Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.

24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Received: 4 December 2018 | Revised: 26 April 2019 | Accepted: 15 May 2019

DOI: 10.1002/jnr.24476

REVIEW

The role of inflammation and the gut microbiome


in depression and anxiety

Jason M. Peirce1,2 | Karina Alviña1

1
Department of Biological Sciences, Texas
Tech University, Lubbock, Texas Abstract
2
Honors College, Texas Tech University, The study of the gut microbiome has increasingly revealed an important role in mod‐
Lubbock, Texas
ulating brain function and mental health. In this review, we underscore specific path‐
Correspondence ways and mechanisms by which the gut microbiome can promote the development of
Karina Alviña, Department of Biological
mental disorders such as depression and anxiety. First, we review the involvement of
Sciences, Texas Tech University, 2901 Main
St. Room #05, Biology Building, Lubbock, the stress response and immune system activation in the development of depression
TX 79409.
and anxiety. Then, we examine germ‐free murine models used to uncover the role
Email: karina.alvina@ttu.edu
of the gut microbiome in developing and modulating pertinent activity in the brain
and the immune system. We also document multiple pathways by which stress‐in‐
duced inflammation harms brain function and ultimately affects mental health, and
review how probiotic and prebiotic treatments have shown to be beneficial. Lastly,
we provide an overview of gut microbiome‐derived compounds (short‐chain fatty
acids, tryptophan catabolites, microbial pattern recognition) and related mechanisms
(vagal nerve activity and fecal microbiota transplants) involved in mediating the influ‐
ence of the gut microbiome to mental health. Overall, a picture of the gut microbiome
playing a facilitating role between stress response, inflammation, and depression, and
anxiety is emerging. Future research is needed to firmly establish the microbiome's
causal role, to further elucidate the mechanisms by which gut microbes influence
brain function and mental health, and to possibly develop treatments that improve
mental health through microbiotic targets.

KEYWORDS
anxiety, depression, gut microbiome, inflammation, stress

1 | I NTRO D U C TI O N a variety of techniques, such as 16S ribosomal RNA gene sequence


and taxonomic profiling, metagenomic sequencing of whole commu‐
nity DNA, and alignment of the assembled sequences to the refer‐
1.1 | The human gut microbiome
ence microbial genomes (Proctor, 2016). One of the main goals of the
The gut microbiome is the collection of microorganisms, mainly bac‐ HMP is to uncover a human microbiome reference genome that can
teria, that reside within the gastrointestinal (GI) tract in the human be used to characterize dysbiosis and could allow the association of
host (Backhed, Ley, Sonnenburg, Peterson, & Gordon, 2005). These it with a variety of conditions, both normal and pathological (Proctor,
microbes are estimated to outnumber human cells (Sender, Fuchs, 2016). This is no small task as the composition of the microbiome is
& Milo, 2016) and have orders of magnitude more genes than their affected by several factors including birth mode (i.e., natural vs. ce‐
human host (Qin et al., 2010). The implications of the gut micro‐ sarean), infant feeding method, social and outdoor exposure, and an‐
biome for human health are so vast that the Human Microbiome tibiotic use, which may, in turn, affect risk of allergies and infections
Project (HMP) has begun to analyze the human microbiome by using (Bloomfield et al., 2016). The composition of the gut microbiome also

J Neuro Res. 2019;97:1223–1241. wileyonlinelibrary.com/journal/jnr


© 2019 Wiley Periodicals, Inc. | 1223
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1224 | PEIRCE and ALVIÑA

Between 2010 and 2015, college students throughout the United


Significance States have reported increased prevalence of depression, general‐
A myriad of recent studies has revealed that the gut micro‐ ized anxiety, and social anxiety (Xiao et al., 2017). These increased
biome, or the community of microorganisms (mostly bac‐ rates of mental health issues have been accompanied by lackluster
teria) that reside in the human gut, seems to be intimately treatments. Across four developed countries including the United
interwoven with mental health. More specifically, both the States, rates of mood and anxiety disorders and symptoms have in‐
function of the Central Nervous System and its stress‐trig‐ creased between 1990 and 2015 despite increased treatment and
gered dysfunction, have emerged as targets of gut microbi‐ medication (Jorm et al., 2017). Across 21 countries surveyed by the
ome imbalance. Here, we review seminal insights and recent World Health Organization, roughly one tenth of responders had
advances outlining this gut–brain relationship and elucidat‐ an anxiety disorder, yet approximately only one fourth of those af‐
ing mechanisms underlying this link. We also delineate ap‐ flicted received treatment, and most of the treatments were deemed
proaches for improving mental health by treating the gut inadequate (Alonso et al., 2018). The rising trend of mental afflic‐
microbiome. tions across the world indicates a dire need for novel treatment ideas
that may include microbiome‐mediated means.

1.3 | Gut microbiome influence on


correlates with host genetic variation in immunity‐related pathways
depression and anxiety
(Blekhman et al., 2015). For instance, the host cytokine production
is linked to the metabolic activity of gut microbes (Schirmer et al., With the advent of improved molecular and metagenomic tools,
2016), which further suggests an intimate relationship between the human and animal studies have provided increasing evidence sug‐
gut microbiome and the immune system. gesting a strong link between the gut microbiome composition and
The gut microbiome appears to significantly interact with the the development of mental disorders such as depression and anxiety
nervous system through the gut–brain axis, a communication channel (Bastiaanssen, Cowan, Claesson, Dinan, & Cryan, 2019; Jiang et al.,
between the central and enteric nervous system (Carabotti, Scirocco, 2018; Rogers et al., 2016; Tognini, 2017). First, it is known that a variety
Maselli, & Severi, 2015). Gut microbes thereby have the potential to of bacterial phyla cohabitate in the human gut, with over 1,000 distinct
influence brain activity and ultimately, mental health (Rogers et al., bacterial species (Human Microbiome Jumpstart Reference Strains
2016). Since recognizing that gut microbes play an important role Consortium et al., 2010). More than 70% of the microbiome comes
in human health, efforts have been made to improve the microbi‐ from the two most prominent phyla Firmicutes and Bacteroides, while
ome composition using probiotics and prebiotics (Schrezenmeir & de Proteobacteria, Actinobacteria, Fusobacteria, and Verrucomicrobia
Vrese, 2001). Probiotics are consumed microorganisms intended to are present in reduced numbers (Eckburg et al., 2005). However, we
alter the host gut microbiome and provide health benefits, whereas are only starting to fully understand the implications of this microbial
prebiotics are consumed dietary fibers meant to improve health by diversity for human health and disease. Regarding mental health spe‐
feeding and promoting a balanced intestinal microbial ecosystem (de cifically, a few studies are beginning to show interesting trends. For in‐
Vrese & Schrezenmeir, 2008). stance, patients diagnosed with major depression disorder (MDD) have
a different fecal microbiome composition compared to healthy con‐
trols: MDD patients showed increased Bacteroidetes, Protobacteria,
1.2 | Mental health trends: Focus on
and Actinobacteria, and less Firmicutes. Further, among genus these
depression and anxiety
patients showed increased Enterobacteriaceae and Allistipes, and less
Mental health is a growing concern in the United States and world‐ Faecalibacterium, the last of which inversely correlated with the se‐
wide as rates of common mental disorders such as depression and verity of the depression (Jiang et al., 2015). Interestingly, in this study,
anxiety have increased over the past several decades (Friedrich, no differences were observed between the gut microbiome of female
2017; Jorm, Patten, Brugha, & Mojtabai, 2017). The prevalence of or male MDD patients although sex differences have been reported
depression in the United States increased significantly between in healthy individuals (more on sex‐dependent differences will be dis‐
2005 and 2015, most markedly among youth (Carrellas, Biederman, cussed on Section 33.2).
& Uchida, 2017; Weinberger et al., 2018). Moreover, the increase Similarly, fecal microbiota of patients suffering from general‐
in depressive symptoms have contributed to a surge in the number ized anxiety disorder (GAD) showed a much lower prevalence of
of adolescent deaths by suicide between 2010 and 2015 (Twenge, five genera: Faecalibacterium, Eubacterium rectale, Lachnospira,
Cooper, Joiner, Duffy, & Binau, 2019). This increase in suicide rates Butyricicoccus, and Sutterella (Jiang et al., 2018). These genera
is common to adults as well (Curtin, Warner, & Hedegaard, 2016; could be relevant to mental health due to their documented pro‐
Hedegaard, Curtin, & Warner, 2018), particularly those living in rural duction of short‐chain fatty acid (SCFA) compounds (van de Wouw
areas with greater social isolation and limited access to proper men‐ et al., 2018). Reduced SCFA production in GAD patients could re‐
tal health care (Kegler, Stone, & Holland, 2017). Furthermore, the sult in intestinal barrier dysfunction (Morris et al., 2017) that could
projection is similarly morose for young adults with elevated anxiety. compromise proper immune responses and ultimately contribute to
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1225

brain dysfunction. We will review in detail mechanisms that compro‐ responses to infection or stress. For example, GF mice have an under‐
mise immune system in the gut. developed mucosal immune system compared with conventionally
raised mice (Macpherson & Harris, 2004; Mazmanian, Liu, Tzianabos,
& Kasper, 2005). Further, GF murines exhibit a far less effective im‐
1.4 | Resident gut microbes support the brain and
mune response to carefully administered infection (Inagaki, Suzuki,
immune system
Nomoto, & Yoshikai, 1996; Nardi et al., 1991; Strauch et al., 2005)
Experimentally, a common approach for uncovering the role of the gut and endotoxin challenge (Clarke et al., 2013). A significant defect is
microbiome has been to raise murines in a germ‐free (GF) environment, that GF mice have fewer regulatory T cells (Tregs) with reduced anti‐
and then compare their characteristics with conventionally raised inflammatory capacities (Ostman, Rask, Wold, Hultkrantz, & Telemo,
counterparts. The most notable differences are seen in the brain and 2006; Strauch et al., 2005). An especially notable finding related to
the immune system, both particularly relevant in addressing anxiety neuroinflammation is that GF mice display global defects in their
and depression‐like behaviors. Moreover, both depression and anxiety microglia, which exhibit an immature phenotype and an impaired
disorders are heavily influenced by stress pathways such as the hypo‐ innate immune response (Erny et al., 2015). A later study demon‐
thalamus–pituitary–adrenal (HPA) axis, which can be strongly modified strated that atypical gene expression in the microglia of GF mice in‐
by the gut microbiome composition (Luo et al., 2018; Sudo et al., 2004). volving endotoxemia recognition and inflammation can be observed
Studies of GF murines revealed important differences in the de‐ even prior to birth (Thion, Ginhoux, Ginhoux, & Garel, 2018). These
velopment and function of the brain when compared with conven‐ studies demonstrate an often‐overlooked role that resident gut mi‐
tionally raised animals. For instance, a seminal study reported that GF crobes can play in promoting the development of the host immune
male mice have an overreactive HPA axis leading to increased concen‐ system. Taken together, these findings and the behavioral abnor‐
trations of corticosterone and adrenocorticotropic hormone (ACTH) malities reported in GF murines, a clear picture of the impact of the
following a stressful stimulus (Sudo et al., 2004). These findings have gut microbiome in shaping the immune and nervous system starts
been replicated several times (Clarke et al., 2013; Luo et al., 2018; K. to emerge.
A. Neufeld, Kang, Bienenstock, & Foster, 2011; K. M. Neufeld, Kang,
Bienenstock, & Foster, 2011); and have been expanded to GF male
rats (Crumeyrolle‐Arias et al., 2014). Furthermore, an in‐depth epigen‐ 2 | S TR E S S A N D D I S O R D E R E D
etic study reported that gene expression in the brain of GF male mice I N FL A M M ATI O N C A N R E S U LT I N
was significantly different from controls, the most notable differences D E PR E S S I O N A N D A N X I E T Y
were observed in the hippocampus, cortex, striatum, and cerebellum
(Heijtz et al., 2011). These alterations in the hippocampus and cortex Many human and animal studies have shown that physiological path‐
are likely to contribute to differences in HPA axis activity (Bellavance ways involving inflammatory and stress responses likely play an impor‐
& Rivest, 2014). Likewise, different levels of neurotransmitters in the tant role in the etiology of depression and anxiety (Bekhbat & Neigh,
brain of GF male rats have been reported: elevated norepinephrine, 2018; Benatti et al., 2016; Dantzer, O'Connor, Freund, Johnson, &
dopamine, and serotonin turnover in the striatum, whereas decreased Kelley, 2008; Dantzer, O'Connor, Lawson, & Kelley, 2011; Derry, Padin,
dopaminergic turnover was reported in the frontal cortex, hippocam‐ Kuo, Hughes, & Kiecolt‐Glaser, 2015). Here, we discuss key findings.
pus, and striatum of GF rats (Crumeyrolle‐Arias et al., 2014). Other
findings in GF mice include decreased expression of brain‐derived
2.1 | Stress‐induced inflammation is involved
neurotropic factor (Arentsen, Raith, Qian, Forssberg, & Heijtz, 2015;
in depression
Clarke et al., 2013; Heijtz et al., 2011; Sudo et al., 2004) and nerve
growth factor‐inducible protein A in several brain regions along with The role of inflammation in depression has been documented in sev‐
increased expression of the plasticity‐related proteins synaptophysin eral studies (Kim, Na, Myint, & Leonard, 2016; Leonard, 2018; Miller
and PSD‐95 in the striatum (Heijtz et al., 2011). Overall, there are & Raison, 2016). A systematic review of genetic contributions to
significant changes in the brain of murines who develop without a depression revealed several gene variants involved in both immune
normal gut microbiome, particularly in regions involved in the stress activation and depression (Barnes, Mondelli, & Pariante, 2017).
response. Increasing evidence suggests that these changes in brain According to the “pathogen host hypothesis,” these depression
development can be detrimental for mental health. It is important to risk alleles promote pathogen–host defense interactions that were
note that only male rodents were used for these studies. adaptive in an ancestral infection‐ridden environment; public health
measures that increased hygiene and reduced infections, meanwhile,
have rendered this pathogen host defense mainly maladaptive as it
1.5 | Germ‐free murines reveal an important
now contributes to depressive symptoms such as withdrawal behav‐
role for resident microbes in host immune system
ior (Raison & Miller, 2013). Using murine models, abnormal behav‐
development
ior has been associated with pro‐inflammatory cytokines also seen
Experiments on GF murines have also uncovered important roles in depressive patients (Dantzer et al., 2008). Studies have reported
for the gut microbiome in shaping the host immune system and its increased pro‐inflammatory cytokine levels in some depressive
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1226 | PEIRCE and ALVIÑA

patients compared to healthy controls (Duivis, Vogelzangs, Kupper, 3 | S E X D I FFE R E N C E S I N R ATE S O F


Jonge, & Penninx, 2013; Lamers et al., 2013). Longitudinal studies, D E PR E S S I O N A N D A N X I E T Y
meanwhile, reported that high levels of pro‐inflammatory cytokines
predict future risk for depression (Gimeno et al., 2009; van den Females show higher rates of anxiety and depression compared
Biggelaar et al., 2007). Further, experiments that initiate an immune to males (Albert, Pruessner, & Newhouse, 2015; Songtachalert,
response induce feelings of depression (Eisenberger et al., 2010; Roomruangwong, Carvalho, Bourin, & Maes, 2018). This can in part
Harrison et al., 2009; Reichenberg et al., 2001; Strike, Wardle, & be explained by sex hormones and their differential effects on gut
Steptoe, 2004; Wright, Strike, Brydon, & Steptoe, 2005), while anti‐ microbiome as well as differences in immune cells (including micro‐
inflammatory drugs further enhance recovery in patients with de‐ glia), discrepant stress, and immune responses between males and
pression (Mendlewicz et al., 2006; Muller et al., 2006). Thus, the link females, and sociological factors.
between inflammatory state and depressive mood is supported by
several lines of evidence and suggests that influencing inflammation
3.1 | Sex hormones differentially affect
can indirectly affect mental health.
microglia, the gut microbiome, and immune status
The stress response has been shown to be altered in some cases
of depression. In fact, a link between depression and hyperactive The higher rates of depression and anxiety observed in females
HPA axis has been reported in several conditions (Juruena, Cleare, may be in part explained by sex hormones, as androgens seem to
& Pariante, 2004; Pariante, 2003; Varghese & Brown, 2001). For in‐ have anxiolytic properties whereas estrogens have been found to
stance, patients with depression have higher amounts of corticotro‐ enhance HPA activity (Maeng & Milad, 2015; Turner‐Cobb, Osborn,
pin‐releasing hormone (CRH) in their cerebrospinal fluid and release Silva, Keogh, & Jessop, 2010). Furthermore, rates of depression in fe‐
less ACTH in response to a CRH signal (Varghese & Brown, 2001). males correlate with hormonal changes, particularly with low levels
A murine model of depression–anxiety has similar overproduction of of estrogen that occur throughout the life cycle (Albert et al., 2015).
CRH indicating a hyperactive HPA axis (Park et al., 2013). Furthermore, These low estrogen periods may involve heightened neuroinflam‐
several studies have suggested that inflammation mediates stress‐in‐ mation as estrogen seems to have anti‐inflammatory effects on mi‐
duced depression (Aschbacher et al., 2012; Koo, Russo, Ferguson, croglia (Hanamsagar & Bilbo, 2016). The microglial cells themselves
Nestler, & Duman, 2010; Pace et al., 2006; Zhang et al., 2015). display sex‐dependent differences, as they express more inflamma‐
tion‐related genes in adult female mice compared to males (Thion,
Low, et al., 2018). Furthermore, female mice have higher amounts of
2.2 | Stress‐induced inflammation is involved
mature microglia compared to age‐matched males (Hanamsagar et
in anxiety
al., 2017). However, when immuno‐challenged, only microglial cells
In addition to depression, inflammation has been implicated in anxi‐ in males showed accelerated maturation (Hanamsagar et al., 2017).
ety. Studies of clinically diagnosed anxious patients have reported These changes may be related to inflammation‐associated conditions
increased levels of pro‐inflammatory cytokines compared to healthy such as depression and anxiety that tend to emerge early during de‐
individuals (Duivis et al., 2013; O'Donovan et al., 2010; Pitsavos velopment and are more prominent in females compared to males
et al., 2006). Moreover, activating an immune response has been (Hanamsagar & Bilbo, 2016; Hanamsagar et al., 2017). Similarly, in a
shown to increase anxiety behaviors in rats (Sominsky et al., 2013) model of microembolic stroke in rats, microglial activation was ob‐
and human subjects (Reichenberg et al., 2001). At a mechanistic served in both females and males, but only males displayed larger
level, stress‐induced interleukin (IL)‐6 activity has been shown to branches in activated microglial cells concurrent with anxiety‐like
alter gene expression in monocytes and cause anxiety‐like behav‐ behaviors (Nemeth, Reddy, Bekhbat, Bailey, & Neigh, 2014).
ior in mice (Niraula, Witcher, Sheridan, & Godbout, 2019). Although Sex hormones also mediate sex differences in gut microbiome
more research is needed, observational and experimental studies composition which may exert adverse inflammatory and psychologi‐
support the overall notion that inflammation is involved in anxiety. cal effects (Yurkovetskiy et al., 2013). For instance, adult women have
The link between stress and anxiety in humans is perhaps the been shown to have fewer Bacteroidetes in their gut microbiome
least concrete of the psychophysiological connections made so far. compared to age‐matched men, while Firmicutes and Actinobacteria
A comprehensive review of this topic finds that the link between were not statistically different between sexes (Dominianni et al.,
anxiety and a hyperactive HPA axis is inconclusive, in part because 2015). This study also showed a strong effect of body mass index
co‐morbid depression may confound analyses and because HPA axis (BMI) on gut microbiome composition, especially in women. Namely,
alterations may be a transient state (Faravelli et al., 2012). Several mu‐ higher BMI was correlated with even fewer Bacteroidetes com‐
rine models of anxiety display increased HPA axis activity (Landgraf, pared to a normal BMI (Dominianni et al., 2015). Lower levels of
Wigger, Holsboer, & Neumann, 1999; Park et al., 2013), and exper‐ Bacteroidetes have also been observed in fecal microbiota from
iments perturbing the stress response in mice result in anxiety‐like patients diagnosed with clinical depression (study combining age‐
behavior (Barbosa Neto et al., 2012; Flandreau, Ressler, Owens, & matched females and males) (Naseribafrouei et al., 2014), and in
Nemeroff, 2012; Niraula et al., 2019). Stress seems to be relevant in obese humans and murine models (Cani et al., 2009; Graessler et al.,
murine models, but its role in human anxiety is still undefined. 2013). Furthermore, the documented sexual dimorphism observed
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1227

in a mouse model of Type I diabetes was attributed to differences in cortisol compared to age‐matched women (Kudielka, Buske‐
in gut microbiome composition between males and females, dif‐ Kirschbaum, Hellhammer, & Kirschbaum, 2004). Meanwhile, a meta‐
ferences that could be reversed in a GF environment or castrating analysis failed to observe any significant differences between men
males (Yurkovetskiy et al., 2013). and women in how acute psychological stress affects inflammatory
In sum, sex hormones differentially affect males and females by markers (Steptoe, Hamer, & Chida, 2007).
altering both their gut microbiome composition and their immune Women's higher rates of depression and anxiety may in part be
status, particularly microglial cells. explained by situational as well as biological factors: women are
more prone to a variety of inflammation‐inducing factors such as
3.2 | Sex differences in stress and immune prior depression, somatic symptomatology, interpersonal stress‐
responses: More research is needed ors, childhood adversity, obesity, and physical inactivity that then
cause mood and behavioral changes (Derry et al., 2015). From a so‐
The existing evidence concerning sex differences in stress and im‐
ciological point of view, traditional gender roles may play a part as
mune responses is mixed. Some studies indicate a greater sensitivity
caregiving is associated with increased stress in women, and the dif‐
of women to stressful and inflammatory conditions, for instance sys‐
ference in stress levels between men and women has decreased as
temic inflammation impairs mood and affective behavior in women
more women have entered the workforce (Mayor, 2015). However,
more than men (Moieni et al., 2015); also women seem to exhibit
women continue to have a greater risk of depression compared to
more robust stress responses and more potent immune responses
men in more liberal, economically developed countries, meaning tra‐
compared to men (Bekhbat & Neigh, 2018), and women show greater
ditional gender roles may be only a small piece of the puzzle (Alonso
activation of the enzyme indole 2,3‐dioxygenase (IDO), which me‐
et al., 2018; Rai, Zitko, Jones, Lynch, & Araya, 2013).
tabolizes tryptophan (Trp) through the kynurenine (Kyn) pathway,
and increased levels of anxiogenic Trp catabolites (Trycats) follow‐
ing immune activation (Songtachalert et al., 2018). On the contrary, 4 | A PL AU S I B LE S TR E S S–
other studies have shown that men display greater sensitivity to I N FL A M M ATI O N–D E PR E S S I O N/A N X I E T Y
stressful and inflammatory conditions: women resolve local inflam‐ D I S O R D E R PATH WAY
matory responses more rapidly than men (Rathod et al., 2017), social
isolation causes more mortality in men mediated by greater immune The mechanisms that connect psychological stress to systemic and
activation (Moieni & Eisenberger, 2018), and adult men seem to neural inflammation to mood disorders such as depression and anxi‐
respond to typical psychological stressors with a greater increase ety are beginning to be uncovered (Figure 1). Psychological stress can

F I G U R E 1 Bidirectional communication between the gut microbiome and the CNS is affected by stress. Stress (a) activates the HPA axis
and results in increased intestinal permeability (b). Intestinal permeability lets bacterial endotoxins leak out of the gut lumen (c) and enter
blood circulation. (d) Endotoxins initiate peripheral inflammation. The immune response then spreads to the CNS (e) and neuroinflammation
compromises mental health. Stress primes microglial cells (f) as part of the neuroinflammatory response. See main text for details
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1228 | PEIRCE and ALVIÑA

increase intestinal permeability and allow bacterial liposaccharides system in preparation of pathogen defense and wound healing
(known as endotoxins) to enter blood circulation when an immune can apparently misfire by instigating endotoxemia and long‐lasting
response is initiated (de Punder & Pruimboom, 2015). This peripheral neuroinflammation.
inflammation then can spread to the central nervous system (CNS) in
various ways and thus affect mental health by promoting neurotox‐
4.3 | Neuroinflammation affects brain activity and
ins and hindering neurotransmitters (Miller & Raison, 2016).
mental health
Once inflammation has reached the brain, it can have serious delete‐
4.1 | Stress induces endotoxemia by increasing
rious consequences for mental health. First, activated microglia con‐
intestinal permeability
tribute to neural toxicity by releasing reactive nitrogen and oxygen
Psychological stress has been shown to reliably induce pro‐inflam‐ species that damage brain epithelial cells and compromise the BBB
matory cytokine production in murine models (Bailey et al., 2011; (Kacimi, Giffard, & Yenari, 2011). These microglia are the resident
Zhang et al., 2015) and in human subjects (Baumeister, Akhtar, immune cells of the CNS that participate in neuroinflammation and
Ciufolini, Pariante, & Mondelli, 2016; Maes et al., 1998; Wright et al., whose development and activity have repeatably been shown to re‐
2005). These inflammation changes may be explained by stress sig‐ spond to changes in gut microbiome composition (Erny et al., 2015;
nals that modify the immune system and the intestinal barrier. Stress Thion & Garel, 2018). Second, pro‐inflammatory cytokines activate
hormones mobilize immune cells throughout the body in prepara‐ the enzyme IDO increasing the metabolism of Trp through the Kyn
tion for pathogen defense and wound healing (Dhabhar, Malarkey, pathway (Dantzer et al., 2008; Schwarcz, Bruno, Muchowski, & Wu,
Neri, & McEwen, 2012). As the stress response increases intestinal 2012). Several studies exploring endotoxin‐induced depressive‐like
epithelial permeability to increase water and sodium availability, en‐ behavior in murines suggest that IDO activation plays a causal role
dotoxins may seep into circulation and initiate an immune response in neuroinflammatory mood disorders by promoting the release of
termed “endotoxemia” (de Punder & Pruimboom, 2015). Stress has these neurotoxins (Dobos et al., 2012; O'Connor et al., 2009; Parrott
indeed been found to increase intestinal permeability in several ani‐ et al., 2016; Smith et al., 2013; Walker et al., 2013; Wichers et al.,
mal models including rats (Maslanik et al., 2012; Meddings & Swain, 2005). Third, synthesis of the monoamine neurotransmitters sero‐
2000), mice (Yu et al., 2013), and pigs (Overman, Rivier, & Moeser, tonin, dopamine, and norepinephrine is hindered during neuroin‐
2012), and humans (Vanuytsel et al., 2014). flammation since pro‐inflammatory cytokines damage and divert
the activity of tetrahydrobiopterin, an essential enzyme co‐factor of
monoamine synthesis (Miller & Raison, 2016). Overall, neuroinflam‐
4.2 | Peripheral inflammation causes central
mation promotes a toxic environment unsuitable for proper brain
inflammation
activity and likely harmful for mental health.
The long‐standing notion that the brain is “immune privileged” has
been updated by discoveries revealing that the brain's immune
system exists and is heavily regulated (Galea, Bechmann, & Perry, 5 | PRO B I OTI C S A N D PR E B I OTI C S H AV E
2007). Many murine studies show that peripheral administration C A PAC IT Y TO I M PROV E M E NTA L H E A LTH
of endotoxins causes global expression of pro‐inflammatory cy‐
tokines in the brain (Gatti & Bartfai, 1993; Laye, Parnet, Goujon, Probiotics are consumable microbes intended to promote a healthier
& Dantzer, 1994; Quan, Stern, Whiteside, & Herkenham, 1999). microbiome; prebiotics, on the other hand, are dietary fibers that nour‐
Several reviews have outlined the variety of pathways by which ish commensal gut microbes (Schrezenmeir & de Vrese, 2001). A great
peripheral inflammation can spread to the brain and initiate neuro‐ number of studies on both murines and humans have shown promise
inflammation (Baganz & Blakely, 2013; Dantzer et al., 2008; Miller for probiotics and prebiotics to improve the stress response, the in‐
& Raison, 2016), namely: (a) peripheral cytokines and pathogen‐ testinal barrier, the immune response, and ultimately mental health.
associated molecular patterns send inflammatory signals to the
brain via afferent nerves, (b) cytokines pass through permeable
5.1 | Probiotics and prebiotics dampen the
sections of the blood–brain barrier (BBB), and (c) activated im‐
stress response
mune cells migrate into the brain. When it comes to mental health,
this central neuroinflammation may be especially insidious since it The administration of probiotics and prebiotics has shown beneficial
may last up to 40 times longer than the initial peripheral immune effects on the physiological stress response. For instance, colonizing
response (Qin et al., 2007). The timing of stress is of importance GF male mice with the strain Bifidobacterium infantis normalizes their
as well: the stress pathway can be activated to counter an immune previously overreactive HPA axis in response to restraint stress and
response by exerting an anti‐inflammatory effect; however, stress returns their stress hormone levels to levels observed in control mice
prior to an immune response primes microglia reactivity and pro‐ (Sudo et al., 2004). Similarly, Lactobacillus (L) spp. probiotics ameliorate
motes a pro‐inflammatory neuroimmune environment (Bellavance overreactive HPA axis in mice facing maternal separation stress (Gareau,
& Rivest, 2014). The stress response that activates the immune Jury, MacQueen, Sherman, & Perdue, 2007). Further, administering
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1229

Lactobacillus rhamnosus significantly reduces corticosterone levels in evidenced by less zonulin in their feces (Lamprecht et al., 2012).
mice subjected to stress‐induced hypothermia or the elevated plus Therefore, as seen in a plethora of murine and human studies, there
maze test; this attenuated stress response was accompanied by region‐ is a handful of bacterial species along with some useful prebiotics
dependent alterations in ɣ‐aminobutyric acid (GABA) receptor subunit that decrease intestinal epithelial permeability and thereby lower
expression throughout the brain (Bravo et al., 2011). Daily adminis‐ the risk of endotoxemia and uncontrolled inflammation.
tration of Lactobacillus helveticus NS8 to adult specific pathogen‐free
(SPF) rats reduced plasma corticosterone and ACTH levels in response
5.3 | Probiotics and prebiotics decrease
to chronic restraint stress compared to a selective serotonin reuptake
inflammation
inhibitor (SSRI) (Liang et al., 2015). Pretreating mice with the probiotics
L. helveticus and B. infantis dampens the stress response during water Resident gut microbes can have a significant effect on limiting the in‐
avoidance stress as well as maintains hippocampal neurogenesis and flammatory response in both murines and humans. B. infantis adminis‐
hypothalamic plasticity (Ait‐Belgnaoui et al., 2014). Prebiotic treatment tration decreases inflammatory cytokine production in rats following
consisting of fructo‐ (FOS) and galactooligosaccharides (GOS) lowers a forced swim test (Desbonnet, Garrett, Clarke, Bienenstock, & Dinan,
corticosterone release in mice exposed to chronic psychosocial stress 2008). Mice subjected to early life maternal separation (MS) have
in part by modulating hippocampal and hypothalamic gene expres‐ enhanced peripheral IL‐6 production after the forced swim test, but
sion (Burokas et al., 2017). In healthy medical students, L. casei intake those given B. infantis had a normalized immune response comparable
significantly alleviates academic stress‐induced rises in cortisol levels to that of non‐MS mice (Desbonnet et al., 2010). Giving L. farciminis
(Kato‐Kataoka et al., 2016). Taken together, these findings indicate that to mice suppresses stress‐induced neuroinflammation during partial
the intake of probiotic strains or prebiotic mixes has a potentially sig‐ restraint stress (Ait‐Belgnaoui et al., 2014). L. helveticus administration
nificant effect on the regulation of the stress response in both murines prevents the negative mental effects of an inflammatory diet in mice
and humans. However, although this evidence is compelling more re‐ (Ohland et al., 2013). Daily L. helveticus NS8 intake also improves IL‐10
search is needed to validate results obtained in animal models. levels in adult SPF rats facing chronic restraint stress in comparison
to a SSRI (Liang et al., 2015). FOS/GOS prebiotic treatment lowers
pro‐inflammatory cytokine levels in mice exposed to chronic psycho‐
5.2 | Probiotics and prebiotics decrease intestinal
social stress (Burokas et al., 2017). A more in‐depth study discovered
permeability
that providing male young adult and middle‐aged mice with FOS‐inulin
Probiotic and/or prebiotic administration can have important effects for 12 weeks prevented stress‐induced peripheral inflammation and
on the integrity of the intestinal epithelial barrier, including decreas‐ microglial activation concomitant with changes of the gut microbiome
ing its permeability and thus inhibiting endotoxemia. For instance, and SCFA profile (van de Wouw et al., 2018). Furthermore, IBS pa‐
administering Lactobacillus farciminis to mice suppresses partial re‐ tients treated with B. infantis showed alleviation of their symptoms
straint stress‐induced hyperpermeability of the colon and thereby and a balanced ratio of anti‐inflammatory to pro‐inflammatory cy‐
prevents endotoxins from entering circulation (Ait‐Belgnaoui et al., tokines (O'Mahony et al., 2005). L. reuteri has an anti‐inflammatory
2014). Mice undergoing water avoidance stress display increased effect on human epithelial cells in part by upregulating an anti‐inflam‐
intestinal permeability but treating them with L. farciminis can help matory molecule, NGF, and by inhibiting inflammatory NF‐kB activity
maintaining the barrier integrity and confers epithelial and mucosal (Ma, Forsythe, & Bienenstock, 2004). Athletes taking a multispecies
barrier strengthening (Da Silva et al., 2015, 2014). Pretreatment with probiotic have lower pro‐inflammatory TNF‐α levels (Lamprecht et al.,
probiotics L. helveticus and Bifidobacterium longum also restores tight 2012). In sum, probiotics and prebiotics decrease inflammation in both
junction (TJ) barrier integrity in mice facing water avoidance stress murines and humans, likely by boosting intestinal barrier integrity and
(Ait‐Belgnaoui et al., 2014). Prebiotics decrease intestinal permeabil‐ by presenting anti‐inflammatory signals to immune cells.
ity and improve TJ integrity in obese mice, thereby limiting metabolic
endotoxemia (Cani et al., 2009). Administering prebiotics to rats with
5.4 | Probiotics and prebiotics improve
acute pancreatitis significantly improves intestinal barrier function
anxiety and depression
(Zhong et al., 2009). Treatment with Lactobacillus plantarum is shown
to improve paracellular sealing between epithelial cells by increasing Administration of probiotics and prebiotics has been shown to dimin‐
expression of scaffold protein zonula occludens (ZO)‐1 and trans‐ ish the stress response during difficult tasks, improve the integrity
membrane protein occludin near TJ structures (Karczewski et al., of the intestinal barrier, and decrease inflammation. There is consid‐
2010). These beneficial effects have also been observed in humans. erable evidence suggesting that these improvements among other
A mix of L. rhamnosus and Lactobacillus reuteri reduces small intestinal effects may contribute to decreased anxiety‐ and depression‐like
permeability in children with eczema (Rosenfeldt, Benfeldt, Valerius, behaviors in murines. L. rhamnosus administration reduces stress‐in‐
Paerregaard, & Michaelsen, 2004). Providing a probiotic fermented duced anxiety‐ and depressive‐like behaviors in mice (Bravo et al.,
milk drink to patients with irritable bowel syndrome (IBS) decreases 2011). B. longum reduces anxiety‐like behavior during the marble
their small bowel permeability (Zeng et al., 2008). Athletes taking burying test and reduces depressive‐like behavior during the tail sus‐
a multispecies probiotic have decreased intestinal permeability as pension test in a mouse model of heightened anxiety (Savignac, Kiely,
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1230 | PEIRCE and ALVIÑA

Dinan, & Cryan, 2014). B. breve intake reduces anxiety‐like behav‐ both the stress and the immune system, it is useful to illuminate the
ior during the elevated maze test in the same anxious mouse strain mechanisms by which the gut microbiome may exert its antidepres‐
(Savignac, Tramullas, Kiely, Dinan, & Cryan, 2015). Mice fed with a sive and anxiolytic effects (Figure 2). Microbial effectors include
diet composed of one‐half refined carbohydrate and one‐third fat SCFAs and Trycats, as well as microbial‐associated molecular pat‐
(called inflammatory diet) have increased anxiety‐like behavior and terns (MAMPs). These factors among others affect intestinal bar‐
memory deficits but L. helveticus administration relieves these ef‐ rier integrity, immune system activity, and vagal nerve stimulation to
fects (Ohland et al., 2013). Likewise, daily L. helveticus NS8 treatment improve depression and anxiety.
improves anxiety‐like and depressive‐like behavioral dysfunctions in
adult SPF rats facing chronic restraint stress in comparison to a SSRI
6.1 | SCFAs improve immune status and
(Liang et al., 2015). Chronic intestinal inflammation induces anxi‐
gastrointestinal health
ety‐like behavior in mice but B. longum treatment counteracts the
pro‐anxiety effect (Bercik et al., 2010). Administration of prebiotic The gut microbiome can ferment host‐indigestible carbohydrates
GOS and polydextrose to rats prevents anxiety‐like and depressive‐ into SCFAs such as acetate, propionate, and butyrate (Miller &
like behaviors that typically occurs following inescapable tail shock Wolin, 1996). Microbiome‐derived SCFAs modulate gastrointestinal
stress (Mika et al., 2017). FOS and GOS treatments have antidepres‐ cells in different conditions. For instance, SCFAs bind to colonic epi‐
sive and anxiolytic effects on mice exposed to chronic psychosocial thelial cells to promote cytokine and chemokine production during
stress (Burokas et al., 2017). Further, an excellent study showed immune response (Fukuda et al., 2011; Kim, Kang, Park, Yanagisawa,
that administering L. helveticus and B. longum to rats for two weeks & Kim, 2013; Singh et al., 2014; Singh et al., 2010). SCFAs also pro‐
decreases anxious‐like behavior during the conditioned defensive mote gut homeostasis in noninflammatory conditions (Povoleri et al.,
burying test (Messaoudi et al., 2011). In summary, there is substan‐ 2018) and promote serotonin production in enterochromaffin cells
tial evidence supporting the notion that probiotics and prebiotics do (Reigstad et al., 2015). SCFAs bolster barrier integrity in intestinal
indeed exert antidepressive and anxiolytic effects in murines. epithelial colon cells and decrease permeability (Kelly et al., 2015;
Similar benefits of probiotic and prebiotic treatments can be Mariadason, Barkla, & Gibson, 1997; Peng, Li, Green, Holzman, &
observed in human studies. For instance, the L. helveticus and B. Lin, 2009; Suzuki, Yoshida, & Hara, 2008) thereby preventing micro‐
longum probiotic mix given to healthy human volunteers for one bial endotoxins from entering circulation and initiating an immune
month alleviated psychological distress in comparison to a control response. In addition, SCFAs affect the development of a variety of
group, matching the positive results seen in rats (Messaoudi et al., immune cells: they inhibit dendritic cell development from bone mar‐
2011). High intake of prebiotic trans‐GOS given to patients with row stem cells (Singh et al., 2010), enhance T cell differentiation into
IBS for 12 weeks significantly improved subjective global assess‐ anti‐inflammatory Tregs (Singh et al., 2014; Smith et al., 2013), and
ment scores and anxiety scores compared to placebo treatment regulate microglia maturity and function (Erny et al., 2015). SCFAs
(Silk, Davis, Vulevic, Tzortzis, & Gibson, 2009). In a study of healthy also affect immune cell activity: butyrate downregulates expression
adult volunteers consuming a probiotic‐containing milk drink daily of pro‐inflammatory mediators in macrophages during an immune
for 3 weeks, those initially in the bottom third on the depressed/ response (Chang, Hao, Offermanns, & Medzhitov, 2014), promotes
elated dimension reported significantly improved mood compared the expression of anti‐inflammatory IL‐10 in macrophages and intes‐
to placebo (Benton, Williams, & Brown, 2007). Healthy human sub‐ tinal dendritic cells, and enhances the anti‐inflammatory activity of
jects receiving a multispecies probiotic show a significantly reduced Tregs (Singh et al., 2014). Hence, by bolstering the intestinal epithe‐
overall cognitive reactivity to sad mood compared to those receiving lial barrier and then promoting an anti‐inflammatory phenotype in
a placebo (Steenbergen, Sellaro, Hemert, Bosch, & Colzato, 2015). immune cells, the gut microbiome defends against stress‐induced
Furthermore, two meta‐studies investigating the efficacy of probi‐ endotoxemia and limits mood‐altering neuroinflammation.
otics on counteracting depression reported an overall improvement
of depressive symptoms following probiotic administration with low
6.2 | Trycats lower inflammation and bolster the
risk of side effects (Huang, Wang, & Hu, 2016; Wallace & Milev,
intestinal barrier
2017). Overall these studies indicate that there is a significant po‐
tential for probiotic and prebiotic treatments to improve anxiety and The gut microbiome metabolizes dietary Trp into Trycats (Gao et al.,
depression in human subjects. 2018). These microbiome‐derived Trycats are agonists of aryl hy‐
drocarbon receptors (AHRs), ligand‐dependent transcription factors
expressed on immune cells and intestinal epithelial cells (Stockinger
6 | G U T M I C RO B I O M E /I N FL A M M ATI O N ‐ et al., 2014). There are several ways in which Trycats affect intesti‐
D E R I V E D M EC H A N I S M S TH AT C A N nal integrity and immune activity through AHR binding. Microbe‐
I N FLU E N C E D E PR E S S I O N A N D A N X I E T Y derived Trycats bind to AHR on innate lymphoid cells and induce
the release of IL‐22 to promote antimicrobial peptide production
Given that stress and inflammation play direct and indirect roles and pathogen defense (Qiu et al., 2012; Romani et al., 2014; Zelante
in modulating brain function and that the gut microbiome affects et al., 2014). Trycats activate AHRs on T cells and dendritic cells to
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1231

F I G U R E 2 Gut microbiome–brain axis mechanisms that can influence CNS function. Short‐chain fatty acids (SCFAs), tryptophan
catabolites (Trycats) catabolites, and microbial‐associated molecular patterns (MAMPs) bolster the intestinal barrier (a) preventing
endotoxins from leaking through the intestinal epithelium. This results in reduced inflammation (b). The vagus nerve (c) communicates with
the gut epithelium, including enteroendocrine cells (gray), to exert antidepressive and anxiolytic effects. Probiotics and prebiotics (Pro/Pre),
and fecal microbiota transplants (FMT) have been shown to have beneficial effects on the gut microbiome (d) and mental health. See main
text for details

favor the differentiation of IL‐10‐producing Tregs cells in place of an evolutionary adaptation to maintain immune homeostasis along‐
pro‐inflammatory Th17 cells (Montagnoli et al., 2006; Quintana, side resident microbes; namely, inflammation‐activated IDO makes
2013). At the neural level, these microbial‐produced Trycats bind Trycats lower inflammation by binding to AHRs and promotion of
to AHRs on astrocytes to limit neuroinflammation (Rothhammer et Tregs activity (Zelante et al., 2014). Similar to SCFAs, Trycats pro‐
al., 2016). However, more research is needed to uncover the roles mote immune homeostasis by boosting the intestinal mucosal layer,
of individual Trycats. Lactobacillus spp., especially L. reuteris, can inducing Tregs differentiation, and tilting the balance of cytokines
catabolize tryptophan into indole‐3‐aldehyde (Zelante et al., 2014), toward an anti‐inflammatory state (Martin‐Subero, Anderson,
which exerts anti‐inflammatory effects by increasing expression of Kanchanatawan, Berk, & Maes, 2016).
the IL‐10 receptor in intestinal epithelial cells (Alexeev et al., 2018).
Indole increases expression of genes involved in strengthening the
6.3 | Microbial pattern recognition contributes to
mucosal barrier, increasing mucin production, and decreasing intes‐
immune and intestinal homeostasis
tinal permeability (Bansal, Alaniz, Wood, & Jayaraman, 2010). Indole
is also anti‐inflammatory by decreasing TNF‐α‐mediated activa‐ Hosts use pattern recognition receptors (PRRs) to keep track of resi‐
tion of NF‐kB, lowering expression of pro‐inflammatory chemokine dent microbes by sensing their MAMPs (Chu & Mazmanian, 2013).
IL‐8, and increasing expression of anti‐inflammatory cytokine IL‐10 Toll‐like receptors (TLRs) are a version of PRRs present on immune
(Bansal et al., 2010). Indole‐3‐propionic acid improves intestinal bar‐ and epithelial cells that are used to maintain immune and intestinal
rier function and decreases TNF‐α expression by binding to the preg‐ homeostasis (Kawai & Akira, 2006). TLR2 stimulation on intestinal
nane X receptor on intestinal epithelial cells (Venkatesh et al., 2014). epithelial cells by gastrointestinal microbes enhances transepithe‐
Kynurenic acid has been reported to inhibit LPS‐induced release of lial resistance and preserves ZO‐1‐associated barrier integrity when
TNF‐α in peripheral immune cells (Bansal et al., 2010). Kynurenine facing stress‐induced damage (Cario, 2008). This effect of the mi‐
leads to AHR‐dependent Tregs differentiation from T cells (Mezrich crobiome is especially importance since stress‐induced intestinal
et al., 2010). Some resident microbes harbor the enzyme IDO, which permeability is the first step toward brain‐harming neuroinflamma‐
catabolizes Trp and is activated by pro‐inflammatory cytokines tion. These intestinal epithelial cells also initiate a defense and re‐
(Morris et al., 2017). The IDO–AHR‐Treg axis has been proposed as pair program in response to microbial TLR2 recognition consisting
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1232 | PEIRCE and ALVIÑA

of increased epithelial cell proliferation, secretion of IgA into the species, B. longum and L. rhamnosus, are known to require an intact
gut lumen, and greater expression of antimicrobial peptides (Abreu, vagus nerve for their beneficial effects on depression and anxiety
2010). Intestinal Paneth cells run an antimicrobial program in re‐ (Bercik et al., 2011; Bravo et al., 2011); the latter was later shown
sponse to enteric bacterial TLR sensing which decreases bacterial to operate by increasing the firing rates of vagal afferents (Perez‐
penetration through the intestinal barrier and maintains gut ho‐ Burgos et al., 2013). Recent studies have sought to characterize the
meostasis (Vaishnava, Behrendt, Ismail, Eckmann, & Hooper, 2008). biochemical flow of information through the vagus nerve in other
Bacteroides fragilis likely exerts many of its beneficial effects by ac‐ contexts (Han et al., 2018; Kaelberer et al., 2018). Previously termed
tivating immunomodulatory TLR2 pathways (Troy & Kasper, 2010). enteroendocrine cells have been discovered to synapse onto vagal
It displays polysaccharide A (PSA) on its outer membrane, which nodose neurons and have been renamed neuropod cells (Kaelberer
promotes a Th1/Th2 balance and induces Tregs cells to limit pro‐in‐ et al., 2018). These neuropod cells detect and relay nutrient con‐
flammatory cytokine expression by Th17 cells and increase release ditions in the gut, inviting the possibility that GI cells serve as an
of anti‐inflammatory IL‐10 (Round et al., 2011; Shen et al., 2012; Troy intermediary between microbial signals and vagus nerve activity.
et al., 2010). In sum, commensal gut microbes activate TLR‐sensing Vagal stimulation has also been shown to induce dopamine release
pathways using their MAMPs to improve intestinal epithelial integ‐ in the substantia nigra (Han et al., 2018). This may have implications
rity and to establish immune homeostasis. for mood disorders, especially since rats with lesions in the dopa‐
mine neurons of the substantia nigra exhibit depressive‐like behav‐
ior (Winter et al., 2007). Given that vagus nerve activity increases
6.4 | The vagus nerve mediates some
GABA transmission in the brain when stimulated by L. rhamnosus
antidepressive and anxiolytic effects of probiotics
(Bravo et al., 2011) but can also induce neural dopamine release in
The vagus nerve is the principal component of the parasympathetic other circumstances (Han et al., 2018), future studies should explore
nervous system and links the internal viscera to the CNS (Bonaz, how different aspects of the vagus nerve mediate this relationship
Sinniger, & Pellissier, 2016). Its afferent fibers can also sense periph‐ between microbial signals in the gut and neurotransmitter activity
eral pro‐inflammatory cytokines and initiate the “cholinergic anti‐in‐ in the brain.
flammatory pathway” which activates the HPA axis to dampen down
the immune response (Borovikova, Ivanova, Nardi, et al., 2000;
6.5 | Fecal microbiota transplants show promise
Borovikova, Ivanova, Zhang, et al., 2000; Pavlov et al., 2009). Clearly,
in the treatment of depression and anxiety
the vagus nerve plays a versatile role in sensing enteric conditions
and, in turn, modulating neural transmission and hormone release. A novel biological treatment for disorders relating to unhealthy gut
Two probiotics have been shown to require an intact vagus nerve microbiome are the fecal microbiota transplants (FMT) (Cohen &
to mediate their beneficial effects. First, mice with intestinal inflam‐ Maharshak, 2017; Gupta, Allen‐Vercoe, & Petrof, 2016). FMT is a
mation that normally exhibit anxiety‐like behavior show less anxious procedure that transfers gut microbes from a healthy donor's fecal
displays when treated with B. longum for 1 week; this anxiolytic ef‐ matter into the recipient's intestinal tract to promote a healthier
fect was not observed in mice with a severed vagus nerve which microbiome. FMTs have been successful in treating Clostridium dif‐
suggests that the parasympathetic vagal tone likely plays a causal ficile antibiotic‐resistant infections (Cohen et al., 2015), and have
role in mediating central modulation of behavior (Bercik et al., 2011). shown promise for improving neurological and GI tract‐related dis‐
This probiotic effect has been confirmed in healthy human volun‐ eases (Cohen et al., 2016). As we have reviewed here, several lines
teers where B. longum treatment reduced stress and anxiety while of evidence support the notion that some neurological disorders and
enhancing memory performance (Allen et al., 2016). Second, chronic GI conditions share a common etiology involving gut dysbiosis (i.e.,
treatment of L. rhamnosus in mice significantly reduces anxiety‐like unhealthy gut microbiome). First, patients with inflammatory bowel
behavior in the elevated plus maze, decreases depressive‐like be‐ disease (IBD) have higher rates of depression and anxiety compared
havior in the forced swim test, lowers stress‐induced corticosteroid to healthy controls (Fond et al., 2014; Graff, Walker, & Bernstein,
response, and induces region‐dependent alterations in GABA recep‐ 2009; Mikocka‐Walus et al., 2007). Second, both patients with de‐
tor subunit expression throughout the brain; however, mice with a pression and IBD have significantly different gut microbiomes com‐
severed vagus nerve did not show these effects (Bravo et al., 2011). pared to healthy controls (Morgan et al., 2012; Zheng et al., 2016).
Moreover, L. rhamnosus was found to increase the firing rate of vagal Third, use of antibiotics correlates with increased risk of subse‐
afferents in the mesenteric nerve bundle, and this stimulation is re‐ quent depression, anxiety, and IBD (Lurie, Yang, Haynes, Mamtani,
quired for GABA modulation in the brain (Perez‐Burgos et al., 2013). & Boursi, 2015; Mendall & Kumar, 1998). Lastly, a common mecha‐
Taken together, these studies indicate that vagal afferents mediate nism involving activated immune‐inflammatory, oxidative, and ni‐
at least partially the antidepressive and anxiolytic effects of probi‐ trosative stress pathways (including Trycats and gut‐brain axis) has
otic strains by modulating neural transmission and perhaps influenc‐ been proposed to explain the co‐morbidity of depression and IBD
ing neuroendocrine activity. (Martin‐Subero et al., 2016). Therefore, since the gut microbiome
The vagus nerve's primary role in the gut–brain axis invites fur‐ appears to regulate immune activity through several mechanisms, it
ther study into its mechanisms of action. As of now, two probiotic is natural to question whether FMTs might be an effective method
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1233

to promote mental health by improving immune status via the gut 2017). Future studies could explore whether probiotics could stim‐
microbiome. This viable biological treatment is especially important ulate vagal afferents and doing so they might parallel or reinforce
since psychological therapy has brought few long‐term benefits to these antidepressive effects. Overall, as the primary link between GI
people with co‐morbid depression and IBD (Gracie et al., 2017). In conditions and the brain, the vagus nerve merits further study into
addition, FMTs have shown to be much more effective than a related its mechanisms and roles in mental health treatment.
biological treatment, probiotics, in restoring a healthy gut microbi‐ Other areas to continue developing are the use of pre/probiotics
ome after antibiotic use (Suez et al., 2018). In support of the idea and FMTs. For example, antibiotic‐resistant C. difficile infection has
that changing gut microbiome composition affects mental health, GF been shown to be effectively treated with FMTs (Cohen et al., 2015).
mice receiving FMT from patients with depression exhibited more Recent findings also show promise for the treatment of other con‐
depressive‐like behaviors compared to GF mice receiving FMT from ditions such as diabetes, obesity, metabolic syndrome, and GI tract‐
healthy control individuals (Zheng et al., 2016). related pathologies (Gupta et al., 2016). Therefore, considering that
the gut microbiome seems to have a strong influence over the GI
physiology it is logic to think that FMTs and pre/probiotic treatments
7 | CO N C LU S I O N S A N D FU T U R E can be beneficial. As discussed here, several reports already have
D I R EC TI O N S shown promise, but more research is needed.
Lastly, several important questions remain unanswered (Foster
The study of the gut microbiome–brain axis is revolutionizing our & McVey Neufeld, 2013). One of the most significant open ques‐
understanding of the mechanisms underlying nervous system dis‐ tions is related to sex‐dependent differences in gut microbiome
orders such as depression and anxiety. However, there are cur‐ composition. Even though progress is being made in this regard,
rently more open questions than answers. What is clear is that this is imperative considering that females are most likely to suffer
the gut microbiome's role in modulating mental health deserves from depression and anxiety than males. Likewise, we are just be‐
increased attention from researchers and clinicians. Across the ginning to understand what role the gut microbiome plays during
developed world, rates of depression and anxiety are increasing brain development. Early life is a period of rapid change when the
(Jorm et al., 2017). At the same time, increased rates of urbaniza‐ developing brain is vulnerable to most internal and external influ‐
tion are bringing about concerns over public health infrastructure ences. Furthermore, many mental disorders can originate from early
and policies (Bloomfield et al., 2016). These two trends make more exposure to stress and inflammation, therefore improving our un‐
sense when seen through the lens of the gut microbiome. Modern derstanding of the gut microbiome influence at this stage will also
urban lifestyles often decrease exposure to microbes early in life have a significant impact on diagnosis and possible treatments.
and lead to an underdeveloped immune system prone to chronic
inflammation later in life, possibly serving as a risk factor for men‐
C O N FL I C T O F I N T E R E S T
tal disorders such as depression and anxiety (Rook, Raison, &
Lowry, 2013). Similarly, urban environments with decreased green The authors declare no conflict of interest.
space combined with heavily marketed fast foods can contribute
to gut dysbiosis, particularly among lower socioeconomic classes
AU T H O R C O N T R I B U T I O N S
(Logan, 2015). Indeed, urbanization is associated with decreased
gut microbiome complexity (Segata, 2015), an indicator of poor Conceptualization, J.M.P. and K.A.; Writing—Original Draft, J.M.P.;
microbiome health (Lloyd‐Price, Abu‐Ali, & Huttenhower, 2016). Writing—Review & Editing, J.M.P. and K.A.; Supervision, K.A.; Funding
Increased urbanization worldwide certainly demands increased Acquisition, K.A.
interest in the ways the gut microbiome can contribute to mental
health. Finally, the economic case for further study is incredibly
ORCID
viable: investments in treatments for mental conditions such as
depression and anxiety are likely to bring very high rates of return Karina Alviña https://orcid.org/0000-0002-8369-0953
through increases in productivity and quality of life (Chisholm et
al., 2016).
In future studies, it is necessary to fully characterize the com‐ REFERENCES
ponents of the gut microbiome–brain axis. In fact, one of the areas
Aaronson, S. T., Sears, P., Ruvuna, F., Bunker, M., Conway, C. R.,
that remains poorly explored is the enteroendocrine cells that com‐
Dougherty, D. D., … Zajecka, J. M. (2017). A 5‐year observational
municate with and activate the vagus nerve. Knowing how these study of patients with treatment‐resistant depression treated
cells operate will allow us to then use that criteria to develop more with vagus nerve stimulation or treatment as usual: Comparison
effective probiotic/prebiotic formulations, for example. On a related of response, remission, and suicidality. American Journal of
Psychiatry, 174(7), 640–648. https​ ://doi.org/10.1176/appi.
note, electric vagus nerve stimulation has been shown to signifi‐
ajp.2017.16010034
cantly improve mental health outcomes in patients with treatment‐ Abreu, M. T. (2010). Toll‐like receptor signalling in the intestinal epithe‐
resistant depression compared to a control group (Aaronson et al., lium: How bacterial recognition shapes intestinal function. Nature
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1234 | PEIRCE and ALVIÑA

Reviews Immunology, 10(2), 131–144. https​ ://doi.org/10.1038/ Baumeister, D., Akhtar, R., Ciufolini, S., Pariante, C. M., & Mondelli, V.
nri2707 (2016). Childhood trauma and adulthood inflammation: A meta‐anal‐
Ait‐Belgnaoui, A., Colom, A., Braniste, V., Ramalho, L., Marrot, A., Cartier, ysis of peripheral C‐reactive protein, interleukin‐6 and tumour ne‐
C., … Tompkins, T. (2014). Probiotic gut effect prevents the chronic crosis factor‐alpha. Molecular Psychiatry, 21(5), 642–649. https​://doi.
psychological stress‐induced brain activity abnormality in mice. org/10.1038/mp.2015.67
Neurogastroenterology and Motility, 26(4), 510–520. https​ ://doi. Bekhbat, M., & Neigh, G. N. (2018). Sex differences in the neuro‐im‐
org/10.1111/nmo.12295​ mune consequences of stress: Focus on depression and anxiety.
Albert, K., Pruessner, J., & Newhouse, P. (2015). Estradiol levels mod‐ Brain, Behavior, and Immunity, 67, 1–12. https​ ://doi.org/10.1016/j.
ulate brain activity and negative responses to psychosocial stress bbi.2017.02.006
across the menstrual cycle. Psychoneuroendocrinology, 59, 14–24. Bellavance, M. A., & Rivest, S. (2014). The HPA ‐ Immune axis and the
https​://doi.org/10.1016/j.psyne​uen.2015.04.022 immunomodulatory actions of glucocorticoids in the brain. Frontiers
Alexeev, E. E., Lanis, J. M., Kao, D. J., Campbell, E. L., Kelly, C. J., Battista, in Immunology, 5, 136. https​ ://doi.org/10.3389/fimmu.2014.
K. D., … Colgan, S. P. (2018). Microbiota‐derived indole metabolites 00136​
promote human and murine intestinal homeostasis through regula‐ Benatti, C., M.C. Blom, J., Rigillo, G., Alboni, S., Zizzi, F., Torta, R., …
tion of interleukin‐10 receptor. American Journal of Pathology, 188(5), Tascedda, F. (2016). Disease‐induced neuroinflammation and de‐
1183–1194. https​://doi.org/10.1016/j.ajpath.2018.01.011 pression. CNS & Neurological Disorders: Drug Targets, 15(4), 414–433.
Allen, A. P., Hutch, W., Borre, Y. E., Kennedy, P. J., Temko, A., Boylan, G., https​://doi.org/10.2174/18715​27315​66616​03211​0 4749​
… Clarke, G. (2016). Bifidobacterium longum 1714 as a translational Benton, D., Williams, C., & Brown, A. (2007). Impact of consuming a
psychobiotic: Modulation of stress, electrophysiology and neuro‐ milk drink containing a probiotic on mood and cognition. European
cognition in healthy volunteers. Translational Psychiatry, 6(11), e939. Journal of Clinical Nutrition, 61(3), 355–361. https​://doi.org/10.1038/
https​://doi.org/10.1038/tp.2016.191 sj.ejcn.1602546
Alonso, J., Liu, Z., Evans‐Lacko, S., Sadikova, E., Sampson, N., Chatterji, Bercik, P., Park, A. J., Sinclair, D., Khoshdel, A., Lu, J., Huang, X., …
S., … the WHO World Mental Health Survey Collaborators. (2018). Verdu, E. F. (2011). The anxiolytic effect of Bifidobacterium longum
Treatment gap for anxiety disorders is global: Results of the World NCC3001 involves vagal pathways for gut‐brain communication.
Mental Health Surveys in 21 countries. Depress Anxiety, 35(3), 195– Neurogastroenterology and Motility, 23(12), 1132–1139. https​ ://doi.
208. https​://doi.org/10.1002/da.22711​ org/10.1111/j.1365-2982.2011.01796.x
Arentsen, T., Raith, H., Qian, Y., Forssberg, H., & Heijtz, R. D. (2015). Bercik, P., Verdu, E. F., Foster, J. A., Macri, J., Potter, M., Huang, X., …
Host microbiota modulates development of social preference in Collins, S. M. (2010). Chronic gastrointestinal inflammation induces
mice. Microbial Ecology in Health & Disease, 26, 29719. https​://doi. anxiety‐like behavior and alters central nervous system biochemis‐
org/10.3402/mehd.v26.29719​ try in mice. Gastroenterology, 139(6), 2102–2112.e2101. https​://doi.
Aschbacher, K., Epel, E., Wolkowitz, O. M., Prather, A. A., Puterman, E., org/10.1053/j.gastro.2010.06.063
& Dhabhar, F. S. (2012). Maintenance of a positive outlook during Blekhman, R., Goodrich, J. K., Huang, K., Sun, Q. I., Bukowski, R., Bell, J.
acute stress protects against pro‐inflammatory reactivity and future T., … Clark, A. G. (2015). Host genetic variation impacts microbiome
depressive symptoms. Brain, Behavior, and Immunity, 26(2), 346–352. composition across human body sites. Genome Biology, 16, 191. https​
https​://doi.org/10.1016/j.bbi.2011.10.010 ://doi.org/10.1186/s13059-015-0759-1
Backhed, F., Ley, R. E., Sonnenburg, J. L., Peterson, D. A., & Gordon, Bloomfield, S. F., Rook, G. A., Scott, E. A., Shanahan, F., Stanwell‐Smith,
J. I. (2005). Host‐bacterial mutualism in the human intestine. R., & Turner, P. (2016). Time to abandon the hygiene hypothesis: New
Science, 307(5717), 1915–1920. https​ ://doi.org/10.1126/scien​ perspectives on allergic disease, the human microbiome, infectious
ce.1104816 disease prevention and the role of targeted hygiene. Perspectives in
Baganz, N. L., & Blakely, R. D. (2013). A dialogue between the immune Public Health, 136(4), 213–224. https​://doi.org/10.1177/17579​13916​
system and brain, spoken in the language of serotonin. ACS Chemical 650225
Neuroscience, 4(1), 48–63. https​://doi.org/10.1021/cn300​186b Bonaz, B., Sinniger, V., & Pellissier, S. (2016). Anti‐inflammatory proper‐
Bailey, M. T., Dowd, S. E., Galley, J. D., Hufnagle, A. R., Allen, R. G., & ties of the vagus nerve: Potential therapeutic implications of vagus
Lyte, M. (2011). Exposure to a social stressor alters the structure of nerve stimulation. Journal of Physiology, 594(20), 5781–5790. https​://
the intestinal microbiota: Implications for stressor‐induced immuno‐ doi.org/10.1113/JP271539
modulation. Brain, Behavior, and Immunity, 25(3), 397–407. https​:// Borovikova, L. V., Ivanova, S., Nardi, D., Zhang, M., Yang, H.,
doi.org/10.1016/j.bbi.2010.10.023 Ombrellino, M., & Tracey, K. J. (2000). Role of vagus nerve sig‐
Bansal, T., Alaniz, R. C., Wood, T. K., & Jayaraman, A. (2010). The bac‐ naling in CNI‐1493‐mediated suppression of acute inflamma‐
terial signal indole increases epithelial‐cell tight‐junction resistance tion. Autonomic Neuroscience, 85(1–3), 141–147. https​ ://doi.
and attenuates indicators of inflammation. Proceedings of the National org/10.1016/S1566-0702(00)00233-2
Academy of Sciences, 107(1), 228–233. https​ ://doi.org/10.1073/ Borovikova, L. V., Ivanova, S., Zhang, M., Yang, H., Botchkina, G. I.,
pnas.09061​12107​ Watkins, L. R., … Tracey, K. J. (2000). Vagus nerve stimulation at‐
Barbosa Neto, J. B., Tiba, P. A., Faturi, C. B., de Castro‐Neto, E. F., da tenuates the systemic inflammatory response to endotoxin. Nature,
Graça Naffah‐Mazacoratti, M., de Jesus Mari, J., … Suchecki, 405(6785), 458–462. https​://doi.org/10.1038/35013070
D. (2012). Stress during development alters anxiety‐like behav‐ Bravo, J. A., Forsythe, P., Chew, M. V., Escaravage, E., Savignac, H. M.,
ior and hippocampal neurotransmission in male and female rats. Dinan, T. G., … Cryan, J. F. (2011). Ingestion of Lactobacillus strain
Neuropharmacology, 62(1), 518–526. https​://doi.org/10.1016/j.neuro​ regulates emotional behavior and central GABA receptor expression
pharm.2011.09.011 in a mouse via the vagus nerve. Proceedings of the National Academy
Barnes, J., Mondelli, V., & Pariante, C. M. (2017). Genetic contributions of of Sciences, 108(38), 16050–16055. https​ ://doi.org/10.1073/
inflammation to depression. Neuropsychopharmacology, 42(1), 81–98. pnas.11029​99108​
https​://doi.org/10.1038/npp.2016.169 Burokas, A., Arboleya, S., Moloney, R. D., Peterson, V. L., Murphy, K.,
Bastiaanssen, T. F. S., Cowan, C. S. M., Claesson, M. J., Dinan, T. G., & Clarke, G., … Cryan, J. F. (2017). Targeting the microbiota‐gut‐brain
Cryan, J. F. (2019). Making sense of … the microbiome in psychiatry. axis: Prebiotics have anxiolytic and antidepressant‐like effects and
International Journal of Neuropsychopharmacology, 22(1), 37–52. https​ reverse the impact of chronic stress in mice. Biological Psychiatry,
://doi.org/10.1093/ijnp/pyy067 82(7), 472–487. https​://doi.org/10.1016/j.biops​ych.2016.12.031
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1235

Cani, P. D., Possemiers, S., Van de Wiele, T., Guiot, Y., Everard, A., Rottier, immune system subjugates the brain. Nature Reviews Neuroscience,
O., … Delzenne, N. M. (2009). Changes in gut microbiota control 9(1), 46–56. https​://doi.org/10.1038/nrn2297
inflammation in obese mice through a mechanism involving GLP‐2‐ Dantzer, R., O'Connor, J. C., Lawson, M. A., & Kelley, K. W. (2011).
driven improvement of gut permeability. Gut, 58(8), 1091–1103. Inflammation‐associated depression: From serotonin to ky‐
https​://doi.org/10.1136/gut.2008.165886 nurenine. Psychoneuroendocrinology, 36(3), 426–436. https​ ://doi.
Carabotti, M., Scirocco, A., Maselli, M. A., & Severi, C. (2015). The gut‐ org/10.1016/j.psyne​uen.2010.09.012
brain axis: Interactions between enteric microbiota, central and en‐ de Punder, K., & Pruimboom, L. (2015). Stress induces endotoxemia and
teric nervous systems. Annals of Gastroenterology, 28(2), 203–209. low‐grade inflammation by increasing barrier permeability. Frontiers
Cario, E. (2008). Barrier‐protective function of intestinal epithelial Toll‐ in Immunology, 6, 223. https​://doi.org/10.3389/fimmu.2015.00223​
like receptor 2. Mucosal Immunology, 1(Suppl 1), S62–66. https​://doi. de Vrese, M., & Schrezenmeir, J. (2008). Probiotics, prebiotics, and syn‐
org/10.1038/mi.2008.47 biotics. Advances in Biochemical Engineering/Biotechnology, 111, 1–66.
Carrellas, N. W., Biederman, J., & Uchida, M. (2017). How prevalent and https​://doi.org/10.1007/10_2008_097
morbid are subthreshold manifestations of major depression in ad‐ Derry, H. M., Padin, A. C., Kuo, J. L., Hughes, S., & Kiecolt‐Glaser, J. K.
olescents? A literature review. Journal of Affective Disorders, 210, (2015). Sex differences in depression: Does inflammation play a
166–173. https​://doi.org/10.1016/j.jad.2016.12.037 role? Current Psychiatry Reports, 17(10), 78. https​://doi.org/10.1007/
Chang, P. V., Hao, L., Offermanns, S., & Medzhitov, R. (2014). The micro‐ s11920-015-0618-5
bial metabolite butyrate regulates intestinal macrophage function via Desbonnet, L., Garrett, L., Clarke, G., Bienenstock, J., & Dinan, T. G. (2008).
histone deacetylase inhibition. Proceedings of the National Academy The probiotic Bifidobacteria infantis: An assessment of potential anti‐
of Sciences, 111(6), 2247–2252. https​://doi.org/10.1073/pnas.13222​ depressant properties in the rat. Journal of Psychiatric Research, 43(2),
69111​ 164–174. https​://doi.org/10.1016/j.jpsyc​hires.2008.03.009
Chisholm, D., Sweeny, K., Sheehan, P., Rasmussen, B., Smit, F., Cuijpers, Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryan, J. F., & Dinan, T.
P., & Saxena, S. (2016). Scaling‐up treatment of depression and anx‐ G. (2010). Effects of the probiotic Bifidobacterium infantis in the ma‐
iety: A global return on investment analysis. Lancet Psychiatry, 3(5), ternal separation model of depression. Neuroscience, 170(4), 1179–
415–424. https​://doi.org/10.1016/S2215-0366(16)30024-4 1188. https​://doi.org/10.1016/j.neuro​scien​ce.2010.08.005
Chu, H., & Mazmanian, S. K. (2013). Innate immune recognition of the Dhabhar, F. S., Malarkey, W. B., Neri, E., & McEwen, B. S. (2012). Stress‐
microbiota promotes host‐microbial symbiosis. Nature Immunology, induced redistribution of immune cells–from barracks to boule‐
14(7), 668–675. https​://doi.org/10.1038/ni.2635 vards to battlefields: A tale of three hormones–Curt Richter Award
Clarke, G., Grenham, S., Scully, P., Fitzgerald, P., Moloney, R. D., winner. Psychoneuroendocrinology, 37(9), 1345–1368. https​ ://doi.
Shanahan, F., … Cryan, J. F. (2013). The microbiome‐gut‐brain axis org/10.1016/j.psyne​uen.2012.05.008
during early life regulates the hippocampal serotonergic system in a Dobos, N., de Vries, E. F. J., Kema, I. P., Patas, K., Prins, M., Nijholt, I. M.,
sex‐dependent manner. Molecular Psychiatry, 18(6), 666–673. https​ … Eisel, U. L. M. (2012). The role of indoleamine 2,3‐dioxygenase in
://doi.org/10.1038/mp.2012.77 a mouse model of neuroinflammation‐induced depression. Journal
Cohen, N. A., Ben Ami, R., Guzner‐Gur, H., Santo, M. E., Halpern, Z., of Alzheimer's Disease, 28(4), 905–915. https​ ://doi.org/10.3233/
& Maharshak, N. (2015). Fecal microbiota transplantation for JAD-2011-111097
Clostridium difficile‐associated diarrhea. The Israel Medical Association Dominianni, C., Sinha, R., Goedert, J. J., Pei, Z., Yang, L., Hayes, R. B., &
Journal, 17(8), 510–514. Ahn, J. (2015). Sex, body mass index, and dietary fiber intake influ‐
Cohen, N. A., Livovsky, D. M., Yaakobovitch, S., Ben Yehoyada, M., Ben ence the human gut microbiome. PLoS ONE, 10(4), e0124599. https​://
Ami, R., Adler, A., … Maharshak, N. (2016). A retrospective com‐ doi.org/10.1371/journ​al.pone.0124599
parison of fecal microbial transplantation methods for recurrent Duivis, H. E., Vogelzangs, N., Kupper, N., de Jonge, P., & Penninx,
Clostridium difficile infection. The Israel Medical Association Journal, B. W. (2013). Differential association of somatic and cogni‐
18(10), 594–599. tive symptoms of depression and anxiety with inflammation:
Cohen, N. A., & Maharshak, N. (2017). Novel indications for fecal micro‐ Findings from the Netherlands Study of Depression and Anxiety
bial transplantation: Update and review of the literature. Digestive (NESDA). Psychoneuroendocrinology, 38(9), 1573–1585. https​://doi.
Diseases and Sciences, 62(5), 1131–1145. https​ ://doi.org/10.1007/ org/10.1016/j.psyne​uen.2013.01.002
s10620-017-4535-9 Eckburg, P. B., Bik, E. M., Bernstein, C. N., Purdom, E., Dethlefsen, L.,
Crumeyrolle‐Arias, M., Jaglin, M., Bruneau, A., Vancassel, S., Cardona, Sargent, M., … Relman, D. A. (2005). Diversity of the human intes‐
A., Daugé, V., … Rabot, S. (2014). Absence of the gut microbiota en‐ tinal microbial flora. Science, 308(5728), 1635–1638. https​ ://doi.
hances anxiety‐like behavior and neuroendocrine response to acute org/10.1126/scien​ce.1110591
stress in rats. Psychoneuroendocrinology, 42, 207–217. https​ ://doi. Eisenberger, N. I., Berkman, E. T., Inagaki, T. K., Rameson, L. T., Mashal, N.
org/10.1016/j.psyne​uen.2014.01.014 M., & Irwin, M. R. (2010). Inflammation‐induced anhedonia: Endotoxin
Curtin, S. C., Warner, M., & Hedegaard, H. (2016). Increase in suicide in reduces ventral striatum responses to reward. Biological Psychiatry,
the United States, 1999–2014. NCHS Data Brief (241), 1–8. 68(8), 748–754. https​://doi.org/10.1016/j.biops​ych.2010.06.010
Da Silva, S., Robbe‐Masselot, C., Ait‐Belgnaoui, A., Mancuso, A., Erny, D., Hrabě de Angelis, A. L., Jaitin, D., Wieghofer, P., Staszewski, O.,
Mercade‐Loubiere, M., Salvador‐Cartier, C., … Mercier‐Bonin, M. David, E., … Prinz, M. (2015). Host microbiota constantly control mat‐
(2014). Stress disrupts intestinal mucus barrier in rats via mucin O‐ uration and function of microglia in the CNS. Nature Neuroscience,
glycosylation shift: Prevention by a probiotic treatment. American 18(7), 965–977. https​://doi.org/10.1038/nn.4030
Journal of Physiology. Gastrointestinal and Liver Physiology, 307(4), Faravelli, C., Lo Sauro, C., Lelli, L., Pietrini, F., Lazzeretti, L., Godini,
G420–429. https​://doi.org/10.1152/ajpgi.00290.2013 L., … Ricca, V. (2012). The role of life events and HPA axis in anx‐
Da Silva, S., Robbe‐Masselot, C., Raymond, A., Mercade‐Loubière, M., iety disorders: A review. Current Pharmaceutical Design, 18(35),
Salvador‐Cartier, C., Ringot, B., … Mercier‐Bonin, M. (2015). Spatial 5663–5674.
localization and binding of the probiotic Lactobacillus farciminis to the Flandreau, E. I., Ressler, K. J., Owens, M. J., & Nemeroff, C. B. (2012).
rat intestinal mucosa: Influence of chronic stress. PLoS ONE, 10(9), Chronic overexpression of corticotropin‐releasing factor from
e0136048. https​://doi.org/10.1371/journ​al.pone.0136048 the central amygdala produces HPA axis hyperactivity and be‐
Dantzer, R., O'Connor, J. C., Freund, G. G., Johnson, R. W., & Kelley, K. havioral anxiety associated with gene‐expression changes in the
W. (2008). From inflammation to sickness and depression: When the hippocampus and paraventricular nucleus of the hypothalamus.
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1236 | PEIRCE and ALVIÑA

Psychoneuroendocrinology, 37(1), 27–38. https​ ://doi.org/10.1016/j. Hanamsagar, R., & Bilbo, S. D. (2016). Sex differences in neurodevel‐
psyne​uen.2011.04.014 opmental and neurodegenerative disorders: Focus on microglial
Fond, G., Loundou, A., Hamdani, N., Boukouaci, W., Dargel, A., Oliveira, function and neuroinflammation during development. Journal of
J., … Boyer, L. (2014). Anxiety and depression comorbidities in irri‐ Steroid Biochemistry and Molecular Biology, 160, 127–133. https​://doi.
table bowel syndrome (IBS): A systematic review and meta‐analysis. org/10.1016/j.jsbmb.2015.09.039
European Archives of Psychiatry and Clinical Neuroscience, 264(8), 651– Harrison, N. A., Brydon, L., Walker, C., Gray, M. A., Steptoe, A., &
660. https​://doi.org/10.1007/s00406-014-0502-z Critchley, H. D. (2009). Inflammation causes mood changes through
Foster, J. A., & McVey Neufeld, K. A. (2013). Gut‐brain axis: How the mi‐ alterations in subgenual cingulate activity and mesolimbic connectiv‐
crobiome influences anxiety and depression. Trends in Neurosciences, ity. Biological Psychiatry, 66(5), 407–414. https​://doi.org/10.1016/j.
36(5), 305–312. https​://doi.org/10.1016/j.tins.2013.01.005 biops​ych.2009.03.015
Friedrich, M. J. (2017). Depression is the leading cause of disability Hedegaard, H., Curtin, S. C., & Warner, M. (2018). Suicide rates in the
around the world. JAMA, 317(15), 1517. https​ ://doi.org/10.1001/ United States continue to increase. NCHS Data Brief, (309), 1–8.
jama.2017.3826 Heijtz, R. D., Wang, S., Anuar, F., Qian, Y., Bjorkholm, B., Samuelsson,
Fukuda, S., Toh, H., Hase, K., Oshima, K., Nakanishi, Y., Yoshimura, K., … A., … Pettersson, S. (2011). Normal gut microbiota modulates brain
Ohno, H. (2011). Bifidobacteria can protect from enteropathogenic development and behavior. Proceedings of the National Academy of
infection through production of acetate. Nature, 469(7331), 543– Sciences, 108(7), 3047–3052. https​://doi.org/10.1073/pnas.10105​
547. https​://doi.org/10.1038/natur​e 09646 29108​
Galea, I., Bechmann, I., & Perry, V. H. (2007). What is immune privilege Huang, R., Wang, K., & Hu, J. (2016). Effect of probiotics on depression: A
(not)? Trends in Immunology, 28(1), 12–18. https​://doi.org/10.1016/j. systematic review and meta‐analysis of randomized controlled trials.
it.2006.11.004 Nutrients, 8(8), 483. https​://doi.org/10.3390/nu808​0 483
Gao, J., Xu, K., Liu, H., Liu, G., Bai, M., Peng, C., … Yin, Y. (2018). Impact Human Microbiome Jumpstart Reference Strains Consortium, Nelson, K.
of the gut microbiota on intestinal immunity mediated by tryptophan E., Weinstock, G. M., Highlander, S. K., Worley, K. C., Creasy, H. H.,
metabolism. Frontiers in Cellular and Infection Microbiology, 8, 13. … Zhu, D. (2010). A catalog of reference genomes from the human
https​://doi.org/10.3389/fcimb.2018.00013​ microbiome. Science, 328(5981), 994–999. https​://doi.org/10.1126/
Gareau, M. G., Jury, J., MacQueen, G., Sherman, P. M., & Perdue, M. H. scien​ce.1183605
(2007). Probiotic treatment of rat pups normalises corticosterone Inagaki, H., Suzuki, T., Nomoto, K., & Yoshikai, Y. (1996). Increased sus‐
release and ameliorates colonic dysfunction induced by mater‐ ceptibility to primary infection with Listeria monocytogenes in
nal separation. Gut, 56(11), 1522–1528. https​ ://doi.org/10.1136/ germfree mice may be due to lack of accumulation of L‐selectin+
gut.2006.117176 CD44+ T cells in sites of inflammation. Infection and Immunity, 64(8),
Gatti, S., & Bartfai, T. (1993). Induction of tumor necrosis factor‐alpha 3280–3287.
mRNA in the brain after peripheral endotoxin treatment: Comparison Jiang, H., Ling, Z., Zhang, Y., Mao, H., Ma, Z., Yin, Y., … Ruan, B. (2015).
with interleukin‐1 family and interleukin‐6. Brain Research, 624(1–2), Altered fecal microbiota composition in patients with major depres‐
291–294. sive disorder. Brain, Behavior, and Immunity, 48, 186–194. https​://doi.
Gimeno, D., Kivimäki, M., Brunner, E. J., Elovainio, M., De Vogli, R., org/10.1016/j.bbi.2015.03.016
Steptoe, A., … Ferrie, J. E. (2009). Associations of C‐reactive protein Jiang, H. Y., Zhang, X., Yu, Z. H., Zhang, Z., Deng, M., Zhao, J. H., & Ruan,
and interleukin‐6 with cognitive symptoms of depression: 12‐year B. (2018). Altered gut microbiota profile in patients with generalized
follow‐up of the Whitehall II study. Psychological Medicine, 39(3), anxiety disorder. Journal of Psychiatric Research, 104, 130–136. https​
413–423. https​://doi.org/10.1017/S0033​29170​8 003723 ://doi.org/10.1016/j.jpsyc​hires.2018.07.007
Gracie, D. J., Irvine, A. J., Sood, R., Mikocka‐Walus, A., Hamlin, P. J., Jorm, A. F., Patten, S. B., Brugha, T. S., & Mojtabai, R. (2017). Has in‐
& Ford, A. C. (2017). Effect of psychological therapy on disease creased provision of treatment reduced the prevalence of common
activity, psychological comorbidity, and quality of life in inflam‐ mental disorders? Review of the evidence from four countries. World
matory bowel disease: A systematic review and meta‐analysis. Psychiatry, 16(1), 90–99. https​://doi.org/10.1002/wps.20388​
Lancet Gastroenterology & Hepatology, 2(3), 189–199. https​ ://doi. Juruena, M. F., Cleare, A. J., & Pariante, C. M. (2004). The hypothalamic
org/10.1016/S2468-1253(16)30206-0 pituitary adrenal axis, glucocorticoid receptor function and rele‐
Graessler, J., Qin, Y., Zhong, H., Zhang, J., Licinio, J., Wong, M.‐L., … vance to depression. Brazilian Journal of Psychiatry, 26(3), 189–201. /
Bornstein, S. R. (2013). Metagenomic sequencing of the human gut S1516-44462​0 0400​0300009
microbiome before and after bariatric surgery in obese patients with Kacimi, R., Giffard, R. G., & Yenari, M. A. (2011). Endotoxin‐activated
type 2 diabetes: Correlation with inflammatory and metabolic pa‐ microglia injure brain derived endothelial cells via NF‐kappaB, JAK‐
rameters. The Pharmacogenomics Journal, 13(6), 514–522. https​://doi. STAT and JNK stress kinase pathways. Journal of Inflammation, 8, 7.
org/10.1038/tpj.2012.43 https​://doi.org/10.1186/1476-9255-8-7
Graff, L. A., Walker, J. R., & Bernstein, C. N. (2009). Depression and anx‐ Kaelberer, M. M., Buchanan, K. L., Klein, M. E., Barth, B. B., Montoya,
iety in inflammatory bowel disease: A review of comorbidity and M. M., Shen, X., & Bohorquez, D. V. (2018). A gut‐brain neural circuit
management. Inflammatory Bowel Diseases, 15(7), 1105–1118. https​ for nutrient sensory transduction. Science, 361(6408), https​://doi.
://doi.org/10.1002/ibd.20873​ org/10.1126/scien​ce.aat5236
Gupta, S., Allen‐Vercoe, E., & Petrof, E. O. (2016). Fecal microbiota trans‐ Karczewski, J., Troost, F. J., Konings, I., Dekker, J., Kleerebezem, M.,
plantation: In perspective. Therapeutic Advances in Gastroenterology, Brummer, R. J., & Wells, J. M. (2010). Regulation of human epithelial
9(2), 229–239. https​://doi.org/10.1177/17562​83X15​607414 tight junction proteins by Lactobacillus plantarum in vivo and protec‐
Han, W., Tellez, L. A., Perkins, M. H., Perez, I. O., Qu, T., Ferreira, J., … tive effects on the epithelial barrier. American Journal of Physiology.
de Araujo, I. E. (2018). A neural circuit for gut‐induced reward. Cell, Gastrointestinal and Liver Physiology, 298(6), G851–859. https​://doi.
175(3), 887–888. https​://doi.org/10.1016/j.cell.2018.10.018 org/10.1152/ajpgi.00327.2009
Hanamsagar, R., Alter, M. D., Block, C. S., Sullivan, H., Bolton, J. L., & Kato‐Kataoka, A., Nishida, K., Takada, M., Kawai, M., Kikuchi‐Hayakawa,
Bilbo, S. D. (2017). Generation of a microglial developmental index H., Suda, K., … Rokutan, K. (2016). Fermented milk containing
in mice and in humans reveals a sex difference in maturation and Lactobacillus casei strain Shirota preserves the diversity of the gut
immune reactivity. Glia, 65(9), 1504–1520. https​://doi.org/10.1002/ microbiota and relieves abdominal dysfunction in healthy medi‐
glia.23176​ cal students exposed to academic stress. Applied and Environment
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1237

Microbiology, 82(12), 3649–3658. https​://doi.org/10.1128/ Luo, Y., Zeng, B., Zeng, L. I., Du, X., Li, B. O., Huo, R., … Xie, P. (2018). Gut
AEM.04134-15 microbiota regulates mouse behaviors through glucocorticoid recep‐
Kawai, T., & Akira, S. (2006). TLR signaling. Cell Death and Differentiation, tor pathway genes in the hippocampus. Translational Psychiatry, 8(1),
13(5), 816–825. https​://doi.org/10.1038/sj.cdd.4401850 187. https​://doi.org/10.1038/s41398-018-0240-5
Kegler, S. R., Stone, D. M., & Holland, K. M. (2017). Trends in sui‐ Lurie, I., Yang, Y. X., Haynes, K., Mamtani, R., & Boursi, B. (2015).
cide by level of urbanization ‐ United States, 1999–2015. MMWR. Antibiotic exposure and the risk for depression, anxiety, or psycho‐
Morbidity and Mortality Weekly Report, 66(10), 270–273. https​://doi. sis: A nested case‐control study. Journal of Clinical Psychiatry, 76(11),
org/10.15585/​mmwr.mm6610a2 1522–1528. https​://doi.org/10.4088/JCP.15m09961
Kelly, C. J., Zheng, L., Campbell, E. L., Saeedi, B., Scholz, C. C., Bayless, Ma, D., Forsythe, P., & Bienenstock, J. (2004). Live Lactobacillus rham‐
A. J., … Colgan, S. P. (2015). Crosstalk between microbiota‐derived nosus [corrected] is essential for the inhibitory effect on tumor
short‐chain fatty acids and intestinal epithelial HIF augments tissue necrosis factor alpha‐induced interleukin‐8 expression. Infection
barrier function. Cell Host & Microbe, 17(5), 662–671. https​://doi. and Immunity, 72(9), 5308–5314. https​ ://doi.org/10.1128/
org/10.1016/j.chom.2015.03.005 IAI.72.9.5308-5314.2004
Kim, M. H., Kang, S. G., Park, J. H., Yanagisawa, M., & Kim, C. H. Macpherson, A. J., & Harris, N. L. (2004). Interactions between com‐
(2013). Short‐chain fatty acids activate GPR41 and GPR43 on in‐ mensal intestinal bacteria and the immune system. Nature Reviews
testinal epithelial cells to promote inflammatory responses in Immunology, 4(6), 478–485. https​://doi.org/10.1038/nri1373
mice. Gastroenterology, 145(2), 396–406.e391‐310. https​ ://doi. Maeng, L. Y., & Milad, M. R. (2015). Sex differences in anxiety disor‐
org/10.1053/j.gastro.2013.04.056 ders: Interactions between fear, stress, and gonadal hormones.
Kim, Y. K., Na, K. S., Myint, A. M., & Leonard, B. E. (2016). The role of Hormones and Behavior, 76, 106–117. https​ ://doi.org/10.1016/j.
pro‐inflammatory cytokines in neuroinflammation, neurogenesis and yhbeh.2015.04.002
the neuroendocrine system in major depression. Progress in Neuro‐ Maes, M., Song, C., Lin, A., De Jongh, R., Van Gastel, A., Kenis, G.,
Psychopharmacology and Biological Psychiatry, 64, 277–284. https​:// … Smith, R. S. (1998). The effects of psychological stress on
doi.org/10.1016/j.pnpbp.2015.06.008 humans: Increased production of pro‐inflammatory cytokines
Koo, J. W., Russo, S. J., Ferguson, D., Nestler, E. J., & Duman, R. S. (2010). and a Th1‐like response in stress‐induced anxiety. Cytokine, 10(4),
Nuclear factor‐kappaB is a critical mediator of stress‐impaired neuro‐ 313–318.
genesis and depressive behavior. Proceedings of the National Academy of Mariadason, J. M., Barkla, D. H., & Gibson, P. R. (1997). Effect of short‐
Sciences, 107(6), 2669–2674. https​://doi.org/10.1073/pnas.09106​58107​ chain fatty acids on paracellular permeability in Caco‐2 intestinal epi‐
Kudielka, B. M., Buske‐Kirschbaum, A., Hellhammer, D. H., & Kirschbaum, thelium model. American Journal of Physiology, 272(4 Pt 1), G705–712.
C. (2004). HPA axis responses to laboratory psychosocial stress in https​://doi.org/10.1152/ajpgi.1997.272.4.G705
healthy elderly adults, younger adults, and children: Impact of age Martin‐Subero, M., Anderson, G., Kanchanatawan, B., Berk, M., & Maes,
and gender. Psychoneuroendocrinology, 29(1), 83–98. https​ ://doi. M. (2016). Comorbidity between depression and inflammatory
org/10.1016/S0306-4530(02)00146-4 bowel disease explained by immune‐inflammatory, oxidative, and ni‐
Lamers, F., Vogelzangs, N., Merikangas, K. R., de Jonge, P., Beekman, A. trosative stress; tryptophan catabolite; and gut‐brain pathways. CNS
T., & Penninx, B. W. (2013). Evidence for a differential role of HPA‐ Spectrums, 21(2), 184–198. https​://doi.org/10.1017/S1092​85291​
axis function, inflammation and metabolic syndrome in melancholic 5000449
versus atypical depression. Molecular Psychiatry, 18(6), 692–699. Maslanik, T., Tannura, K., Mahaffey, L., Loughridge, A. B., Beninson, L.,
https​://doi.org/10.1038/mp.2012.144 Ursell, L., … Fleshner, M. (2012). Commensal bacteria and MAMPs
Lamprecht, M., Bogner, S., Schippinger, G., Steinbauer, K., Fankhauser, F., are necessary for stress‐induced increases in IL‐1beta and IL‐18
Hallstroem, S., … Greilberger, J. F. (2012). Probiotic supplementation but not IL‐6, IL‐10 or MCP‐1. PLoS ONE, 7(12), e50636. https​://doi.
affects markers of intestinal barrier, oxidation, and inflammation in org/10.1371/journ​al.pone.0050636
trained men; a randomized, double‐blinded, placebo‐controlled trial. Mayor, E. (2015). Gender roles and traits in stress and health. Frontiers in
Journal of the International Society of Sports Nutrition, 9(1), 45. https​:// Psychology, 6, 779. https​://doi.org/10.3389/fpsyg.2015.00779​
doi.org/10.1186/1550-2783-9-45 Mazmanian, S. K., Liu, C. H., Tzianabos, A. O., & Kasper, D. L. (2005). An
Landgraf, R., Wigger, A., Holsboer, F., & Neumann, I. D. (1999). Hyper‐ immunomodulatory molecule of symbiotic bacteria directs matura‐
reactive hypothalamo‐pituitary‐adrenocortical axis in rats bred tion of the host immune system. Cell, 122(1), 107–118. https​://doi.
for high anxiety‐related behaviour. Journal of Neuroendocrinology, org/10.1016/j.cell.2005.05.007
11(6), 405–407. https​://doi.org/10.1046/j.1365-2826.1999.00342.x Meddings, J. B., & Swain, M. G. (2000). Environmental stress‐induced
Laye, S., Parnet, P., Goujon, E., & Dantzer, R. (1994). Peripheral admin‐ gastrointestinal permeability is mediated by endogenous glucocor‐
istration of lipopolysaccharide induces the expression of cytokine ticoids in the rat. Gastroenterology, 119(4), 1019–1028. https​://doi.
transcripts in the brain and pituitary of mice. Molecular Brain Research, org/10.1053/gast.2000.18152​
27(1), 157–162. https​://doi.org/10.1016/0169-328X(94)90197-X Mendall, M. A., & Kumar, D. (1998). Antibiotic use, childhood affluence and
Leonard, B. E. (2018). Inflammation and depression: A causal or coin‐ irritable bowel syndrome (IBS). European Journal of Gastroenterology
cidental link to the pathophysiology? Acta Neuropsychiatrica, 30(1), and Hepatology, 10(1), 59–62. https​://doi.org/10.1097/00042​737-
1–16. https​://doi.org/10.1017/neu.2016.69 19980​1000-00011​
Liang, S., Wang, T., Hu, X., Luo, J., Li, W., Wu, X., … Jin, F. (2015). Mendlewicz, J., Kriwin, P., Oswald, P., Souery, D., Alboni, S., & Brunello,
Administration of Lactobacillus helveticus NS8 improves behavioral, N. (2006). Shortened onset of action of antidepressants in major de‐
cognitive, and biochemical aberrations caused by chronic restraint pression using acetylsalicylic acid augmentation: A pilot open‐label
stress. Neuroscience, 310, 561–577. https​://doi.org/10.1016/j.neuro​ study. International Clinical Psychopharmacology, 21(4), 227–231.
scien​ce.2015.09.033 https​://doi.org/10.1097/00004​850-20060​7000-00005​
Lloyd‐Price, J., Abu‐Ali, G., & Huttenhower, C. (2016). The healthy human Messaoudi, M., Lalonde, R., Violle, N., Javelot, H., Desor, D., Nejdi, A.,
microbiome. Genome Medicine, 8(1), 51. https​ ://doi.org/10.1186/ … Cazaubiel, J. M. (2011). Assessment of psychotropic‐like proper‐
s13073-016-0307-y ties of a probiotic formulation (Lactobacillus helveticus R0052 and
Logan, A. C. (2015). Dysbiotic drift: Mental health, environmental grey Bifidobacterium longum R0175) in rats and human subjects. British
space, and microbiota. Journal of Physiological Anthropology, 34, 23. Journal of Nutrition, 105(5), 755–764. https​://doi.org/10.1017/S0007​
https​://doi.org/10.1186/s40101-015-0061-7 11451​0 004319
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1238 | PEIRCE and ALVIÑA

Mezrich, J. D., Fechner, J. H., Zhang, X., Johnson, B. P., Burlingham, W. Neufeld, K. A., Kang, N., Bienenstock, J., & Foster, J. A. (2011). Effects
J., & Bradfield, C. A. (2010). An interaction between kynurenine and of intestinal microbiota on anxiety‐like behavior. Communicative
the aryl hydrocarbon receptor can generate regulatory T cells. The & Integrative Biology, 4(4), 492–494. https​ ://doi.org/10.4161/
Journal of Immunology, 185(6), 3190–3198. https​://doi.org/10.4049/ cib.4.4.15702​
jimmu​nol.0903670 Neufeld, K. M., Kang, N., Bienenstock, J., & Foster, J. A. (2011). Reduced
Mika, A., Day, H. E. W., Martinez, A., Rumian, N. L., Greenwood, B. N., anxiety‐like behavior and central neurochemical change in germ‐free
Chichlowski, M., … Fleshner, M. (2017). Early life diets with prebi‐ mice. Neurogastroenterology and Motility, 23(3), 255–264.e119. https​
otics and bioactive milk fractions attenuate the impact of stress on ://doi.org/10.1111/j.1365-2982.2010.01620.x
learned helplessness behaviours and alter gene expression within Niraula, A., Witcher, K. G., Sheridan, J. F., & Godbout, J. P. (2019).
neural circuits important for stress resistance. European Journal of Interleukin‐6 induced by social stress promotes a unique tran‐
Neuroscience, 45(3), 342–357. https​://doi.org/10.1111/ejn.13444​ scriptional signature in the monocytes that facilitate anxiety.
Mikocka‐Walus, A. A., Turnbull, D. A., Moulding, N. T., Wilson, I. G., Biological Psychiatry, 85(8), 679–689. https​://doi.org/10.1016/j.biops​
Andrews, J. M., & Holtmann, G. J. (2007). Controversies surround‐ ych.2018.09.030
ing the comorbidity of depression and anxiety in inflammatory bowel O’Mahony, L., McCarthy, J., Kelly, P., Hurley, G., Luo, F., Chen, K., …
disease patients: A literature review. Inflammatory Bowel Diseases, Quigley, E. M. M. (2005). Lactobacillus and bifidobacterium in ir‐
13(2), 225–234. https​://doi.org/10.1002/ibd.20062​ ritable bowel syndrome: Symptom responses and relationship to
Miller, A. H., & Raison, C. L. (2016). The role of inflammation in depres‐ cytokine profiles. Gastroenterology, 128(3), 541–551. https​ ://doi.
sion: From evolutionary imperative to modern treatment target. org/10.1053/j.gastro.2004.11.050
Nature Reviews Immunology, 16(1), 22–34. https​://doi.org/10.1038/ O'Connor, J. C., Lawson, M. A., André, C., Moreau, M., Lestage, J.,
nri.2015.5 Castanon, N., … Dantzer, R. (2009). Lipopolysaccharide‐induced de‐
Miller, T. L., & Wolin, M. J. (1996). Pathways of acetate, propionate, and pressive‐like behavior is mediated by indoleamine 2,3‐dioxygenase
butyrate formation by the human fecal microbial flora. Applied and activation in mice. Molecular Psychiatry, 14(5), 511–522. https​://doi.
Environment Microbiology, 62(5), 1589–1592. org/10.1038/sj.mp.4002148
Moieni, M., & Eisenberger, N. I. (2018). Effects of inflammation on social O'Donovan, A., Hughes, B. M., Slavich, G. M., Lynch, L., Cronin, M. T.,
processes and implications for health. Annals of the New York Academy O'Farrelly, C., & Malone, K. M. (2010). Clinical anxiety, cortisol and
of Sciences, 1428(1), 5–13. https​://doi.org/10.1111/nyas.13864​ interleukin‐6: Evidence for specificity in emotion‐biology relation‐
Moieni, M., Irwin, M. R., Jevtic, I., Olmstead, R., Breen, E. C., & ships. Brain, Behavior, and Immunity, 24(7), 1074–1077. https​://doi.
Eisenberger, N. I. (2015). Sex differences in depressive and socio‐ org/10.1016/j.bbi.2010.03.003
emotional responses to an inflammatory challenge: Implications Ohland, C. L., Kish, L., Bell, H., Thiesen, A., Hotte, N., Pankiv, E., &
for sex differences in depression. Neuropsychopharmacology, 40(7), Madsen, K. L. (2013). Effects of Lactobacillus helveticus on murine
1709–1716. https​://doi.org/10.1038/npp.2015.17 behavior are dependent on diet and genotype and correlate with
Montagnoli, C., Bozza, S., Gaziano, R., Zelante, T., Bonifazi, P., Moretti, alterations in the gut microbiome. Psychoneuroendocrinology, 38(9),
S., … Romani, L. (2006). Immunity and tolerance to Aspergillus fumi‐ 1738–1747. https​://doi.org/10.1016/j.psyne​uen.2013.02.008
gatus. Novartis Foundation Symposium, 279, 66–77; discussion 77–69, Ostman, S., Rask, C., Wold, A. E., Hultkrantz, S., & Telemo, E. (2006).
216–219. Impaired regulatory T cell function in germ‐free mice. European
Morgan, X. C., Tickle, T. L., Sokol, H., Gevers, D., Devaney, K. L., Ward, D. Journal of Immunology, 36(9), 2336–2346. https​://doi.org/10.1002/
V., … Huttenhower, C. (2012). Dysfunction of the intestinal microbi‐ eji.20053​5244
ome in inflammatory bowel disease and treatment. Genome Biology, Overman, E. L., Rivier, J. E., & Moeser, A. J. (2012). CRF induces intesti‐
13(9), R79. https​://doi.org/10.1186/gb-2012-13-9-r79 nal epithelial barrier injury via the release of mast cell proteases and
Morris, G., Berk, M., Carvalho, A., Caso, J. R., Sanz, Y., Walder, K., & TNF‐alpha. PLoS ONE, 7(6), e39935. https​://doi.org/10.1371/journ​
Maes, M. (2017). The role of the microbial metabolites including al.pone.0039935
tryptophan catabolites and short chain fatty acids in the patho‐ Pace, T. W., Mletzko, T. C., Alagbe, O., Musselman, D. L., Nemeroff, C.
physiology of immune‐inflammatory and neuroimmune disease. B., Miller, A. H., & Heim, C. M. (2006). Increased stress‐induced in‐
Molecular Neurobiology, 54(6), 4432–4451. https​://doi.org/10.1007/ flammatory responses in male patients with major depression and
s12035-016-0004-2 increased early life stress. American Journal of Psychiatry, 163(9),
Müller, N., Schwarz, M. J., Dehning, S., Douhe, A., Cerovecki, A., 1630–1633. https​://doi.org/10.1176/ajp.2006.163.9.1630
Goldstein‐Müller, B., … Riedel, M. (2006). The cyclooxygenase‐2 in‐ Pariante, C. M. (2003). Depression, stress and the adrenal axis. Journal of
hibitor celecoxib has therapeutic effects in major depression: Results Neuroendocrinology, 15(8), 811–812.
of a double‐blind, randomized, placebo controlled, add‐on pilot study Park, A. J., Collins, J., Blennerhassett, P. A., Ghia, J. E., Verdu, E. F., Bercik,
to reboxetine. Molecular Psychiatry, 11(7), 680–684. https​ ://doi. P., & Collins, S. M. (2013). Altered colonic function and microbiota
org/10.1038/sj.mp.4001805 profile in a mouse model of chronic depression. Neurogastroenterology
Nardi, R. M., Vieira, E. C., Crocco‐Afonso, L. C., Silva, M. E., Bambirra, E. and Motility, 25(9), 733–e575. https​://doi.org/10.1111/nmo.12153​
A., Andrade, A. M., & Nicoli, J. R. (1991). Bacteriological and immu‐ Parrott, J. M., Redus, L., Santana‐Coelho, D., Morales, J., Gao, X., &
nological aspects of conventional and germfree mice infected with O'Connor, J. C. (2016). Neurotoxic kynurenine metabolism is in‐
Salmonella typhimurium. Revista Latinoamericana De Microbiologia, creased in the dorsal hippocampus and drives distinct depressive
33(4), 239–243. behaviors during inflammation. Translational Psychiatry, 6(10), e918.
Naseribafrouei, A., Hestad, K., Avershina, E., Sekelja, M., Linlokken, A., https​://doi.org/10.1038/tp.2016.200
Wilson, R., & Rudi, K. (2014). Correlation between the human fecal Pavlov, V. A., Parrish, W. R., Rosas‐Ballina, M., Ochani, M., Puerta, M.,
microbiota and depression. Neurogastroenterology and Motility, 26(8), Ochani, K., … Tracey, K. J. (2009). Brain acetylcholinesterase activ‐
1155–1162. https​://doi.org/10.1111/nmo.12378​ ity controls systemic cytokine levels through the cholinergic anti‐
Nemeth, C. L., Reddy, R., Bekhbat, M., Bailey, J., & Neigh, G. N. (2014). inflammatory pathway. Brain, Behavior, and Immunity, 23(1), 41–45.
Microglial activation occurs in the absence of anxiety‐like behav‐ https​://doi.org/10.1016/j.bbi.2008.06.011
ior following microembolic stroke in female, but not male, rats. Peng, L., Li, Z. R., Green, R. S., Holzman, I. R., & Lin, J. (2009). Butyrate
Journal of Neuroinflammation, 11, 174. https​ ://doi.org/10.1186/ enhances the intestinal barrier by facilitating tight junction assem‐
s12974-014-0174-7 bly via activation of AMP‐activated protein kinase in Caco‐2 cell
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1239

monolayers. Journal of Nutrition, 139(9), 1619–1625. https​ ://doi. and mental illness: Mechanisms and pathways. Molecular Psychiatry,
org/10.3945/jn.109.104638 21(6), 738–748. https​://doi.org/10.1038/mp.2016.50
Perez‐Burgos, A., Wang, B., Mao, Y. K., Mistry, B., McVey Neufeld, K. Romani, L., Zelante, T., Luca, A. D., Iannitti, R. G., Moretti, S., Bartoli, A., …
A., Bienenstock, J., & Kunze, W. (2013). Psychoactive bacteria Puccetti, P. (2014). Microbiota control of a tryptophan‐AhR pathway
Lactobacillus rhamnosus (JB‐1) elicits rapid frequency facilitation in disease tolerance to fungi. European Journal of Immunology, 44(11),
in vagal afferents. American Journal of Physiology. Gastrointestinal 3192–3200. https​://doi.org/10.1002/eji.20134 ​4 406
and Liver Physiology, 304(2), G211–220. https​ ://doi.org/10.1152/ Rook, G. A., Raison, C. L., & Lowry, C. A. (2013). Childhood microbial
ajpgi.00128.2012 experience, immunoregulation, inflammation and adult susceptibility
Pitsavos, C., Panagiotakos, D. B., Papageorgiou, C., Tsetsekou, E., to psychosocial stressors and depression in rich and poor countries.
Soldatos, C., & Stefanadis, C. (2006). Anxiety in relation to inflam‐ Evolution, Medicine, and Public Health, 2013(1), 14–17. https​ ://doi.
mation and coagulation markers, among healthy adults: The ATTICA org/10.1093/emph/eos005
study. Atherosclerosis, 185(2), 320–326. https​ ://doi.org/10.1016/j. Rosenfeldt, V., Benfeldt, E., Valerius, N. H., Paerregaard, A., & Michaelsen,
ather​oscle​rosis.2005.06.001 K. F. (2004). Effect of probiotics on gastrointestinal symptoms and
Povoleri, G. A. M., Nova‐Lamperti, E., Scottà, C., Fanelli, G., Chen, small intestinal permeability in children with atopic dermatitis.
Y.‐C., Becker, P. D., … Afzali, B. (2018). Human retinoic acid‐regu‐ Journal of Pediatrics, 145(5), 612–616. https​ ://doi.org/10.1016/j.
lated CD161(+) regulatory T cells support wound repair in intes‐ jpeds.2004.06.068
tinal mucosa. Nature Immunology, 19(12), 1403–1414. https​ ://doi. Rothhammer, V., Mascanfroni, I. D., Bunse, L., Takenaka, M. C.,
org/10.1038/s41590-018-0230-z Kenison, J. E., Mayo, L., … Quintana, F. J. (2016). Type I interfer‐
Proctor, L. M. (2016). The National Institutes of Health Human ons and microbial metabolites of tryptophan modulate astrocyte
Microbiome Project. Seminars in Fetal & Neonatal Medicine, 21(6), activity and central nervous system inflammation via the aryl hy‐
368–372. https​://doi.org/10.1016/j.siny.2016.05.002 drocarbon receptor. Nature Medicine, 22(6), 586–597. https​://doi.
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K. S., Manichanh, C., … org/10.1038/nm.4106
Wang, J. (2010). A human gut microbial gene catalogue established Round, J. L., Lee, S. M., Li, J., Tran, G., Jabri, B., Chatila, T. A., &
by metagenomic sequencing. Nature, 464(7285), 59–65. https​://doi. Mazmanian, S. K. (2011). The Toll‐like receptor 2 pathway es‐
org/10.1038/natur​e 08821 tablishes colonization by a commensal of the human microbi‐
Qin, L., Wu, X., Block, M. L., Liu, Y., Breese, G. R., Hong, J.‐S., … Crews, ota. Science, 332(6032), 974–977. https​ ://doi.org/10.1126/scien​
F. T. (2007). Systemic LPS causes chronic neuroinflammation and ce.1206095
progressive neurodegeneration. Glia, 55(5), 453–462. https​ ://doi. Savignac, H. M., Kiely, B., Dinan, T. G., & Cryan, J. F. (2014). Bifidobacteria
org/10.1002/glia.20467​ exert strain‐specific effects on stress‐related behavior and physiol‐
Qiu, J., Heller, J. J., Guo, X., Chen, Z. M., Fish, K., Fu, Y. X., & Zhou, L. ogy in BALB/c mice. Neurogastroenterology and Motility, 26(11), 1615–
(2012). The aryl hydrocarbon receptor regulates gut immunity 1627. https​://doi.org/10.1111/nmo.12427​
through modulation of innate lymphoid cells. Immunity, 36(1), 92– Savignac, H. M., Tramullas, M., Kiely, B., Dinan, T. G., & Cryan, J. F.
104. https​://doi.org/10.1016/j.immuni.2011.11.011 (2015). Bifidobacteria modulate cognitive processes in an anxious
Quan, N., Stern, E. L., Whiteside, M. B., & Herkenham, M. (1999). mouse strain. Behavioral Brain Research, 287, 59–72. https​ ://doi.
Induction of pro‐inflammatory cytokine mRNAs in the brain after org/10.1016/j.bbr.2015.02.044
peripheral injection of subseptic doses of lipopolysaccharide in Schirmer, M., Smeekens, S. P., Vlamakis, H., Jaeger, M., Oosting, M.,
the rat. Journal of Neuroimmunology, 93(1–2), 72–80. https​ ://doi. Franzosa, E. A., … Xavier, R. J. (2016). Linking the human gut micro‐
org/10.1016/S0165-5728(98)00193-3 biome to inflammatory cytokine production capacity. Cell, 167(4),
Quintana, F. J. (2013). The aryl hydrocarbon receptor: A molec‐ 1125–1136.e1128. https​://doi.org/10.1016/j.cell.2016.10.020
ular pathway for the environmental control of the immune Schrezenmeir, J., & de Vrese, M. (2001). Probiotics, prebiotics, and synbi‐
response. Immunology, 138(3), 183–189. https​ ://doi.org/10.1111/ otics–approaching a definition. American Journal of Clinical Nutrition,
imm.12046​ 73(2 Suppl), 361S–364S. https​://doi.org/10.1093/ajcn/73.2.361s
Rai, D., Zitko, P., Jones, K., Lynch, J., & Araya, R. (2013). Country‐ and in‐ Schwarcz, R., Bruno, J. P., Muchowski, P. J., & Wu, H. Q. (2012).
dividual‐level socioeconomic determinants of depression: Multilevel Kynurenines in the mammalian brain: When physiology meets pa‐
cross‐national comparison. British Journal of Psychiatry, 202(3), 195– thology. Nature Reviews Neuroscience, 13(7), 465–477. https​://doi.
203. https​://doi.org/10.1192/bjp.bp.112.112482 org/10.1038/nrn3257
Raison, C. L., & Miller, A. H. (2013). The evolutionary significance of Segata, N. (2015). Gut microbiome: Westernization and the disappear‐
depression in Pathogen Host Defense (PATHOS‐D). Molecular ance of intestinal diversity. Current Biology, 25(14), R611–613. https​
Psychiatry, 18(1), 15–37. https​://doi.org/10.1038/mp.2012.2 ://doi.org/10.1016/j.cub.2015.05.040
Rathod, K. S., Kapil, V., Velmurugan, S., Khambata, R. S., Siddique, U., Sender, R., Fuchs, S., & Milo, R. (2016). Revised estimates for the num‐
Khan, S., … Ahluwalia, A. (2017). Accelerated resolution of inflam‐ ber of human and bacteria cells in the body. PLoS Biology, 14(8),
mation underlies sex differences in inflammatory responses in hu‐ e1002533. https​://doi.org/10.1371/journ​al.pbio.1002533
mans. Journal of Clinical Investigation, 127(1), 169–182. https​://doi. Shen, Y., Giardino Torchia, M. L., Lawson, G. W., Karp, C. L., Ashwell,
org/10.1172/JCI89429 J. D., & Mazmanian, S. K. (2012). Outer membrane vesicles of a
Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A., & human commensal mediate immune regulation and disease protec‐
Pollmacher, T. (2001). Cytokine‐associated emotional and cognitive tion. Cell Host & Microbe, 12(4), 509–520. https​://doi.org/10.1016/j.
disturbances in humans. Archives of General Psychiatry, 58(5), 445– chom.2012.08.004
452. https​://doi.org/10.1001/archp​syc.58.5.445 Silk, D. B., Davis, A., Vulevic, J., Tzortzis, G., & Gibson, G. R. (2009).
Reigstad, C. S., Salmonson, C. E., Rainey, J. F. 3rd, Szurszewski, J. H., Clinical trial: The effects of a trans‐galactooligosaccharide prebiotic
Linden, D. R., Sonnenburg, J. L., … Kashyap, P. C. (2015). Gut mi‐ on faecal microbiota and symptoms in irritable bowel syndrome.
crobes promote colonic serotonin production through an effect of Alimentary Pharmacology & Therapeutics, 29(5), 508–518. https​://doi.
short‐chain fatty acids on enterochromaffin cells. The FASEB Journal, org/10.1111/j.1365-2036.2008.03911.x
29(4), 1395–1403. https​://doi.org/10.1096/fj.14-259598 Singh, N., Gurav, A., Sivaprakasam, S., Brady, E., Padia, R., Shi, H., …
Rogers, G. B., Keating, D. J., Young, R. L., Wong, M. L., Licinio, J., & Ganapathy, V. (2014). Activation of Gpr109a, receptor for nia‐
Wesselingh, S. (2016). From gut dysbiosis to altered brain function cin and the commensal metabolite butyrate, suppresses colonic
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1240 | PEIRCE and ALVIÑA

inflammation and carcinogenesis. Immunity, 40(1), 128–139. https​:// Thion, M. S., Low, D., Silvin, A., Chen, J., Grisel, P., Schulte‐Schrepping, J.,
doi.org/10.1016/j.immuni.2013.12.007 … Garel, S. (2018). Microbiome influences prenatal and adult microg‐
Singh, N., Thangaraju, M., Prasad, P. D., Martin, P. M., Lambert, N. A., lia in a sex‐specific manner. Cell, 172(3), 500–516.e516. https​://doi.
Boettger, T., … Ganapathy, V. (2010). Blockade of dendritic cell de‐ org/10.1016/j.cell.2017.11.042
velopment by bacterial fermentation products butyrate and propio‐ Tognini, P. (2017). Gut microbiota: A potential regulator of neurode‐
nate through a transporter (Slc5a8)‐dependent inhibition of histone velopment. Frontiers in Cellular Neuroscience, 11, 25. https​ ://doi.
deacetylases. Journal of Biological Chemistry, 285(36), 27601–27608. org/10.3389/fncel.2017.00025​
https​://doi.org/10.1074/jbc.M110.102947 Troy, E. B., & Kasper, D. L. (2010). Beneficial effects of Bacteroides fragilis
Smith, P. M., Howitt, M. R., Panikov, N., Michaud, M., Gallini, C. A., polysaccharides on the immune system. Frontiers in Bioscience, 15,
Bohlooly‐Y, M., … Garrett, W. S. (2013). The microbial metabo‐ 25–34. https​://doi.org/10.2741/3603
lites, short‐chain fatty acids, regulate colonic Treg cell homeosta‐ Turner‐Cobb, J. M., Osborn, M., da Silva, L., Keogh, E., & Jessop, D. S.
sis. Science, 341(6145), 569–573. https​ ://doi.org/10.1126/scien​ (2010). Sex differences in hypothalamic‐pituitary‐adrenal axis func‐
ce.1241165 tion in patients with chronic pain syndrome. Stress, 13(4), 292–300.
Sominsky, L., Fuller, E. A., Bondarenko, E., Ong, L. K., Averell, L., Nalivaiko, https​://doi.org/10.3109/10253​89090​3524785
E., … Hodgson, D. M. (2013). Functional programming of the auto‐ Twenge, J. M., Cooper, A. B., Joiner, T. E., Duffy, M. E., & Binau, S. G.
nomic nervous system by early life immune exposure: Implications (2019). Age, period, and cohort trends in mood disorder indicators
for anxiety. PLoS ONE, 8(3), e57700. https​://doi.org/10.1371/journ​ and suicide‐related outcomes in a nationally representative dataset,
al.pone.0057700 2005–2017. Journal of Abnormal Psychology, 128(3), 185–199. https​://
Songtachalert, T., Roomruangwong, C., Carvalho, A. F., Bourin, M., & doi.org/10.1037/abn00​0 0410​
Maes, M. (2018). Anxiety disorders: Sex differences in serotonin and Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L., & Hooper, L.
tryptophan metabolism. Current Topics in Medicinal Chemistry, 18(19), V. (2008). Paneth cells directly sense gut commensals and maintain
1704–1715. https​://doi.org/10.2174/15680​26618​66618​11150​ homeostasis at the intestinal host‐microbial interface. Proceedings of
93136​ the National Academy of Sciences, 105(52), 20858–20863. https​://doi.
Steenbergen, L., Sellaro, R., van Hemert, S., Bosch, J. A., & Colzato, org/10.1073/pnas.08087​23105​
L. S. (2015). A randomized controlled trial to test the effect of van de Wouw, M., Boehme, M., Lyte, J. M., Wiley, N., Strain, C., O'Sullivan,
multispecies probiotics on cognitive reactivity to sad mood. Brain, O., … Cryan, J. F. (2018). Short‐chain fatty acids: Microbial metabo‐
Behavior, and Immunity, 48, 258–264. https​ ://doi.org/10.1016/j. lites that alleviate stress‐induced brain‐gut axis alterations. Journal of
bbi.2015.04.003 Physiology, 596(20), 4923–4944. https​://doi.org/10.1113/JP276431
Steptoe, A., Hamer, M., & Chida, Y. (2007). The effects of acute psy‐ van den Biggelaar, A. H., Gussekloo, J., de Craen, A. J., Frolich, M., Stek,
chological stress on circulating inflammatory factors in humans: A M. L., van der Mast, R. C., & Westendorp, R. G. (2007). Inflammation
review and meta‐analysis. Brain, Behavior, and Immunity, 21(7), 901– and interleukin‐1 signaling network contribute to depressive symp‐
912. https​://doi.org/10.1016/j.bbi.2007.03.011 toms but not cognitive decline in old age. Experimental Gerontology,
Stockinger, S., Duerr, C. U., Fulde, M., Dolowschiak, T., Pott, J., Yang, I., … 42(7), 693–701. https​://doi.org/10.1016/j.exger.2007.01.011
Hornef, M. W. (2014). TRIF signaling drives homeostatic intestinal ep‐ Vanuytsel, T., van Wanrooy, S., Vanheel, H., Vanormelingen, C.,
ithelial antimicrobial peptide expression. The Journal of Immunology, Verschueren, S., Houben, E., … Tack, J. (2014). Psychological stress
193(8), 4223–4234. https​://doi.org/10.4049/jimmu​nol.1302708 and corticotropin‐releasing hormone increase intestinal permeability
Strauch, U. G., Obermeier, F., Grunwald, N., Gurster, S., Dunger, N., in humans by a mast cell‐dependent mechanism. Gut, 63(8), 1293–
Schultz, M., … Rath, H. C. (2005). Influence of intestinal bacteria on in‐ 1299. https​://doi.org/10.1136/gutjnl-2013-305690
duction of regulatory T cells: Lessons from a transfer model of colitis. Varghese, F. P., & Brown, E. S. (2001). The hypothalamic‐pituitary‐ad‐
Gut, 54(11), 1546–1552. https​://doi.org/10.1136/gut.2004.059451 renal axis in major depressive disorder: A brief primer for primary
Strike, P. C., Wardle, J., & Steptoe, A. (2004). Mild acute inflamma‐ care physicians. The Primary Care Companion to the Journal of Clinical
tory stimulation induces transient negative mood. Journal of Psychiatry, 3(4), 151–155. https​://doi.org/10.4088/PCC.v03n0401
Psychosomatic Research, 57(2), 189–194. https​ ://doi.org/10.1016/ Venkatesh, M., Mukherjee, S., Wang, H., Li, H., Sun, K., Benechet, A. P.,
S0022-3999(03)00569-5 … Mani, S. (2014). Symbiotic bacterial metabolites regulate gastro‐
Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X.‐N., … Koga, intestinal barrier function via the xenobiotic sensor PXR and Toll‐
Y. (2004). Postnatal microbial colonization programs the hypotha‐ like receptor 4. Immunity, 41(2), 296–310. https​://doi.org/10.1016/j.
lamic‐pituitary‐adrenal system for stress response in mice. Journal immuni.2014.06.014
of Physiology, 558(Pt 1), 263–275. https​ ://doi.org/10.1113/jphys​ Walker, A. K., Budac, D. P., Bisulco, S., Lee, A. W., Smith, R. A., Beenders,
iol.2004.063388 B., … Dantzer, R. (2013). NMDA receptor blockade by ketamine
Suez, J., Zmora, N., Zilberman‐Schapira, G., Mor, U., Dori‐Bachash, M., abrogates lipopolysaccharide‐induced depressive‐like behavior in
Bashiardes, S., … Elinav, E. (2018). Post‐antibiotic gut mucosal mi‐ C57BL/6J mice. Neuropsychopharmacology, 38(9), 1609–1616. https​
crobiome reconstitution is impaired by probiotics and improved ://doi.org/10.1038/npp.2013.71
by autologous FMT. Cell, 174(6), 1406–1423.e1416. https​ ://doi. Wallace, C. J. K., & Milev, R. (2017). The effects of probiotics on depres‐
org/10.1016/j.cell.2018.08.047 sive symptoms in humans: A systematic review. Annals of General
Suzuki, T., Yoshida, S., & Hara, H. (2008). Physiological concentrations of Psychiatry, 16, 14. https​://doi.org/10.1186/s12991-017-0138-2
short‐chain fatty acids immediately suppress colonic epithelial per‐ Weinberger, A. H., Gbedemah, M., Martinez, A. M., Nash, D., Galea, S.,
meability. British Journal of Nutrition, 100(2), 297–305. https​://doi. & Goodwin, R. D. (2018). Trends in depression prevalence in the
org/10.1017/S0007​11450​8888733 USA from 2005 to 2015: Widening disparities in vulnerable groups.
Thion, M. S., & Garel, S. (2018). Microglia under the spotlight: Activity Psychological Medicine, 48(8), 1308–1315. https​://doi.org/10.1017/
and complement‐dependent engulfment of synapses. Trends S0033​29171​7002781
in Neurosciences, 41(6), 332–334. https​ ://doi.org/10.1016/j. Wichers, M. C., Koek, G. H., Robaeys, G., Verkerk, R., Scharpe, S., & Maes,
tins.2018.03.017 M. (2005). IDO and interferon‐alpha‐induced depressive symptoms:
Thion, M. S., Ginhoux, F., & Garel, S. (2018). Microglia and early brain de‐ A shift in hypothesis from tryptophan depletion to neurotoxic‐
velopment: An intimate journey. Science, 362(6411), 185–189. https​ ity. Molecular Psychiatry, 10(6), 538–544. https​://doi.org/10.1038/
://doi.org/10.1126/scien​ce.aat0474 sj.mp.4001600
10974547, 2019, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jnr.24476 by Spanish Cochrane National Provision (Ministerio de Sanidad), Wiley Online Library on [20/11/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PEIRCE and ALVIÑA | 1241

Winter, C., von Rumohr, A., Mundt, A., Petrus, D., Klein, J., Lee, T., … Zeng, J., Li, Y. Q., Zuo, X. L., Zhen, Y. B., Yang, J., & Liu, C. H. (2008).
Juckel, G. (2007). Lesions of dopaminergic neurons in the substan‐ Clinical trial: Effect of active lactic acid bacteria on mucosal barrier
tia nigra pars compacta and in the ventral tegmental area enhance function in patients with diarrhoea‐predominant irritable bowel syn‐
depressive‐like behavior in rats. Behavioral Brain Research, 184(2), drome. Alimentary Pharmacology & Therapeutics, 28(8), 994–1002.
133–141. https​://doi.org/10.1016/j.bbr.2007.07.002 https​://doi.org/10.1111/j.1365-2036.2008.03818.x
Wright, C. E., Strike, P. C., Brydon, L., & Steptoe, A. (2005). Acute inflam‐ Zhang, Y., Liu, L., Liu, Y.‐Z., Shen, X.‐L., Wu, T.‐Y., Zhang, T., … Jiang, C.‐L.
mation and negative mood: Mediation by cytokine activation. Brain, (2015). NLRP3 inflammasome mediates chronic mild stress‐induced
Behavior, and Immunity, 19(4), 345–350. https​://doi.org/10.1016/j. depression in mice via neuroinflammation. International Journal of
bbi.2004.10.003 Neuropsychopharmacology, 18(8). https​://doi.org/10.1093/ijnp/pyv006
Xiao, H., Carney, D. M., Youn, S. J., Janis, R. A., Castonguay, L. G., Hayes, Zheng, P., Zeng, B., Zhou, C., Liu, M., Fang, Z., Xu, X., … Xie, P. (2016). Gut
J. A., & Locke, B. D. (2017). Are we in crisis? National mental health microbiome remodeling induces depressive‐like behaviors through
and treatment trends in college counseling centers. Psychological a pathway mediated by the host's metabolism. Molecular Psychiatry,
Services, 14(4), 407–415. https​://doi.org/10.1037/ser00​0 0130​ 21(6), 786–796. https​://doi.org/10.1038/mp.2016.44
Yu, Y., Liu, Z.‐Q., Liu, X.‐Y., Yang, L. I., Geng, X.‐R., Yang, G., … Yang, Zhong, Y., Cai, D., Cai, W., Geng, S., Chen, L., & Han, T. (2009). Protective
P.‐C. (2013). Stress‐derived corticotropin releasing factor breaches effect of galactooligosaccharide‐supplemented enteral nutrition
epithelial endotoxin tolerance. PLoS ONE, 8(6), e65760. https​://doi. on intestinal barrier function in rats with severe acute pancreati‐
org/10.1371/journ​al.pone.0065760 tis. Clinical Nutrition, 28(5), 575–580. https​ ://doi.org/10.1016/j.
Yurkovetskiy, L., Burrows, M., Khan, A. A., Graham, L., Volchkov, P., clnu.2009.04.026
Becker, L., … Chervonsky, A. V. (2013). Gender bias in autoimmunity
is influenced by microbiota. Immunity, 39(2), 400–412. https​://doi.
How to cite this article: Peirce JM, Alviña K. The role of
org/10.1016/j.immuni.2013.08.013
Zelante, T., Iannitti, R. G., Fallarino, F., Gargaro, M., De Luca, A., Moretti, inflammation and the gut microbiome in depression and
S., … Romani, L. (2014). Tryptophan feeding of the IDO1‐AhR axis in anxiety. J Neuro Res. 2019;97:1223–1241. https​://doi.
host‐microbial symbiosis. Frontiers in Immunology, 5, 640. https​://doi. org/10.1002/jnr.24476​
org/10.3389/fimmu.2014.00640​

You might also like