotional function in older adults. Limited research supports this, requiring more scientific evidence for probiotics as an effective therapy for cognitive decline. This study has been prospectively registered at ClinicalTrials.gov (NCT04828421; 2020/July/17).
Article
The gut microbiota emerged as a potential modulator of brain connectivity in health and disease. This systematic review details current evidence on the gut-brain axis and its influence on brain connectivity. The initial set of studies included 532 papers, updated to January 2024. Studies were selected based on employed techniques. We excluded reviews, studies without connectivity focus, studies on non-human subjects. Forty-nine papers were selected. Employed techniques in healthy subjects included 15 functional magnetic resonance imaging studies (fMRI), 5 diffusion tensor imaging, (DTI) 1 electroencephalography (EEG), 6 structural magnetic resonance imaging, 2 magnetoencephalography, 1 spectroscopy, 2 arterial spin labeling (ASL); in patients 17 fMRI, 6 DTI, 2 EEG, 9 structural MRI, 1 transcranial magnetic stimulation, 1 spectroscopy, 2 R2*MRI. In healthy subjects, the gut microbiota was associated with connectivity of areas implied in cognition, memory, attention and emotions. Among the tested areas, amygdala and temporal cortex showed functional and structural differences based on bacteria abundance, as well as frontal and somatosensory cortices, especially in patients with inflammatory bowel syndrome. Several studies confirmed the connection between microbiota and brain functions in healthy subjects and patients affected by gastrointestinal to renal and psychiatric diseases.
Article
Full-text available
The gut–brain axis, a bidirectional communication network between the gastrointestinal system and the brain, significantly influences mental health and behavior. Probiotics, live microorganisms conferring health benefits, have garnered attention for their potential to modulate this axis. However, their effects on brain function through gut microbiota modulation remain controversial. This systematic review examines the effects of probiotics on brain activity and functioning, focusing on randomized controlled trials using both resting-state and task-based functional magnetic resonance imaging (fMRI) methodologies. Studies investigating probiotic effects on brain activity in healthy individuals and clinical populations (i.e., major depressive disorder and irritable bowel syndrome) were identified. In healthy individuals, task-based fMRI studies indicated that probiotics modulate brain activity related to emotional regulation and cognitive processing, particularly in high-order areas such as the amygdala, precuneus, and orbitofrontal cortex. Resting-state fMRI studies revealed changes in connectivity patterns, such as increased activation in the Salience Network and reduced activity in the Default Mode Network. In clinical populations, task-based fMRI studies showed that probiotics could normalize brain function in patients with major depressive disorder and irritable bowel syndrome. Resting-state fMRI studies further suggested improved connectivity in mood-regulating networks, specifically in the subcallosal cortex, amygdala and hippocampus. Despite promising findings, methodological variability and limited sample sizes emphasize the need for rigorous, longitudinal research to clarify the beneficial effects of probiotics on the gut–brain axis and mental health.
Article
Full-text available
Introduction Depression and anxiety are pervasive mental health disorders with substantial global burdens. Probiotics, live microorganisms known for their health benefits, have emerged as a potential therapeutic intervention for these conditions. This systematic review and meta-analysis aim to evaluate the strain-specific effects of probiotics on relieving depressive and anxiety symptoms while elucidating underlying mechanisms. Methods EMBASE, Cochrane CENTRAL and PubMed/Medline were systematically queried to identify studies released until May 15, 2024. Randomized Controlled Trials (RCTs) that employed standardized assessment tools for depression and anxiety namely Beck Depression Inventory (BDI), Hamilton Depression Rating Scale (HAMD), Depression Anxiety Stress Scales (DASS), or Montgomery-Asberg Depression Rating Scale (MADRS) were included. Results 12 RCTs involving 707 participants were included. Seven RCTs utilizing the BDI questionnaire demonstrated a significant decrease in depressive symptoms favoring probiotics containing strains such as Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus salivarius, Bifidobacterium bifidum, Bifidobacterium lactis, Bifidobacterium breve, and Bifidobacterium longum (MD: -2.69, CI95%: -4.22/-1.16, p value: 0.00). Conversely, RCTs using HAMD showed a non-significant reduction in depressive symptoms (MD: -1.40, CI95%: -3.29/0.48, p value: 0.14). RCTs employing DASS and MADRS scales also showed no significant differences. Conclusion This meta-analysis offers valuable insights into the strain-specific effects of probiotics containing Lactobacillus and Bifidobacterium species on depressive and anxiety symptoms. While our findings suggest a significant reduction in depressive symptoms based on the BDI scale favoring probiotics, the lack of significant effects observed on the HAMD, DASS, and MADRS scales underscores the complexity inherent in these conditions. It is imperative to acknowledge the mixed results across different measurement scales, indicating the need for cautious interpretation. Therefore, we advocate for a nuanced understanding of probiotics’ impacts on various dimensions of mood, emphasizing the necessity for further research.
Article
Full-text available
We recently indicated that four-week probiotic supplementation significantly reduced depression along with microbial and neural changes in people with depression. Here we further elucidated the biological modes of action underlying the beneficial clinical effects of probiotics by focusing on immune-inflammatory processes. The analysis included a total of N = 43 participants with depression, from which N = 19 received the probiotic supplement and N = 24 received a placebo over four weeks, in addition to treatment as usual. Blood and saliva were collected at baseline, at post-intervention (week 4) and follow-up (week 8) to assess immune-inflammatory markers (IL-1β, IL-6, CRP, MIF), gut-related hormones (ghrelin, leptin), and a stress marker (cortisol). Furthermore, transcriptomic analyses were conducted to identify differentially expressed genes. Finally, we analyzed the associations between probiotic-induced clinical and immune-inflammatory changes. We observed a significant group x time interaction for the gut hormone ghrelin, indicative of an increase in the probiotics group. Additionally, the increase in ghrelin was correlated with the decrease in depressive symptoms in the probiotics group. Transcriptomic analyses identified 51 up- and 57 down-regulated genes, which were involved in functional pathways related to enhanced immune activity. We identified a probiotic-dependent upregulation of the genes ELANE, DEFA4 and OLFM4 associated to immune activation and ghrelin concentration. These results underscore the potential of probiotic supplementation to produce biological meaningful changes in immune activation in patients with depression. Further large-scale mechanistic trials are warranted to validate and extend our understanding of immune-inflammatory measures as potential biomarkers for stratification and treatment response in depression. Trial Registration: www.clinicaltrials.gov, identifier: NCT02957591.
Article
Full-text available
Nowadays, depressive disorder is spreading rapidly all over the world. Therefore, attention to the studies of the pathogenesis of the disease in order to find novel ways of early diagnosis and treatment is increasing among the scientific and medical communities. Special attention is drawn to a biomarker and therapeutic strategy through the microbiota–gut–brain axis. It is known that the symbiotic interactions between the gut microbes and the host can affect mental health. The review analyzes the mechanisms and ways of action of the gut microbiota on the pathophysiology of depression. The possibility of using knowledge about the taxonomic composition and metabolic profile of the microbiota of patients with depression to select gene compositions (metagenomic signature) as biomarkers of the disease is evaluated. The use of in silico technologies (machine learning) for the diagnosis of depression based on the biomarkers of the gut microbiota is given. Alternative approaches to the treatment of depression are being considered by balancing the microbial composition through dietary modifications and the use of additives, namely probiotics, postbiotics (including vesicles) and prebiotics as psychobiotics, and fecal transplantation. The bacterium Faecalibacterium prausnitzii is under consideration as a promising new-generation probiotic and auxiliary diagnostic biomarker of depression. The analysis conducted in this review may be useful for clinical practice and pharmacology.
Article
Full-text available
A promising new treatment approach for major depressive disorder (MDD) targets the microbiota-gut-brain (MGB) axis, which is linked to physiological and behavioral functions affected in MDD. This is the first randomized controlled trial to determine whether short-term, high-dose probiotic supplementation reduces depressive symptoms along with gut microbial and neural changes in depressed patients. Patients with current depressive episodes took either a multi-strain probiotic supplement or placebo over 31 days additionally to treatment-as-usual. Assessments took place before, immediately after and again four weeks after the intervention. The Hamilton Depression Rating Sale (HAM-D) was assessed as primary outcome. Quantitative microbiome profiling and neuroimaging was used to detect changes along the MGB axis. In the sample that completed the intervention (probiotics N = 21, placebo N = 26), HAM-D scores decreased over time and interactions between time and group indicated a stronger decrease in the probiotics relative to the placebo group. Probiotics maintained microbial diversity and increased the abundance of the genus Lactobacillus , indicating the effectivity of the probiotics to increase specific taxa. The increase of the Lactobacillus was associated with decreased depressive symptoms in the probiotics group. Finally, putamen activation in response to neutral faces was significantly decreased after the probiotic intervention. Our data imply that an add-on probiotic treatment ameliorates depressive symptoms (HAM-D) along with changes in the gut microbiota and brain, which highlights the role of the MGB axis in MDD and emphasizes the potential of microbiota-related treatment approaches as accessible, pragmatic, and non-stigmatizing therapies in MDD. Trial Registration: www.clinicaltrials.gov , identifier: NCT02957591.
Article
Full-text available
Anhedonia has been associated with abnormal reward-related striatal dopamine functioning in patients with different psychiatric disorders. Here, we tested whether anhedonia expression mapped onto striatal volume across several psychiatric diagnoses. T1-weighted images from 313 participants including 89 healthy controls (HC), 22 patients with opioid use disorder (OUD), 50 patients with major depressive disorder (MDD), 45 patients with borderline personality disorder (BPD), 49 patients with first-episode psychosis (FEP), 43 patients with cocaine use disorder (CUD) and 15 patients with schizophrenia (SZ) were included. Anhedonia was assessed with subscores of the Beck Depression Inventory (BDI) and/or the Scale for the Assessment of Negative Symptoms (SANS). Voxel-based morphometry (VBM) was conducted for identifying dimensional symptom-structure associations using region of interest (ROI, dorsal and ventral striatum) and whole-brain analyses, as well as for group comparisons of striatal volume. ROI analyses revealed significant negative relationships between putamen volume and BDI and SANS anhedonia scores across OUD, MDD, BPD, CUD and SZ patients (n = 175) and MDD, FEP and SZ patients (n = 114), respectively. Whole-brain VBM analyses confirmed these associations and further showed negative relationships between anhedonia severity and volume of the bilateral cerebellum. There were group differences in right accumbens volume, which however were not related to anhedonia expression across the different diagnoses. Our findings indicate volumetric abnormalities in the putamen and cerebellum as a common neural substrate of anhedonia severity that cut across psychiatric entities.
Article
Full-text available
The field of probiotic has been exponentially expanding over the recent decades with a more therapeutic-centered research. Probiotics mediated microbiota modulation within the microbiota–gut–brain axis (MGBA) have been proven to be beneficial in various health domains through pre-clinical and clinical studies. In the context of mental health, although probiotic research is still in its infancy stage, the promising role and potential of probiotics in various mental disorders demonstrated via in-vivo and in-vitro studies have laid a strong foundation for translating preclinical models to humans. The exploration of the therapeutic role and potential of probiotics in major depressive disorder (MDD) is an extremely noteworthy field of research. The possible etio-pathological mechanisms of depression involving inflammation, neurotransmitters, the hypothalamic–pituitary–adrenal (HPA) axis and epigenetic mechanisms potentially benefit from probiotic intervention. Probiotics, both as an adjunct to antidepressants or a stand-alone intervention, have a beneficial role and potential in mitigating anti-depressive effects, and confers some advantages compared to conventional treatments of depression using anti-depressants.
Article
Full-text available
The orbitofrontal cortex in primates including humans is the key brain area in emotion, and in the representation of reward value and in non-reward, that is not obtaining an expected reward. Cortical processing before the orbitofrontal cortex is about the identity of stimuli, i.e. ′what′ is present, and not about reward value. There is evidence that this holds for taste, visual, somatosensory, and olfactory stimuli. The human medial orbitofrontal cortex represents many different types of reward, and the lateral orbitofrontal cortex represents non-reward and punishment. Not obtaining an expected reward can lead to sadness, and feeling depressed. The concept is advanced that an important brain region in depression is the orbitofrontal cortex, with depression related to over-responsiveness and over-connectedness of the non-reward-related lateral orbitofrontal cortex, and to under-responsiveness and under-connectivity of the reward-related medial orbitofrontal cortex. Evidence from large-scale voxel-level studies and supported by an activation study is described that provides support for this hypothesis. Increased functional connectivity of the lateral orbitofrontal cortex with brain areas that include the precuneus, posterior cingulate cortex, and angular gyrus is found in patients with depression, and is reduced towards the levels in controls when treated with medication. Decreased functional connectivity of the medial orbitofrontal cortex with medial temporal lobe areas involved in memory is found in patients with depression. Some treatments for depression may act by reducing activity or connectivity of the lateral orbitofrontal cortex. New treatments that increase activity or connectivity of the medial orbitofrontal cortex may be useful for depression. These concepts, and that of increased activity in non-reward attractor networks, have potential for advancing our understanding and treatment of depression. The focus is on the orbitofrontal cortex in primates including humans, because of differences of operation of the orbitofrontal cortex, and indeed of reward systems, in rodents. Finally, the hypothesis is developed that the orbitofrontal cortex has a special role in emotion and decision-making in part because as a cortical area it can implement attractor networks useful in maintaining reward and emotional states online, and in decision-making.
Article
Full-text available
Preclinical studies have demonstrated that antidepressant treatment in juvenile rodents affect the ontogeny of the serotonin system. However, whether early antidepressant use has similar effects on the development of the serotonin system in humans remains unknown. Therefore, we investigated whether effects of selective serotonin reuptake inhibitor (SSRI) treatment on the serotonin system are modulated by age. With pharmacological Magnetic Resonance Imaging the cerebral blood flow (CBF) response to an acute citalopram challenge was measured, as a proxy for serotonin function. Fifty-one females with major depressive disorder or anxiety disorder were stratified into three groups: 1) those treated with SSRIs <23 years of age, 2) those treated with SSRIs >23 years of age, and 3) those that were never treated with SSRIs. Additionally, a group of 14 healthy controls was included. CBF decreased after a citalopram challenge in the amygdala, hippocampus and orbitofrontal cortex across the whole sample. However, in contrast to preclinical studies, we did not find any age-dependent effect of SSRI exposure on the CBF response. In view of recent concerns on potential adverse effects of SSRIs administered to children, future studies are needed to replicate our negative findings in larger samples sizes and potentially in a prospective design.
Article
Full-text available
Probiotics have been proposed to ameliorate cognitive impairment and depressive disorder via the gut-brain axis in patients and experimental animal models. However, the beneficial role of probiotics in brain functions of healthy elderly remains unclear. Therefore, a randomized, double-blind, and placebo-controlled multicenter trial was conducted to determine the effects of probiotics on cognition and mood in community-dwelling elderly. Sixty-three healthy elders (≥ 65 years) consumed either placebo or probiotics containing Bifidobacteriumbifidum BGN4 and Bifidobacteriumlongum BORI (1 × 109 CFU/d) for 12 weeks. The gut microbiota was analyzed using 16S rRNA sequencing and bioinformatics. Brain functions were measured using the Consortium to Establish a Registry for Alzheimer’s Disease, Satisfaction with life scale, stress questionnaire, Geriatric depression scale, and Positive affect and negative affect schedule. Blood brain-derived neurotrophic factor (BDNF) was determined using ELISA. Relative abundance of gut bacteria was significantly reduced at week 12 in the probiotics group (p <0.05). The probiotics group showed greater improvement in mental flexibility test and stress score than the placebo group (p <0.05). Contrary to placebo, probiotics significantly increased serum BDNF level (p <0.05). Notably, the gut microbes significantly shifted by probiotics (Eubacterium and Clostridiales) showed significant negative correlation with serum BDNF level only in the probiotics group (RS = -0.37, RS = -0.39, p <0.05). In conclusion, probiotics promote mental flexibility and alleviate stress in healthy elderly, along with causing changes in gut microbiota. These results provide evidence supporting health-promoting properties of probiotics as a part of healthy diet in the elderly.
Article
Full-text available
The accumulating knowledge of the host-microbiota interplay gives rise to the microbiota-gut-brain (MGB) axis. The MGB axis depicts the interkingdom communication between the gut microbiota and the brain. This communication process involves the endocrine, immune and neurotransmitters systems. Dysfunction of these systems, along with the presence of gut dysbiosis, have been detected among clinically depressed patients. This implicates the involvement of a maladaptive MGB axis in the pathophysiology of depression. Depression refers to symptoms that characterize major depressive disorder (MDD), a mood disorder with a disease burden that rivals that of heart diseases. The use of probiotics to treat depression has gained attention in recent years, as evidenced by increasing numbers of animal and human studies that have supported the antidepressive efficacy of probiotics. Physiological changes observed in these studies allow for the elucidation of probiotics antidepressive mechanisms, which ultimately aim to restore proper functioning of the MGB axis. However, the understanding of mechanisms does not yet complete the endeavor in applying probiotics to treat MDD. Other challenges remain which include the heterogeneous nature of both the gut microbiota composition and depressive symptoms in the clinical setting. Nevertheless, probiotics offer some advantages over standard pharmaceutical antidepressants, in terms of residual symptoms, side effects and stigma involved. This review outlines antidepressive mechanisms of probiotics based on the currently available literature and discusses therapeutic potentials of probiotics for depression.
Article
The human body can be considered a superorganism in which it's eukaryotic cells and prokaryotic microorganisms coexist. Almost every organ system of the body lives a symbiotic life with these commensal bacteria. Intestinal microbiota has an important role in shaping, organizing and maintaining mental functions from as early as the intrauterine period. Microbiota-based approaches are becoming more prominent in understanding and treating the etiopathogenesis of neuropsychiatric disorders, especially depression. Antidepressant drugs, which are the first-line option in the treatment of depression today, also contain antimicrobial and immunomodulatory mechanisms of action. Treatment options for directly modifying the microbiota composition include prebiotics, probiotics (psychobiotics) and fecal microbiota transplantation. There are few preclinical and clinical studies on the efficacy and reliability of these treatment options in depression. This article will review pertinent studies on the role of intestinal microbiota in depression and discuss the treatment potential of altering ones gut microbiome.
Article
Accumulating data show that probiotics may be beneficial in reducing depressive symptoms. We conducted an updated meta-analysis and evaluated the effects of probiotics on depressive symptoms. A systematic search of six databases was performed, and the results were reported according to Preferred Reporting Items for Systematic Reviews and Meta-analyses, with the priori-defined protocol registered at PROSPERO (CRD42018107426). In total, 19 double-blind, randomized, placebo-controlled trials with a total of 1901 participants were included in the qualitative synthesis. Participants treated with probiotics showed significantly greater improvement in depressive symptoms than those receiving placebo. The clinical population was stratified by clinical diagnosis into those with major depressive disorder (MDD) and those with other clinical conditions. The beneficial effect of probiotics on depressive symptoms was significant in patients with MDD, but not in those with other clinical conditions and in the general population. In addition, multiple strains of probiotics were more effective in reducing depressive symptoms. In conclusion, altering the gut-brain axis with probiotics may be an approach to improve depression severity. It is essential to verify the efficacy of specific combinations or strains of probiotics for depressive symptoms by conducting studies with a larger sample size in the future.
Article
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.