Sie sind auf Seite 1von 22

809379

research-article2019
PPSXXX10.1177/1745691618809379Smith, WisselMicrobes and the Mind

ASSOCIATION FOR
PSYCHOLOGICAL SCIENCE

Perspectives on Psychological Science

Microbes and the Mind: How Bacteria 2019, Vol. 14(3) 397­–418
© The Author(s) 2019
Article reuse guidelines:
Shape Affect, Neurological Processes, sagepub.com/journals-permissions
DOI: 10.1177/1745691618809379
https://doi.org/10.1177/1745691618809379

Cognition, Social Relationships, www.psychologicalscience.org/PPS

Development, and Pathology

Leigh K. Smith1 and Emily F. Wissel2


1
Department of Psychology, University of California, Davis, and 2School of Nursing, Emory University

Abstract
Recent data suggest that the human body is not so exclusively human after all. Specifically, humans share their bodies
with approximately 10 trillion microorganisms, collectively known as the microbiome. Chief among these microbes
are bacteria, and there is a growing consensus that they are critical to virtually all facets of normative functioning.
This article reviews the ways in which bacteria shape affect, neurological processes, cognition, social relationships,
development, and psychological pathology. To date, the vast majority of research on interactions between microbes
and humans has been conducted by scientists outside the field of psychology, despite the fact that psychological
scientists are experts in many of the topics being explored. This review aims to orient psychological scientists to the
most relevant research and perspectives regarding the microbiome so that we might contribute to the now widespread,
interdisciplinary effort to understand the relationship between microbes and the mind.

Keywords
microbiome, bacteria, emotion, gut–brain axis, cognition, social relationships, development, mental health

It is widely accepted that mental states are influenced science). Each section presents a rigorous but concise
by bodily processes (Blascovich & Mendes, 2010), but overview of how bacteria shape variables typically
it is time to expand the concept of “body” to include investigated in each subfield. Collectively, therefore,
the 10 trillion microorganisms that live on and inside this review aims to be accessible to a diverse audience
of humans (P. Kramer & Bressan, 2015; Sender, Fuchs, of researchers. Of key importance is that this review
& Milo, 2016). Collectively known as the microbiome, highlights a large collection of nonclinical findings,
these organisms include fungi, archaea, and viruses, stepping beyond the notion that contact with bacteria
but chief among them are bacteria—which, as recent is harmful (R. T. Liu, 2017) and into the fascinating body
investigations have uncovered, seem to influence virtu- of work that demonstrates how bacteria critically sup-
ally all facets of the human experience (Anderson, port normative, everyday functioning. Specifically, we
Cryan, & Dinan, 2017). This review outlines the ways review how an array of psychological experiences are
in which bacteria shape affect, cognition, neurological influenced by both commensal bacteria (those that tend
processing, social relationships, development, and psy- to be beneficial or harmless, e.g., Lactobacillus species)
chological pathologies. In an interdisciplinary effort, and pathogenic bacteria (those that cause disease or
biological and clinical scientists have carefully seeded
this new research terrain, and psychological scientists
Corresponding Authors:
have an important role to play in cultivating the soil— Leigh K. Smith, University of California, Davis, Department of
both theoretically and empirically. Psychology, 135 Young Hall, 1 Shields Ave., Davis, CA 95616
To efficiently orient psychological scientists to the E-mail: leismith@ucdavis.edu
most relevant research and perspectives regarding the Emily F. Wissel, Emory University, School of Nursing, Wesley Woods
microbiome, this review is organized by psychological Hospital Office A2076, 1821 Clifton Rd. NE, Atlanta, GA 30329
subfield (e.g., affective science, neuroscience, cognitive E-mail: ewissel@emory.edu
398 Smith, Wissel

dysfunction, e.g., Clostridium difficile). An important induced colitis—inflammation of the colon—led to


note, however, is that whether bacteria play a beneficial decreases in beneficial intestinal bacteria in rodents and
or harmful role is not exclusively determined by their increases in their behaviors associated with negative
type. Factors such as their proportion relative to other affect, such as anxiety (Bercik, Park, et al., 2011; Davis
microbes, the physiology of the host, and the external et al., 2016) and depression (Slyepchenko et al., 2016).
environment all contribute to the type of effects they Bacteria are also related to chronic, trait-like affective
will exert. Last, because of recent advances in both states. For example, Martin et al. (2009) reported that
animal and human experimental research, a substantial human subjects who had chronically low or high levels
amount of the work covered in this review moves past of anxiety had distinct metabolic profiles indicative of
correlational associations to include direct, causal influ- different gut microbial activity. Furthermore, extended
ences of the microbiome on psychological and neuro- dietary interventions that directly influenced gut motil-
logical processes. ity partially normalized these trait-like anxiety-related
differences. Recent studies have also shown that individu-
als who experience chronic negative affect (as indexed
Emotion and Affective Science
via personality traits such as high neuroticism) have gut
Bacteria play an important role in shaping affective microbiota that are distinct from individuals whose per-
states. Affect forms the basis for processing a wide range sonalities are characterized by less negative affect and
of sensory and physiological information (L. F. Barrett lower levels of neuroticism (H. Kim et al., 2018).
& Bliss-Moreau, 2009; Siegel, Wormwood, Quigley, & In a similar demonstration from animal research,
Barrett, 2018), as well as developing a sense of self colonizing phenotypically anxious mice (BALB/c mice)
(Damasio, 2003), establishing meaningful social bonds with the microbiota of phenotypically gregarious mice
(Algoe, 2012), and promoting approach versus avoid- (Swiss mice) resulted in the typically anxious mice
ance behaviors (Phaf, Mohr, Rotteveel, & Wicherts, becoming more gregarious. The reverse was also true—
2014). The brain regions that support affective states colonizing gregarious mice with the microbiota of anx-
also support essential physiological functioning (e.g., ious mice induced heightened anxiety-like behaviors
allostasis; Kleckner et al., 2017), suggesting that affect (Bercik, Park, et  al., 2011). Although conducted with
is deeply embodied and fundamentally intertwined with animals, this study and others like it (e.g., Collins, Kassam,
humans’ physical preservation and survival. The micro- & Bercik, 2013) demonstrate that trait-like affective states
biome’s ability to shape affect and emotion therefore can be “transferred” from one individual to another via
indicates that the influence of bacteria may spill over manipulation of the microbiome.
into every aspect of what it means to be a conscious, In one of the first studies to examine how bacteria
living organism. are related to acute affective experiences, Lencner et al.
(1984) reported that astronauts experiencing momen-
tary emotional distress during shuttle launches showed
Chronic and acute affective states
distinct alterations in the beneficial bacteria present in
At the most general level, maintaining a robust popula- their saliva and feces. Experiments with rodents have
tion of beneficial bacteria in the gut can improve mood repeatedly demonstrated similar findings. Acute nega-
and well-being, whereas the depletion of these bacteria tive affect brought on by momentary stress (e.g., fear
can negatively affect well-being (Cryan & Dinan, 2012; conditioning, forced swim tests, stress-induced hyper-
Cryan & O’Mahony, 2011; Forsythe, Sudo, Dinan, Taylor, thermia) is associated with or directly caused by the
& Bienenstock, 2010). For example, in a double-blind overgrowth of pathogenic bacteria or the depletion of
placebo-controlled study, medical students consumed beneficial bacteria (Bravo et al., 2011; McLean, Bergonzelli,
a daily drink composed of probiotics, which are supple- Collins, & Bercik, 2012; Neufeld, Kang, Bienenstock, &
ments containing live, beneficial bacteria, for 3 months Foster, 2011).
leading up to their qualifying exams. The students
showed improvements in psychological symptoms
Mood beyond health
related to chronic stress, as indexed by attenuated
increases in cortisol, improved sleep quality, and Although it may be tempting to simply conclude that
improved parasympathetic activity (Nishida et  al., the microbiome influences health and that health in
2017). Recent studies have further demonstrated that turn influences affective states (e.g., “good” bacteria
male rats that consumed a diet rich in prebiotics, which make you healthy, and healthy people are happy), there
is fiber that beneficial microbes can digest, exhibited are also several compelling examples of bacteria influ-
decreases in anxiety-like behaviors and altered gene encing emotion in which improvements in physical
expression in neural circuits that may support emotion health were not the primary mediating force. For exam-
regulation (Mika et al., 2018). By contrast, chemically ple, a probiotic yogurt drink developed to reduce
Microbes and the Mind 399

constipation was administered to human subjects daily Dinan, & Cryan, 2016). Specifically, there is a consensus
for 3 weeks. Although the probiotic drink did not influ- emerging that the microbiome and brain communicate
ence the frequency of defecation (the targeted health regularly and bidirectionally along a network termed
outcome), it did influence discrete emotional outcomes. the gut–brain axis (Allen, Dinan, Clarke, & Cryan, 2017;
Consumption of the probiotic drink (compared with a Cryan & Dinan, 2012; Wiley et al., 2017). The gut–brain
placebo drink) improved the self-reported mood of axis facilitates molecular signaling between microbes
those whose mood was initially poor (Benton, Williams, in the GI tract and the central nervous system (CNS)
& Brown, 2007). In another study, healthy human sub- and peripheral nervous system (PNS). It also facilitates
jects with no symptoms of physical illness who ingested communication to and from the enteric nervous system
a probiotic supplement for 30 days reported significant (ENS), which is embedded in the lining of the GI tract
decreases in symptoms of psychological distress over and can function independently of the CNS to regulate
time. Specifically, they exhibited decreases in anxiety gut motility and composition (Carabotti, Scirocco,
(as assessed by the Hospital Anxiety and Depression Maselli, & Severi, 2015; Cryan & Dinan, 2012). One of
Scale; Zigmond & Snaith, 1983) and fewer symptoms of the most important and well-researched features of gut-
depression, anger, and hostility (as assessed by the brain communication is the ability of bacteria to both
respective subscales of the Hopkins Symptom Checklist; produce and directly respond to a variety of neuro-
Derogatis, Lipman, Rickels, Uhlenhuth, & Covi, 1974) chemicals. These neurochemical signals can travel to
compared with those taking a placebo (Messaoudi et al., the brain via several biochemical and structural path-
2011). ways along the gut–brain axis. These pathways are
Strikingly, the positive effects of bacteria on mood illustrated in Figure 1 and more extensively reviewed
are not limited to how people perceive their own emo- in the “Pathways of Communication” section. The cur-
tions but also how people perceive emotions on the rent section introduces several key bacterially produced
faces of others. To this end, researchers found that neurochemicals that have been shown to affect psycho-
when healthy women with no gastrointestinal (GI) or logical outcomes relevant to research in our field.
psychiatric symptoms consumed a nonfermented pro-
biotic drink daily for 4 weeks, they showed decreased
reactivity to negative facial expressions on other peo-
Serotonin
ple’s faces (angry and sad faces) compared with sub- It is well established that serotonin is fundamental to
jects who consumed a placebo (Tillisch et  al., 2012). physical and psychological health, but less is known
Taken together, these results suggest that bacteria do about how vitally important it is for GI health ( J. J. Chen
not influence emotions simply by ameliorating poor et al., 2001). In fact, approximately 90% of the body’s
health, as all of these human subjects were already serotonin is located along the intestinal tract, where its
healthy. Examples from animal research also support primary function is regulating gut motility (Berger,
this claim. Healthy mice administered beneficial bacte- Gray, & Roth 2009). The serotonin produced in the gut
ria (compared with healthy mice given a placebo) tend is also central to healthy nervous system development,
to be more exploratory and outgoing in their behavior and research with rodents indicates that the activation
and show fewer symptoms of helplessness and depres- of serotonin receptors in the ENS is linked to its
sion (Bravo et al., 2011; Davis et al., 2016). neurogenesis and neuroprotection (De Vadder et  al.,
How do bacteria influence emotions, then, if improved 2018). Given that many people diagnosed with serotonin-
mood is not just an outcome of improved health from deficit-based mood disorders (e.g., depression, anxiety)
the presence of beneficial bacteria and negative mood have a comorbid gut disorder (Martin-Subero, Anderson,
is not just an outcome of poor health induced by patho- Kanchanatawan, Berk, & Maes, 2016), serotonin-based
genic bacteria? Although health almost certainly medi- interventions may be effective in treating both psycho-
ates the influence of bacteria on emotion in many cases, logical and gut-related symptoms. In fact, depression
experimental studies also point to evidence that bacteria and anxiety symptoms in humans tend to increase in
can more directly modify a host’s neural functions, parallel with GI symptom severity and frequency (Pinto-
affecting a variety of psychological processes and men- Sanchez et  al., 2015), making this comorbidity even
tal states. more relevant. Current treatment methods, however,
involve administering a selective serotonin-reuptake
inhibitor that, although effective in ameliorating behav-
Neuroscience and the Gut–Brain Axis ioral symptoms, can have negative effects from long-
Evidence from the past decade suggests that the micro- term use and does not address the root cause of the
biome may have just as strong an influence over the issue (Cohen & Baldessarini, 1985). Alternatively, admin-
brain as the brain has over the microbiome (Stilling, istering beneficial bacteria that are known to increase
400 Smith, Wissel

Neurometabolite Production

Endocrine Response

Immune Activation

Vagal Communication
Inner
Intestines

GABA

SCFA Serotonin
IL-10
IgA
LPS IL-6

al Wall
Intestin

Fig. 1.  Pathways of communication along the gut–brain axis. Bacteria can secrete neu-
rotransmitters (e.g., γ-aminobutyric acid, or GABA) that induce intestinal cells to release
molecules that modulate neural signaling within the neurometabolite production pathway—
the enteric nervous system (ENS), central nervous system (CNS), and peripheral nervous
system (PNS). Bacteria can also secrete short-chain fatty acids (SCFAs) that induce neuro-
endocrine cells to convert amino acids into serotonin, which is communicated to the brain
by ENS activity (neuroendocrine response pathway). The overgrowth of pathogenic bacteria
can increase intestinal permeability, releasing harmful bacteria into the bloodstream. This
triggers an immune response and leads to the production of inflammatory immune mol-
ecules—such as interleukin- 6 (IL-6) or IL-10—and antibodies (e.g., immunoglobulin A,
or IgA) that signal to the CNS and PNS (immune activation pathway). Last, information is
routinely passed between the gut and brain along the vagus nerve. Signals from the CNS
can influence motor, sensory, and secretory functions of the intestines, and the intestinal
microbiome can send information about gut motility and composition via afferent vagal
pathways to the CNS (vagal communication pathway).

serotonin levels (Neufeld et  al., 2011; Reigstad et  al., 2003; Nemeroff, 2003). Benzodiazepines are primarily
2015) in the form of a probiotic or by following a diet used to treat anxiety disorders because they bind to
that supports bacteria critical for serotonin production GABA receptors in the brain, making GABA more effi-
may effectively influence mood as well as concomitant cacious. Antidepressants can also be used to treat GABA
GI symptoms. Current research with humans has already imbalances in people who have been diagnosed with
shown that an oral probiotic can improve the affective depression. It is known that certain gut bacteria natu-
symptoms of those suffering from a poor mood (Benton rally produce GABA (E. Barrett, Ross, O’Toole,
et al., 2007), and this may be due in part to the influence Fitzgerald, & Stanton, 2012). Furthermore, when probi-
bacterial populations exert on key neurochemicals. otics are ingested by rodents, they have been found to
alter GABA in the same regionally dependent manner
as benzodiazepines and antidepressants (Bravo et al.,
GABA 2011; Janik et al., 2016). After probiotic treatment with
γ-Aminobutyric acid (GABA) has a strong connection these GABA-producing bacteria, healthy mice demon-
to depression and anxiety disorders; low levels of strate an increase in exploratory behavior (Collins,
GABA correspond to increased depression- and anxiety- Surette, & Bercik, 2012), showing that even a healthy
related symptoms (Cryan & Kaupmann, 2005; Lydiard, population can benefit from probiotics. Likewise, the
Microbes and the Mind 401

administration of commensal bacteria that have been to be not only beneficial but also vital to brain function
shown to modulate GABA receptor mRNA expression and development.
in rodents can reduce anxiety, increase the experience
of pleasure, and modulate other affectively laden moti- Cognitive Science and Information
vational states (Bravo et al., 2011).
Processing
In addition to affecting the brain at both the molecular
Brain-derived neurotrophic factor and structural level, bacteria influence the emergent
Brain-derived neurotrophic factor (BDNF) has been properties of neurological activity. Specifically, the
implicated in an array of psychological disorders—such microbiome has been implicated in a range of cognitive
as major depressive disorder (Kielstein et al., 2015) and processes related to an individual’s ability to process,
schizophrenia (Nieto, Kukuljan, & Silva, 2013). It has integrate, and make sense of both novel and familiar
been consistently found that low levels of BDNF in information.
humans are associated with poorer psychological out-
comes and that certain medications, such as antidepres-
Memory
sants, seem to restore BDNF levels for those who have
an abnormally low supply (B. Chen, Dowlatshahi, Encoding and storing information is essential to every-
MacQueen, Wang, & Young, 2001; Shimizu et al., 2003). day functioning both for humans and animals, and bac-
Recent research has found that having an overabun- teria play a role in these cognitive processes (Benton,
dance of pathogenic bacteria results in significantly less Williams, & Brown, 2007; Magnusson et  al., 2015;
hippocampal BDNF in mice, which covaries with more Messaoudi et al., 2011). For example, in a series of stud-
anxious and depressed behaviors. Similar to the effect ies, Gareau et al. (2011) found that having a bacterial
of antidepressants, these behavioral markers of anxiety infection that leads to the proliferation of pathogenic
and depression were attenuated when a probiotic con- bacteria resulted in memory dysfunction in mice that
taining Bifidobacterium longum was administered to had been exposed to a prolonged water-avoidance
the mice (Bercik, Denou, et  al., 2011). Other studies stressor, which is a well-established model of psycho-
have also found that having a healthier microbial profile, logical stress in rodents (Saunders et al., 2002). In addi-
either by receiving a probiotic, reducing the abundance tion, germ-free mice exhibited similar stress-induced
of pathogenic bacteria, or supporting communities of memory dysfunction. This memory dysfunction was
beneficial bacteria that are already present, results in prevented when mice received a probiotic containing
more BDNF and exploratory behavior in mice (Collins an array of commensal bacteria, suggesting that probi-
et  al., 2012). These findings suggest that optimizing otics designed to normalize gut bacteria can be benefi-
one’s microbial profile could effectively increase low cial for cognitive processing. Likewise, mice with
BDNF levels. enriched diets display increased working and reference
memory (Collins et  al., 2013; Li, Dowd, Scurlock,
Acosta-Martinez, & Lyte, 2009). Recent research with
Deficits in bacteria rodents has demonstrated that probiotics can have
Studies in germ-free animals—which live in sterile envi- strong and significant effects on both long-term object
ronments and have no exposure to bacteria—have recognition and short-term memory for objects in place
shown that a lack of bacteria results in many detrimen- (O’Hagan et al., 2017).
tal neurological consequences (Al-Asmakh & Zadjali, These findings may, however, have important mod-
2015; Cebra, 1999; Collins et  al., 2012). Specifically, erators. For example, in a recent experimental study
germ-free animals lack key neurometabolites (Wikoff with rodents, the administration of probiotics improved
et  al., 2009), and their brains lack structural integrity some types of memory (spatial) but impaired others
(Luczynski et al., 2016). For instance, the blood–brain (object recognition; Beilharz, Kaakoush, Maniam, &
barrier of germ-free mice is not as tight or well pro- Morris, 2018). Likewise, antibiotics administered to mice
tected as it is in conventional mice (Braniste et  al., to cause dysbiosis—which is an unhealthy imbalance
2014). Furthermore, the microbiome supports the of their internal microbial populations—led to impair-
growth and functioning of microglia cells, which are ments in object recognition but not spatial memory
essential for mounting immune responses to protect the (Frohlich et al., 2016). These findings suggest that the
brain from pathogens or internal damage. Even the type of memory task being assessed and the cognitive
temporary depletion of a host’s microbiota leads to processes it recruits may be differentially affected by
structurally and functionally defective microglia in bacteria populations. In addition, the health of subjects’
rodents (Erny et al., 2015). Altogether, bacteria prove microbiomes before probiotic intervention as well as
402 Smith, Wissel

their routine diets may also be key variables to account more quickly than the control animals at all levels of
for. For example, Beilharz, Kaakoush, Maniam, and complexity. This performance was in part attributed to
Morris (2018) found that administering probiotics to corresponding decreases in anxiety-related behaviors.
rats with severely dysregulated microbiomes as a result Furthermore, the bacteria-fed mice continued to out-
of a high-fat diet did indeed improve some types of perform the control mice for a week after the bacteria
memory performance. By comparison, the probiotic had been withdrawn from their diets. However, this
intervention led to memory deficits in healthy rats with effect did not persist beyond that time frame, suggesting
typical microbiomes. In humans, many diets support a that the continual rather than intermittent presence of
healthy microbiome and, in general, eating more pre- healthy bacteria is necessary to induce positive cogni-
biotic fiber and fewer processed foods will confer a tive and affective outcomes (Matthews & Jenks, 2013).
health benefit. However, further research will need to These findings also must be interpreted with caution,
examine when and which bacterial strains most directly as there exists at least one study with humans wherein
influence memory-related processes and how this inter- the candidate probiotic Lactobacillus rhamnosus failed
acts with the host diet. to modulate cognitive performance in a randomized
placebo-controlled trial (Kelly et al., 2017). As discussed
above, this highlights the need for further research
Learning and attention
aimed at successfully identifying what moderators, as
Research suggests that the microbiome also influences well as which strains of bacteria, are most likely to
the ability to process, apply, and ultimately learn from affect cognitive processing in healthy human subjects.
information. For example, when mice were fed com-
mensal bacteria daily for 11 weeks, they showed sig- Social Networks and Close Relationships
nificant improvements on cognitive tasks. Note that the
improvement depended on the exact type of bacteria The relationship that humans have with their commen-
mice ingested. Those fed B. longum showed the most sal bacteria is typically mutually beneficial, and the
improvement in object recognition and object discrimi- reach of people’s microbiomes can extend into their
nation, followed by those fed Bifidobacterium breve, social networks and influence their interpersonal
whereas control animals who did not receive a bacteria behaviors.
supplement demonstrated the least improvement and
the poorest performance overall (Savignac, Tramullas, Social niches and interpersonal
Kiely, Dinan, & Cryan, 2015). A study that examined the
effects of diet on the microbiome showed similar results.
interactions
When tested 2 weeks after a controlled diet change, mice The social niche people live in affects their microbiome
fed a high-fat or high-sucrose diet that resulted in the from birth to death. Being delivered via vaginal birth
proliferation of pathogenic bacteria exhibited impaired versus cesarean section sets the stage for the forthcoming
cognitive flexibility compared with control mice that bacterial profile of the infant (Dominguez-Bello et  al.,
were fed a normal diet (Magnusson et al., 2015). 2010), and the nutrients present in a mother’s breast milk
In a recent study with human subjects, participants feed the growing bacterial population in a developing
who received a 4-week probiotic food supplement child (Hinde & German, 2012). This means that within the
experienced significant reductions in cognitive reactivity first few days after birth, social networks and social prac-
to sad moods compared with a control group that received tices are already exerting an influence on the bacteria that
a placebo. This effect was driven primarily by decreases humans will share their bodies with for the rest of their
in the rumination and aggressive thoughts linked to sad lives. In fact, all humans emit a relatively unique “micro-
moods and therefore provides some of the first evidence bial cloud” (Meadow et  al., 2015)—a collection of per-
that beneficial bacterial species can influence emotional sonal bacteria—that is shed into the surrounding air. This
states via cognitive pathways (O’Mahony et al., 2014). cloud influences the microbial composition of humans’
Likewise, human subjects who ingested a prebiotic external environment, including shared social spaces and
every day for 3 weeks exhibited less attentional vigi- the people in them. Even ethnic affiliation is correlated with
lance to negative versus positive stimuli and a subse- microbiota composition (Mason, Nagaraja, Camerlengo,
quent decrease in cortisol levels (Schmidt et al., 2015). Joshi, & Kumar, 2013; Ravel et al., 2011), presumably
In another study that explored the ways in which because of shared genes, shared diets, shared living spaces,
cognition and affect may interact to influence learning, and other shared cultural experiences.
mice were fed the ambient bacteria Mycobacterium Bacteria respond not only to whom people are sur­-
vaccae before completing increasingly complex maze round­e d by but also to the content of their social
tasks. The bacteria-fed animals completed the mazes interactions—especially social stress (Parashar &
Microbes and the Mind 403

Udayabanu, 2016). For instance, mice exposed to socially fluids produced by an interaction partner (Borg & de
aggressive cage mates exhibited substantially altered Jong, 2012). Kissing, for example, may contribute to
gut microbial communities (Bailey et  al., 2011), and mate selection by offering the opportunity to sample
mice exposed to repeated experiences of social defeat chemical cues from the saliva that indicate health and
showed disruptions in their balance of intestinal bac- compatibility (Nicholson, 1984), and smelling or tasting
teria (Galley et al., 2014). Furthermore, the depletion a partner’s sweat may provide similarly rich mating-
of beneficial bacteria in mice has been shown to sup- relevant information (Bhutta, 2007; Gildersleeve,
press normative immune responses after social stress- Haselton, Larson, & Pillsworth, 2012; Singh & Bronstad,
ors, such as being repeatedly physically attacked by 2001). Not surprisingly, bacteria are an essential ingredi-
other mice (Galley et al., 2014). This suggests that bac- ent in each of these mate-selection signals. For exam-
teria not only respond to stress but may also aid indi- ple, approximately 80 million bacteria are exchanged
viduals in the ability to recover after major social in a single mouth-to-mouth kiss, and the more fre-
disruptions. Maternal separation, for example, is an quently partners kiss, the more similar their oral and
early-life relational stressor that can result in lifelong tongue microbiota become (Kort et al., 2014). Likewise,
disturbances in stress reactivity (e.g., chronic or pro- sweat itself does not have an odor. Rather, the bacteria
longed activation of the hypothalamic-pituitary-adrenal, colonizing the skin generate the odors that serve as
or HPA, axis). After maternal separation, baby mice olfactory cues for health, similarity, and social recogni-
suffer a significant decrease in beneficial bacteria such tion (Archie & Theis, 2011; Kligman & Strauss, 1956;
as Lactobacillus species (Bailey & Coe, 1999), whereas Montiel-Castro, Augusto, Báez-Yáñez Mario, & Pacheco-
being treated with that same (and other similar) bacteria López, 2014). Additional markers of reproductive fitness
before the separation attenuates the stress response, as that bacteria directly shape include clear skin, lustrous
indexed by reduced corticosterone levels (Desbonnet hair, and body mass index (Levkovich et  al., 2013;
et al., 2010). Poutahidis et al., 2013; Poutahidis et al., 2014). This work
A microbiome abundant in beneficial bacteria spe- demonstrates that the microbiome plays an important
cies may even promote prosocial behaviors. Social role in promoting traits that are used in the mate-selection
interactions are most likely to occur when exploratory process.
behaviors are enhanced and anxious behaviors are Experimental research has demonstrated that bacte-
decreased. There are at least a dozen known strains of ria can also shape mate preferences at the actor level.
bacteria that exert anxiolytic—or anxiety-reducing— For example, in an experimental study, administering
effects on their hosts and promote exploration of the antibiotics to fruit flies eliminated their normative pref-
environment (Wissel, 2018). Consistent with these find- erence for mates that inhabited environments similar
ings, germ-free mice exhibit significant social impair- to their own and maintained similar diets (Sharon et al.,
ments, lacking the normative preference for spending 2010). In a departure from their normative behavior,
time with other mice compared with spending time the fruit flies given the antibiotics selected mates more
alone in an empty chamber. Mice with healthy micro- indiscriminately, with no preference given to similarity
biomes, on the other hand, are more likely to explore, factors. This suggests that the disruption of the micro-
interact with novel mice, and demonstrate the norma- biome can alter individuals’ mating preferences and
tive motivation to socialize with others (Desbonnet, behaviors. Likewise, sexual practices can influence the
Clarke, Shanahan, Dinan, & Cryan, 2014). microbiome. For example, non-monogamous female
lizards show a more diverse microbiome in their cloaca
than do monogamous females (White, Murielle, Massot,
Mate selection and intimate relationships
& Meylan, 2011). In humans, men who maintain monog-
Once people have established a social niche, instances amous relationships can be differentiated from those
of interpersonal bacterial exchange become frequent who have had extramarital affairs by the bacterial pro-
and periodic. Human romantic relationships may have file of their reproductive organs (or their community
an especially strong influence on the composition of state type; C. M. Liu et al., 2015).
the microbiome, as they offer virtually unparalleled Bacteria also shape mating and relationship pro-
opportunities for bacterial exchange and colonization cesses at the hormonal level. Feeding mice certain types
through multiple channels (e.g., mucus membranes, of bacteria elicits the production of oxytocin, a hor-
prolonged skin-to-skin contact). mone that has been associated with monogamy and
There is already evidence that people use informa- pair bonding, in the brain and bloodstream (Varian
tion from other people’s bodies to assess their repro- et al., 2016, 2017). Mice treated with the purified bac-
ductive fitness. To this end, sexual arousal engenders teria Lactobacillus reuteri exhibit increases in testoster-
a higher tolerance, and even a desire, for biochemical one, larger testicles, and higher sperm counts and tend
404 Smith, Wissel

to build extra muscle and maintain a competitive edge numerous bacteria isolated in the umbilical cord, sug-
in their reproductive efforts (Poutahidis et al., 2014). gesting that they were translocated from the mother to
At the genetic level, microbes can promote bacteria- the fetus during pregnancy (Matamoros, Gras-Leguen,
induced alterations in nuclear genes that code for the Le Vacon, Potel, & de La Cochetiere, 2013). More recent
production of sex pheromones in animals or that gener- attempts to replicate these findings have not always
ate molecules that act as sex attractants (Brucker & been successful, however, and some researchers have
Bordenstein, 2012). It should be noted that the type of argued that the bacterial presence in the placenta could
bacteria that are known to possess some of the most be caused by contamination of the sample rather than
robust psychoactive properties are overrepresented in translocation from mother to baby (de Goffau et  al.,
the human reproductive organs (e.g., species of Lacto- 2018). Regardless, the microbiome of a pregnant woman
bacillus; Perez-Burgos et al., 2013). This suggests that experiences several pronounced changes throughout
bacterial transfer via intimate or sexual contact may be gestation that can affect the fetus. Specifically, there is
especially likely to influence downstream neurological a decrease in proinflammatory bacteria and an increase
and psychological processes. Thus, in an effort to sup- in anti-inflammatory bacteria from the first to third tri-
port their own reproductive success, bacteria have also mester, as well as a massive proliferation of bacteria
shaped the traits and behaviors that drive the reproduc- that expertly assist in nutrient uptake (Koren et  al.,
tive success of their hosts. 2012; Mueller, Bakacs, Combellick, Grigoryan, &
Dominguez-Bello, 2015). This trajectory of microbial
Developmental Psychology and development is an example of how bacteria respond
to the physiological needs of the host and aid in the
Bacteria Across the Life Span safe delivery and development of the baby.
The human microbiota undergoes a process of develop- Psychological factors also have an important effect
ment that is coordinated with the development of the on the microbiome during pregnancy. Maternal stress
CNS, PNS, ENS, endocrine system, and immune system. in particular influences both the microbiome of the
This symbiotic relationship is established early in life mother as well as the fetus. For example, the self-
but changes dynamically to meet specific developmen- reported stress of mothers during pregnancy—as mea-
tal needs—sometimes prioritizing the needs of the hosts sured by the Pregnancy-Related Anxiety Questionnaire
and other times the needs of the bacteria. For example, (PRAQ-R; Van den Bergh, 1990) and the reported fre-
individuals often exhibit different microbial profiles quency of pregnancy-related hassles—predicted dif-
across different stages of the life span (Hopkins, Sharp, ferential gut microbiota in infants as well as their
& Macfarlane, 2002). Specifically, higher proportions of susceptibility to GI disease and allergic reactions
bacteria dedicated to enriching growth and develop- (Zijlmans, Korpela, Riksen-Walraven, de Vos, & de
ment are often found in infants and children (birth to Weerth, 2015). Maternal stress has also been positively
11 years old), and larger proportions of bacteria associ- correlated with the prevalence of bacteria associated
ated with inflammation and obesity are found in adults with respiratory symptoms, HPA reactivity (Golubeva
over 70 years old (Hollister et  al., 2015). Thus, the et al., 2015), and the disruption of amino-acid profiles
development of the microbiome can be examined from in the developing brain ( Jašarević, Howerton, et  al.,
a functional perspective. This allows us to identify not 2015). One possible mechanism of action is that emo-
only how humans’ resident bacteria change over time tional and physiological stress influence the microbial
but also how they shape physical and psychological population of the vagina, decreasing commensal and
development across the life span. increasing pathogenic bacteria, which then seed the
infant’s microbiome as it passes through the birth canal
( Jašarević, Howerton, et al., 2015).
Gestation, birth, and early infancy
A variety of behavioral factors are also known to
The gut and vaginal microbiomes undergo considerable affect the infant microbiome. For example, the mode
changes during pregnancy, resulting in a strong mater- of delivery (vaginal vs. cesarean section), the location
nal signature on the microbial profile of newborns of delivery (hospital vs. home birth), whether caregivers
(Palmer, Bik, DiGiulio, Relman, & Brown, 2007). The breastfeed or provide the infant with formula, and
human uterus was previously thought to be sterile, but whether mothers and babies receive any type of anti-
recent research suggests that bacteria may actually be biotic treatment early in life all have a lasting impact
transferred to the fetus via the bloodstream and pla- on the microbiome. Infants born vaginally tend to have
centa while still in utero ( Jiménez et al., 2008; Rautava, microbiomes dominated by Lactobacillus strains from
Collado, Salminen, & Isolauri, 2012; Satokari et  al., their mothers’ vaginas and fecal matter (Dominguez-Bello
2009). Although this work is still in its early stages, et al., 2010; Mueller, Bakacs, et al., 2015; Vaishampayan
researchers have now catalogued the presence of et al., 2010), whereas the microbiomes of infants born
Microbes and the Mind 405

via caesarian section are largely populated by bacteria et al., 2010; Iyer & Vaishnava, 2016; Mutlu et al., 2009).
found on the skin and immediate external environment— Thus, adolescence is a stage during which neurological,
such as the room in which the baby was delivered biological, social, and bacterial changes all unfold in
(Dominguez-Bello et al., 2010). Relative to their formula- concert with one another in a manner that ultimately
fed counterparts, infants that are breastfed exhibit a sets the stage for healthy or pathological development
healthy population of Bifidobacterium and Lactobacil- well into adulthood.
lus species (Fernández et al., 2013; Jiménez et al., 2012).
These bacteria can enhance the ability to uptake and
Aging populations and older adults
process nutrients (M. S. Kramer et  al., 2008; Roger,
Costabile, Holland, Hoyles, & McCartney, 2010), which By 2030, people over the age of 60 years will outnum-
may in turn lead to positive neurodevelopmental out- ber children under the age of 9 years, and by 2050,
comes (e.g., improved intelligence and cognitive abili- there will be more people over the age of 60 years than
ties; M. S. Kramer et al., 2008). Last, the administration people between the ages of 10 and 24 years (United
of antibiotics during gestation and infancy is robustly Nations, 2015). Although this area of research is in its
associated with decreased bacterial diversity—including nascent stages, physiological processes in these older
and especially the depletion of beneficial bacteria adult populations are especially likely to be connected
(Mueller, Whyatt, et al., 2015). Although bacteria colo- to the microbiome. For example, inflammation worsens
nies may repopulate eventually, massive bacterial extinc- as the body ages (Guigoz, Doré, & Schiffrin, 2008), and
tion during these critical periods of development may bacteria play an important role in regulating immune
have long-term deleterious effects on a range of neuro- responses at the systemic and molecular level. In addi-
logical and psychological outcomes. tion, gut motility decreases as individuals age, leading
to changes in nutrient uptake, extended digestion times,
and irregularity in bowel movements (Camilleri, Lee,
Adolescence and early adulthood Viramontes, Bharucha, & Tangalos, 2000). All of these
Adolescence is a transitional period characterized by changes affect the composition of the microbiome and
major neuroanatomical changes that can partly be the proportion of bacteria colonizing people’s intes-
mapped onto the developmental changes observed in tines. Although seemingly trivial, chewing strength may
bacterial populations. For example, two of the most also decline with age, which can change the integrity
critical alterations in the human cortex during late and type of food the intestines are challenged to digest
childhood/early adolescence involve axonal myelina- (Watanabe, 1998). This can lead to different and pro-
tion and synaptic pruning. Research with germ-free longed functional demands on the bacteria tasked with
animals has implicated bacteria in both of these pro- decomposing the food and extracting appropriate nutri-
cesses. Specifically, animals bred without a microbiome ents. Medication regimens as people age may include
show hypermyelination in the prefrontal cortex (Hoban the prolonged use of antibiotics, which dramatically
et al., 2016) as well as defective microglia cells (Erny influence bacteria populations. For all of these reasons,
et al., 2015), which are crucial for pruning back neurons the gut microbiota of human adults approximately 70
during development (Paolicelli et  al., 2011; Schafer years of age and older is significantly different from
et al., 2012). Of course, the disruption of neurological that of younger adults and infants (O’Toole & Claesson,
development during adolescence as a result of bacterial 2010). Specifically, researchers have observed a decline
imbalances will likely have adverse consequences for in the bacterial diversity of older human populations
concomitant cognitive developments, and several (70–99 years old), and, not surprisingly, this decline
experimental studies offer supporting evidence. For was comorbid with decreases in brain weight, brain
example, infecting healthy rodents with harmful bacte- volume, and cognitive impairments (Biagi et al., 2010,
rial strains can lead to learning deficits (Gareau et al., 2013; Claesson et al., 2012).
2011; Li et al., 2009), whereas administering healthy bac-
teria to infected mice can ameliorate symptoms of cogni-
Critical windows and sensitive periods
tive dysfunction (Bercik, 2011; Gareau, Jury, MacQueen,
Sherman, & Perdue, 2007). Microbial and neurological development unfold
As mentioned throughout this review, environmental together and thus may have overlapping windows of
and psychological stressors alter people’s microbiota, time during which they are more or less responsive to
and adolescence is a developmental period rife with change and intervention. Specifically, emotional and
these types of assaults. Changes in diet, disrupted sleep physiological stress during early life have a substantial
patterns, access to drugs and alcohol, and navigating impact on both the brain and microbiome, and disrup-
sexual relationships all influence the microbiome, and tions may be difficult to normalize if interventions are
all typically have onsets during adolescence (De Filippo applied too late in development. For example, colonizing
406 Smith, Wissel

mice with commensal bacteria shortly after birth cor- psychopathological conditions. In fact, applied clinical
rected deficits in the development of the HPA axis researchers were among the first psychological scien-
caused by restraint stress, whereas colonizing mice later tists to collaborate with microbiologists to understand
in life did not produce the same therapeutic outcomes the ways in which pathologies of the gut were comor-
(Sudo et al., 2004). Likewise, deficits in social-cognitive bid with pathologies of the brain and behavior. This
processes in mice established early in life (because of section provides a brief overview of some of the most
an absence of key bacterial populations) could not be frequently studied clinical conditions that sit at the
reversed by bacterial interventions later in life (post- intersection of psychological and microbial processes
weaning), although some behaviors (such as social (for extensive reviews, see R. T. Liu, 2017; Nowakowski
avoidance) were more amenable to treatment (Desbonnet et al., 2016).
et al., 2014).
Adolescence may also constitute a sensitive devel-
Autism spectrum disorder
opmental period. For example, bacterial profiles were
significantly altered in rats exposed to exercise during The microbiome has recently been placed at the fore-
their juvenile years, whereas exercise exposure during front of autism spectrum disorder (ASD) research, and
their adult years showed no such effect. Furthermore, although the role of the microbiome in ASD has enough
the exercise-induced changes in the microbiota of these complexity for its own review article (see Luna, Savidge,
adolescent rats were associated with an increase in lean & Williams, 2016), some general conclusions can be
body mass (Mika et al., 2015), suggesting that earlier summarized here. Germ-free mice show similar social
(but not later) exercise-related interventions may facili- deficits and repetitive behaviors as those observed in
tate the development of a microbiome that promotes humans with ASD (Desbonnet et al., 2014), and mouse
healthy weight maintenance. Likewise, colonization models of ASD have an altered microbiota (de Theije
with commensal bacteria during the adolescent period et al., 2014), suggesting that when beneficial bacteria
reversed stress hyperresponsivity in mice (i.e., reversed are missing ASD-like deficits can appear. Because ASD
the prolonged elevation of adrenocorticotropin, or is so diverse and heterogeneous in nature, it is difficult
ACTH, after a stressful event). However, bacteria colo- for specific microbiota-related findings to generalize
nization introduced in later adulthood seemed to be across the entire spectrum (as addressed in Song, Liu,
ineffective at ameliorating these stress-induced eleva- & Finegold, 2004). Despite this limitation, some findings
tions in ACTH (Neufeld, Luczynski, Oriach, Dinan, & do have more general implications for ASD. For exam-
Cryan, 2016). These findings suggest that early adoles- ple, rates of GI pathologies and symptoms are dispro-
cence is another critical window of development during portionately high in people with ASD compared with
which gut bacteria can shape the stress response. neurotypical populations (de Theije et al., 2011). Pro-
It is important to note that despite having sensitive biotics, prebiotics, and antibiotics have all shown
periods of development, the microbiome may be more behavioral benefits for people with ASD (Critchfield,
flexible across the life span and more amenable to van Hemert, Ash, Mulder, & Ashwood, 2011; Grimaldi
change than other physiological systems. Researchers et  al., 2018), but whether this is due to the bacteria
are currently examining a variety of restorative inter- themselves or the natural cycle of ASD is still unclear.
ventions geared toward transforming the microbiome All of these findings do, however, suggest that a base-
after it has been altered by early-life stress and assault line dysbiotic state may exist in those with ASD.
(Cowan, Callaghan, & Richardson, 2016; Mueller,
Bakacs, et al., 2015; Salazar et al., 2014). Several pre-
Clinical depression and anxiety
clinical bacteria-based treatments are being investigated
for their therapeutic and medical properties (Dinan, There has been an abundance of research on the ways
Stanton, & Cryan, 2013). Indeed, the microbiome may in which the microbiome shapes depression and anxi-
offer a pathway to correct neurodevelopmental patholo- ety (for recent and thorough reviews, see Foster &
gies by acting on elements that influence function and Neufeld, 2013; Huang, Wang, & Hu, 2016). However,
process (e.g., neurochemicals excreted by bacteria) most of this work has been conducted with rodents
rather than relatively unalterable morphological struc- rather than humans. Nonetheless, some recent research
tures (e.g., brain tissue). in humans has found that depression and anxiety symp-
toms increase stepwise as GI symptoms and severity
increase (Pinto-Sanchez et al., 2015). GI illness is also
Clinical Psychology and Psychopathology highly comorbid with clinical depression and anxiety
Studies in humans and animals have repeatedly shown (Maes, Kubera, Leunis, & Berk, 2012; Maes et al., 2013).
that the microbiome is implicated in a variety of In addition, probiotics have been found to improve
Microbes and the Mind 407

both GI and clinical depression and anxiety symptoms Cryan, & Dinan, 2018; Dinan et  al., 2013; Misra &
for those with disorders such as irritable bowel syn- Mohanty, 2017; Wall et al., 2014). Although there is still
drome (Pinto-Sanchez et  al., 2017; Wallace & Milev, a need for more randomized, controlled, clinical trials
2017). Although under some circumstances probiotics investigating the impact of psychobiotics on humans
can have antidepressant or anxiolytic effects robust (Anglin, Surette, Moayyedi, & Bercik, 2015), their thera-
enough to relieve clinically classified symptoms in peutic potential for a broad range of mental-health
humans, how this happens and which combination of experiences is a promising future direction for pharma-
probiotics may be most optimal for treatment requires cologically driven treatments.
further investigation.

Pathways of Communication: Connecting


Posttraumatic stress disorder
the Mind and Gut via the Vagus Nerve
The microbiome is implicated in both the normative
Microbiologists are beginning to home in on which
and hyperactivated stress response in humans (R. T.
biological structures may facilitate communication
Liu, 2017) and animals (e.g., van de Wouw et al., 2018).
between the gut and the brain. A promising and pri-
Given that severe and prolonged stress is a central
mary contender is the vagus nerve—a component of
feature of posttraumatic stress disorder (PTSD),
the autonomic nervous system that has been of special
researchers have begun to explore the ways in which
interest to psychological scientists investigating social
the microbiome is related to the prevalence and time
emotions (Muhtadie, Koslov, Akinola, & Mendes, 2015;
course of this disorder (Malan-Muller et al., 2018). For
Quintana, Guastella, Outhred, Hickie, & Kemp, 2012;
example, in a recent exploratory study researchers
Shahrestani, Stewart, Quintana, Hickie, & Guastella,
reported that individuals with a PTSD diagnosis had
2015), subjective well-being (Geisler, Vennewald,
lower proportions of three distinct phyla of bacteria
Kubiak, & Weber, 2010), mindfulness (Kok et al., 2013;
(Actinobacteria, Lentisphaerae, and Verrucomicrobia)
Svendsen et al., 2016), health (Thayer, Åhs, Fredrikson,
compared with controls who had also been exposed to
Sollers, & Wager, 2012), and psychopathology (Austin,
a traumatic event but did not meet the clinical threshold
Riniolo, & Porges, 2007; Patriquin, Scarpa, Friedman, &
for a PTSD diagnosis. In addition, increased scores on
Porges, 2013). Indeed, there may be no other discipline
the Clinician-Administered PTSD Scale (CAPS-5;
outside the biological sciences that has examined the
Weathers et al., 2013) were associated with decreases
influence of the vagus nerve on cognitive, social, and
in the three bacterial taxa (Hemmings et al., 2017).
emotional outcomes as widely as psychological science.
This suggests, therefore, that psychological scientists
Developing future clinical treatments may be uniquely positioned to help microbiologists
answer questions that sit at the intersection of psycho-
In addition to the topics reviewed above, clinical
logical, microbial, and vagal processes.
research has also been conducted on how the micro-
biome may be associated with many other challenges
related to physical and mental health, including chronic Why are microbiologists interested in
fatigue syndrome ( Jackson, Butt, Ball, Lewis, & Bruck,
2015; Rao et  al., 2009), anorexia (Armougom, Henry,
the vagus nerve?
Vialettes, Raccah, & Raoult, 2009), alcoholism (Engen, The vagus nerve plays a central role in conveying infor-
Green, Voigt, Forsyth, & Keshavarzian, 2015; Mutlu mation about the state of the GI tract, as well as other
et  al., 2009; Yan & Schnabl, 2012), schizophrenia organs, to the CNS, with approximately 20% of its fibers
(Severance, Yolken, & Eaton, 2016), and manic depres- dedicated to GI–CNS communication (Forsythe,
sion (Dickerson, Severance, & Yolken, 2017; Kanji et al., Bienenstock, & Kunze, 2014; Stakenborg, Di Giovangiulio,
2018). It is clear from this wide range of examples that Boeckxstaens, & Matteoli, 2013). Note that certain bac-
factors that affect the microbiome are relevant to physi- teria that have colonized the gut may only be capable
cal and mental health. Exactly which mechanisms are of exerting effects on mental and neurological pro-
at play, how these mechanisms operate, and what route cesses if the vagus nerve is functionally intact. That is,
may be most optimal for treatment is still being debated, the vagus nerve may constitute a direct line of com-
but it seems that the administration of probiotics has munication between bacteria and the brain. Microbiolo-
the strongest impact on behavioral change. Live bacte- gists have reached this conclusion after conducting
ria that, when administered in a clinical or medical multiple studies demonstrating that surgically severing
context, have a significant beneficial effect on the host the vagus nerve eliminates, or in some cases reverses,
have been termed psychobiotics (Bambury, Sandhu, both the positive psychological effects of commensal
408 Smith, Wissel

bacteria and the negative psychological effects of appropriate and fruitful time to begin considering how
pathogenic bacteria. For example, severing the vagus the bacteria that people share their bodies with interact
nerve of mice eliminated the antianxiety effects of at least with the vagus nerve and, in turn, influence outcomes
two species of commensal bacteria—L. rhamnosus (Bravo of great interest to psychological science.
et al., 2011) and B. longum (Bercik, Park, et al., 2011)—as
well as the antidepressant effects of L. rhamnosus (Bravo
Other pathways of bacteria-brain
et al., 2011). Likewise, when mice were colonized with
pathogenic bacteria, severing the vagus nerve pre- communication
vented maladaptive behavior from emerging, whereas Researchers are also beginning to identify several addi-
administering pathogenic bacteria to mice with intact tional mechanisms by which the microbiome influences
vagus nerves led to increased anxiety-like behaviors neurological and psychological outcomes (see Fig. 1;
(Bercik, Park, et  al., 2011). Taken together, there is for a detailed and accessible review, see Wiley et al.,
compelling preliminary evidence that one process by 2017). For example, bacterial communities can interface
which bacteria influence psychological outcomes with the CNS by excreting metabolites with neuromodu-
depends, at least in part, on vagal integrity. latory properties and release hormone-signaling pep-
tides in the intestines that act on both sympathetic and
Vagal influence on psychological parasympathetic pathways (Schele et al., 2013; Wren &
Bloom, 2007). Different species of bacteria are also
outcomes capable of interacting with the immune system at the
Over the past 20 years, psychological scientists have molecular level, resulting in changes to the circulation
implicated the vagus nerve in a range of psychological of pro- and anti-inflammatory cytokines that influence
outcomes that are related to those currently being CNS functioning (Dantzer, 2009; Dinan & Cryan, 2017;
examined by microbiologists. For example, in human Petra et al., 2015). Furthermore, dysbiosis or an over-
subjects, higher heart-rate variability (a measure of growth of pathogenic bacteria can lead to increased
healthy vagal functioning) is associated with lower lev- permeability of both the intestinal lining (e.g., “leaky
els of trait anxiety (Svendsen et al., 2016) as well as a gut”; Hollander, 1999) and the blood-brain barrier
more habitually positive mood (e.g., cheerfulness, (Logsdon, Erickson, Rhea, Salameh, & Banks, 2018),
calmness), higher life satisfaction (Geisler et al., 2010), accelerating the rate and proportion of microbial infil-
and more competent social communication (Quintana tration (de Theije et  al., 2011). As the science of the
et  al., 2012; Shahrestani et  al., 2015). People with gut–brain axis advances, microbiologists, neuroscien-
greater vagal flexibility (another measure of adaptive tists, immunologists, and medical professionals will
vagus nerve functioning) exhibit more accurate percep- continue to uncover more detail concerning these path-
tions of social-emotional cues and are more sensitive ways of communication—and psychological scientists
to their social context (Muhtadie et  al., 2015). In a motivated to deeply understand the mind-body con-
longitudinal intervention-based study designed to nection will undoubtedly have important contributions
improve psychological well-being, individuals who had to make.
higher baseline vagal tone (an index of both vagal and
cardiovascular health) showed significantly larger
Practical Applications and Future
increases in positive emotions compared with individuals
who had low vagal tone (Kok et al., 2013). Furthermore, Directions
researchers have found that vagus nerve stimulation can There is meaningful overlap in the topics psychological
reduce symptoms of depression and anxiety and point scientists and microbiologists have been studying, but
to the gut–brain axis as an important mechanism of research exploring these topics has largely taken place
action (Kong, Fang, Park, Li, & Rong, 2018). in parallel rather than as interdisciplinary collabora-
Psychological scientists have thus repeatedly dem- tions. There are, however, a few notable exceptions that
onstrated that there is a link between the vagus nerve illustrate the important role psychological scientists can
and emotion, cognition, and behavior, whereas micro- play in advancing work on how bacteria shape mental
biologists have demonstrated that the vagus nerve states. For example, Callaghan and colleagues have
mediates the relationship between bacterial communi- integrated microbiology into their groundbreaking
ties and these same outcomes. Psychological scientists work on intergenerational stress in rodents, demonstrat-
have often situated their findings in the context of ing that the toxic effects of stress brought on by early-
broader physical health (e.g., HPA reactivity), and given life adversity (such as maternal or paternal separation)
the overlap between psychological and microbial out- can be ameliorated by probiotic interventions (Callaghan,
comes outlined above, it may be a particularly Cowan, and Richardson, 2016; Callaghan, 2017). This is
Microbes and the Mind 409

in line with recent research by microbiologists demon- overrepresented in both human and rodent males, and
strating that microbiota can both cause or reduce stress both are naturally inclined toward social interaction
depending on the type and ratio of bacteria present in and stability; Desbonnet et  al., 2014). Psychological
people’s bodies and that it is a key regulator of the scientists now have the opportunity to test generaliz-
stress response (i.e., HPA reactivity, the release of glu- ability as well as explore the subjective mental states
cocorticoids; self-reported or subjectively experienced that are associated with physiological and behavioral
stress; Wiley et al., 2017). In fact, microbiologists have outcomes. Take, for example, a common behavioral
already undertaken quite a bit of work demonstrating paradigm used with rodent models: the forced swim
that bacteria influence the experience of stress in a test (FST). The FST involves placing a rodent in a con-
variety of contexts relevant to psychological scientists tainer of water with smooth surfaces so that it must
such as academic pressure (Kato-Kataoka et al., 2016; either actively swim or passively float. Depending on
Nishida et  al., 2017) and familial disruptions such as the research hypotheses, the outcome of interest may
maternal separation ( Jašarević, Rodgers, & Bale, 2015; be time spent swimming (a proxy for perseverance) or
Waworuntu et  al., 2017). Yet collaborations between time spent immobile (a proxy for depression). Creating
microbiologists and psychological scientists on this appropriate human analogs for such behavioral mea-
topic are rare, despite the fact that they would almost sures and augmenting them with self-reports of intero-
certainly enrich our understanding of the microbiome ceptive, emotional, and cognitive states may provide
and stress in ways neither field could accomplish alone. useful insight into how bacteria influence conscious,
Another area of research in which psychological sci- psychological perceptions rather than simply modulate
entists may fruitfully integrate microbial measures is innate behavioral action plans.
work on Gene × Environment interactions. Recent Psychological scientists are well positioned to har-
reports confirm that most genes in the human body do vest a variety of low-hanging fruit simply by investigat-
not belong to the human genome but rather to the col- ing how bacteria influence the areas of research in
lective bacterial genome (Qin et al., 2010). In addition, which they are the experts, and now is the time to start.
human and microbial genes interact regularly and recip- A rapidly increasing number of programs are already
rocally to influence each other’s expression and, there- collecting microbiome-related information from a stag-
fore, function (Bordenstein & Theis, 2015; Fellows gering number of participants thanks to large-scale ini-
et al., 2018; Theis et al., 2016). This has important and tiatives at both the national and international level. For
possibly dramatic implications for the ways in which example, the National Institutes of Health, National
psychological scientists study Gene × Environment Science Foundation, National Aeronautics and Space
interactions. Consider, for example, the work of Tucker- Administration, Department of Energy, and Department
Drob and colleagues reporting that outcomes such as of Agriculture recently announced a generously funded
cognitive ability, intelligence, and academic achieve- collaboration called the National Microbiome Initiative.
ment are shaped by interactions between genetic fac- This initiative aims to expand interdisciplinary research
tors and socioeconomic status (Tucker-Drob & Bates, on the microbiome and includes the support of more
2016; Tucker-Drob & Harden, 2012; Tucker-Drob, than 100 external institutions across the private sector
Rhemtulla, Harden, Turkheimer, & Fask, 2011). We now and academia (White House Office of Science and
know, of course, that the microbiome is related to each Technology Policy, 2016). In addition, dozens of citizen-
of these variables, but perhaps most important is the scientist projects such as uBiome and Your Private
degree to which socioeconomic status (the environ- Biome have transformed the swelling public interest in
mental factor) influences the microbiome through path- the microbiome into an interactive scientific effort in
ways such as diet, sanitation opportunities (e.g., clean which thousands of everyday people share their micro-
water, clean air), and access to green space. Researchers biome data with scientists, and scientists in turn have
could thus predict that a meaningful source of vari- access to highly powered, incredibly diverse data sets
ability in these types of Gene × Environment interac- for analyses and exploration. Although breaking into
tions is accounted for not only by how the environment the microbial world may be intimidating for the nonmi-
affects the human genome but also by how it affects crobial scientist, the relationship between microbes and
each individual’s microbial genome. the mind cannot fully be understood if the perspectives
One limitation of the extant work exploring and expertise of psychological scientists remain absent.
microbe–host interactions is that it has predominantly
been conducted with nonhuman models. This can make Action Editor
it difficult to generalize findings across species, despite June Gruber served as action editor and interim editor-in-chief
relevant similarities (e.g., pathological symptoms are for this article.
410 Smith, Wissel

Author Contributions Bambury, A., Sandhu, K., Cryan, J. F., & Dinan, T. G. (2018).
Finding the needle in the haystack: Systematic identifica-
E. F. Wissel and L. K. Smith are co-first authors and con-
tion of psychobiotics. British Journal of Pharmacology,
tributed equally to this work. Both authors approved the 175, 4430–4438. doi:10.1111/bph.14127
final manuscript for submission. Barrett, E., Ross, R. P., O’Toole, P. W., Fitzgerald, G. F.,
& Stanton, C. (2012). Aminobutyric acid production by
ORCID iD culturable bacteria from the human intestine. Journal of
Emily F. Wissel https://orcid.org/0000-0003-2275-8456 Applied Microbiology, 113, 411–417. doi:10.1111/j.1365-
2672.2012.05344.x
Barrett, L. F., & Bliss-Moreau, E. (2009). Affect as a psy-
Acknowledgments
chological primitive. In M. P. Zanna (Ed.), Advances in
We thank Timothy Loving for his feedback on this manuscript ex­per­imental social psychology (Vol. 41, pp. 167–218).
and for always being supportive of our research ambitions. San Diego, CA: Academic Press.
Beilharz, J. E., Kaakoush, N. O., Maniam, J., & Morris, M. J.
Declaration of Conflicting Interests (2018). Cafeteria diet and probiotic therapy: Cross talk
The author(s) declared that there were no conflicts of interest among memory, neuroplasticity, serotonin receptors and
with respect to the authorship or the publication of this gut microbiota in the rat. Molecular Psychiatry, 23, 351–
361. doi:10.1038/mp.2017.38
article.
Benton, D., Williams, C., & Brown, A. (2007). Impact of con-
suming a milk drink containing a probiotic on mood
References and cognition. European Journal of Clinical Nutrition,
Al-Asmakh, M., & Zadjali, F. (2015). Use of germ-free ani- 61, 355–361. doi:10.1038/sj.ejcn.1602546
mal models in microbiota-related research. Journal of Bercik, P. (2011). The microbiota-gut-brain axis: Learning
Microbiology and Biotechnology, 25, 1583–1588. doi:10 from intestinal bacteria? Gut, 60, 288–289.
.4014/jmb.1501.01039 Bercik, P., Denou, E., Collins, J., Jackson, W., Lu, J., Jury, J.,
Algoe, S. B. (2012). Find, remind, and bind: The functions of . . . Verdu, E. F. (2011). The intestinal microbiota affect
gratitude in everyday relationships. Social and Personality central levels of brain-derived neurotropic factor and
Psychology Compass, 6, 455–469. behavior in mice. Gastroenterology, 141, 599–609. doi:10
Allen, A. P., Dinan, T. G., Clarke, G., & Cryan, J. F. (2017). .1053/j.gastro.2011.04.052
A psychology of the human brain–gut–microbiome axis. Bercik, P., Park, A. J., Sinclair, D., Khoshdel, A., Lu, J.,
Social & Personality Psychology Compass, 11(4), Article Huang, X., . . . Verdu, E. F. (2011). The anxiolytic effect
e12309. doi:10.1111/spc3.12309 of Bifidobacterium longum NCC3001 involves vagal path-
Anderson, S. C., Cryan, J. F., & Dinan, T. (2017). The psy- ways for gut-brain communication. Neurogastroenterology
chobiotic revolution: Mood, food, and the new science and Motility, 23, 1132–1139. doi:10.1111/j.1365-2982
of the gut-brain connection. Washington, DC: National .2011.01796.x
Geographic Society. Berger, M., Gray, J. A., & Roth, B. L. (2009). The expanded
Anglin, R., Surette, M., Moayyedi, P., & Bercik, P. (2015). biology of serotonin. Annual Review of Medicine, 60,
Lost in translation: The gut microbiota in psychiatric 355–366.
illness. Canadian Journal of Psychiatry, 60, 460–463. Bhutta, M. F. (2007). Sex and the nose: Human pheromonal
doi:10.1177/070674371506001007 responses. Journal of the Royal Society of Medicine, 100,
Archie, E. A., & Theis, K. R. (2011). Animal behaviour meets 268–278. doi:10.1177/014107680710000612
microbial ecology. Animal Behaviour, 82, 425–436. Biagi, E., Candela, M., Turroni, S., Garagnani, P., Franceschi, C.,
doi:10.1016/j.anbehav.2011.05.029 & Brigidi, P. (2013). Ageing and gut microbes: Perspectives
Armougom, F., Henry, M., Vialettes, B., Raccah, D., & Raoult, D. for health maintenance and longevity. Pharmacological
(2009). Monitoring bacterial community of human gut Research, 69, 11–20. doi:10.1016/j.phrs.2012.10.005
microbiota reveals an increase in Lactobacillus in obese Biagi, E., Nylund, L., Candela, M., Ostan, R., Bucci, L., Pini, E.,
patients and Methanogens in anorexic patients. PLOS ONE, . . . Brigidi, P. (2010). Through ageing, and beyond: Gut
4(9), Article e7125. doi:10.1371/journal.pone.0007125 microbiota and inflammatory status in seniors and cen-
Austin, M. A., Riniolo, T. C., & Porges, S. W. (2007). Borderline tenarians. PLOS ONE, 5(5), Article e10667. doi:10.1371/
personality disorder and emotion regulation: Insights journal.pone.0010667
from the polyvagal theory. Brain and Cognition, 65, Blascovich, J., & Mendes, W. B. (2010). Social psychophysiol-
69–76. doi:10.1016/j.bandc.2006.05.007 ogy and embodiment. In S. T. Fiske, D. T. Gilbert, & G.
Bailey, M. T., & Coe, C. L. (1999). Maternal separation disrupts Lindzey (Eds.), Handbook of social psychology, Vol. 1 (5th
the integrity of the intestinal microflora in infant rhesus ed., pp. 194–227). Hoboken, NJ: John Wiley & Sons.
monkeys. Developmental Psychobiology, 35, 146–155. Bordenstein, S. R., & Theis, K. R. (2015). Host biology in
Bailey, M. T., Dowd, S. E., Galley, J. D., Hufnagle, A. R., Allen, light of the microbiome: Ten principles of holobionts
R. G., & Lyte, M. (2011). Exposure to a social stressor alters and hologenomes. PLOS Biology, 13(8), Article e1002226.
the structure of the intestinal microbiota: Implications for doi:10.1371/journal.pbio.1002226
stressor-induced immunomodulation. Brain, Behavior, Borg, C., & de Jong, P. J. (2012). Feelings of disgust and
and Immunity, 25, 397–407. doi:10.1016/j.bbi.2010.10.023 disgust-induced avoidance weaken following induced
Microbes and the Mind 411

sexual arousal in women. PLOS ONE, 7(9), Article e44111. microbiota: Experimental evidence and clinical impli-
doi:10.1371/journal.pone.0044111 cations. Current Opinion in Microbiology, 16, 240–245.
Braniste, V., Al-Asmakh, M., Kowal, C., Anuar, F., Abbaspour, A., doi:10.1016/j.mib.2013.06.004
Tóth, M., . . . Pettersson, S. (2014). The gut microbi- Collins, S. M., Surette, M., & Bercik, P. (2012). The inter-
ota influences blood-brain barrier permeability in mice. play between the intestinal microbiota and the brain.
Science Translational Medicine, 6(263), Article 263ra158. Nature Reviews Microbiology, 10, 735–742. doi:10.1038/
doi:10.1126/scitranslmed.3009759 nrmicro2876
Bravo, J. A., Forsythe, P., Chew, M. V., Escaravage, E., Savignac, H., Cowan, C. S. M., Callaghan, B. L., & Richardson, R. (2016). The
Dinan, T. G., . . . Cryan, J. F. (2011). Ingestion of effects of a probiotic formulation (Lactobacillus rhamnosus
Lactobacillus strain regulates emotional behavior and cen- and L. helveticus) on developmental trajectories of emotional
tral GABA receptor expression in a mouse via the vagus learning in stressed infant rats. Translational Psychiatry,
nerve. Proceedings of the National Academy of Sciences, 6(5), Article e823. doi:10.1038/tp.2016.94
USA, 108, 16050–16055. doi:10.1073/pnas.1102999108 Critchfield, J. W., van Hemert, S., Ash, M., Mulder, L., &
Brucker, R. M., & Bordenstein, S. R. (2012). Speciation by Ashwood, P. (2011). The potential role of probiotics in
symbiosis. Trends in Ecology & Evolution, 27, 443–451. the management of childhood autism spectrum disorders.
doi:10.1016/j.tree.2012.03.011 Gastroenterology Research and Practice, 2011(161358),
Callaghan, B. L. (2017). Generational patterns of stress: Help Article 161358. doi:10.1155/2011/161358
from our microbes? Current Directions in Psychological Cryan, J. F., & Dinan, T. G. (2012). Mind-altering microor-
Science, 26, 323–329. doi:10.1177/0963721417697737 ganisms: The impact of the gut microbiota on brain and
Callaghan, B. L., Cowan, C. S., & Richardson, R. (2016). behaviour. Nature Reviews Neuroscience, 13, 701–712.
Treating generational stress: The effect of paternal stress doi:10.1038/nrn3346
on development of memory and extinction in offspring Cryan, J. F., & Kaupmann, K. (2005). Don’t worry ‘B’ happy!:
is reversed by probiotic treatment. Psychological Science, A role for GABA B receptors in anxiety and depres-
27, 1171–1180. doi:10.1177/0956797616653103 sion. Trends in Pharmacological Sciences, 26, 36–43.
Camilleri, M., Lee, J. S., Viramontes, B., Bharucha, A. E., doi:10.1016/j.tips.2004.11.004
& Tangalos, E. G. (2000). Insights into the pathophysi- Cryan, J. F., & O’Mahony, S. M. (2011). The microbiome-gut-
ology and mechanisms of constipation, irritable bowel brain axis: From bowel to behavior. Neurogastroenterology
syndrome, and diverticulosis in older people. Journal of and Motility, 23, 187–192. doi:10.1111/j.1365-2982.2010
the American Geriatrics Society, 48, 1142–1150. doi:10 .01664.x
.1111/j.1532-5415.2000.tb04793.x Damasio, A. (2003). Feelings of emotion and the self. Annals
Carabotti, M., Scirocco, A., Maselli, M. A., & Severi, C. (2015). of the New York Academy of Sciences, 1001, 253–261. doi:
The gut-brain axis: Interactions between enteric micro- 10.1196/annals.1279.014
biota, central and enteric nervous systems. Annals of Dantzer, R. (2009). Cytokine, sickness behavior, and depres-
Gastroenterology, 28, 203–209. sion. Immunology and Allergy Clinics of North America,
Cebra, J. J. (1999). Influences of microbiota on intestinal 29, 247–264. doi:10.1016/j.iac.2009.02.002
immune system development. American Journal of Davis, D. J., Doerr, H. M., Grzelak, A. K., Busi, S. B., Jasarevic, E.,
Clinical Nutrition, 69, 1046s–1051s. doi:10.1093/ajcn/ Ericsson, A. C., & Bryda, E. C. (2016). Lactobacillus
69.5.1046s plantarum attenuates anxiety-related behavior and pro-
Chen, B., Dowlatshahi, D., MacQueen, G. M., Wang, J., & tects against stress-induced dysbiosis in adult zebrafish.
Young, L. T. (2001). Increased hippocampal BDNF immu- Scientific Reports, 6, 33726. doi:10.1038/srep33726
noreactivity in subjects treated with antidepressant medi- de Filippo, C., Cavalieri, D., Di Paola, M., Ramazzotti, M.,
cation. Biological Psychiatry, 50, 260–265. doi:10.1016/ Poullet, J. B., Massart, S., . . . Lionetti, P. (2010). Impact
S0006-3223(01)01083-6 of diet in shaping gut microbiota revealed by a com-
Chen, J. J., Li, Z., Pan, H., Murphy, D. L., Tamir, H., Koepsell, H., parative study in children from Europe and rural Africa.
& Gershon, M. D. (2001). Maintenance of serotonin in the Proceedings of the National Academy of Sciences, USA,
intestinal mucosa and ganglia of mice that lack the high- 107, 14691–14696. doi:10.1073/pnas.1005963107
affinity serotonin transporter: Abnormal intestinal motility de Goffau, M. C., Lager, S., Salter, S. J., Wagner, J.,
and the expression of cation transporters. The Journal of Kronbichler, A., Charnock-Jones, D. S., . . . Parkhill, J.
Neuroscience, 21, 6348–6361. (2018). Recognizing the reagent microbiome. Nature
Claesson, M. J., Jeffery, I. B., Conde, S., Power, S. E., O’Connor, Microbiology, 3, 851–853. doi:10.1038/s41564-018-0202-y
E. M., Cusack, S., . . . O’Toole, P. W. (2012). Gut micro- Derogatis, L. R., Lipman, R. S., Rickels, K., Uhlenhuth, E. H.,
biota composition correlates with diet and health in the & Covi, L. (1974). The Hopkins Symptom Checklist (HSCL):
elderly. Nature, 488, 178–184. doi:10.1038/nature11319 A self-report symptom inventory. Behavioral Science,
Cohen, B. M., & Baldessarini, R. J. (1985). Tolerance to thera- 19(1), 1–15.
peutic effects of antidepressants. American Journal of Desbonnet, L., Clarke, G., Shanahan, F., Dinan, T. G., &
Psychiatry, 142, 489–490. doi:10.1176/ajp.142.4.489 Cryan, J. F. (2014). Microbiota is essential for social devel-
Collins, S. M., Kassam, Z., & Bercik, P. (2013). The adop- opment in the mouse. Molecular Psychiatry, 19, 146–148.
tive transfer of behavioral phenotype via the intestinal doi:10.1038/mp.2013.65
412 Smith, Wissel

Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryan, J. F., Advances in Experimental Medicine and Biology, 817,
& Dinan, T. G. (2010). Effects of the probiotic Bifidobacterium 115–133. doi:10.1007/978-1-4939-0897-4_5
infantis in the maternal separation model of depression. Forsythe, P., Sudo, N., Dinan, T., Taylor, V. H., & Bienenstock, J.
Neuroscience, 170, 1179–1188. doi:10.1016/j.neurosci (2010). Mood and gut feelings. Brain, Behavior, and
ence.2010.08.005 Immunity, 24, 9–16. doi:10.1016/j.bbi.2009.05.058
de Theije, C. G. M., Wu, J., da Silva, S. L., Kamphuis, P. J., Foster, J. A., & Neufeld, K. A. M. (2013). Gut-brain axis:
Garssen, J., Korte, S. M., & Kraneveld, A. D. (2011). How the microbiome influences anxiety and depression.
Pathways underlying the gut-to-brain connection in autism Trends in Neurosciences, 36, 305–312. doi:10.1016/j.tins
spectrum disorders as future targets for disease manage- .2013.01.005
ment. European Journal of Pharmacology, 668(Suppl. 1), Frohlich, E. E., Farzi, A., Mayerhofer, R., Reichmann, F., Jacan,
S70–S80. doi:10.1016/j.ejphar.2011.07.013 A., Wagner, B., . . . Holzer, P. (2016). Cognitive impair-
de Theije, C. G. M., Wopereis, H., Ramadan, M., van ment by antibiotic-induced gut dysbiosis: Analysis of gut
Eijndthoven, T., Lambert, J., Knol, J., . . . Oozeer, R. microbiota-brain communication. Brain, Behavior, and
(2014). Altered gut microbiota and activity in a murine Immunity, 56, 140–155. doi:10.1016/j.bbi.2016.02.020
model of autism spectrum disorders. Brain, Behavior, Galley, J. D., Nelson, M. C., Yu, Z., Dowd, S. E., Walter, J.,
and Immunity, 37, 197–206. doi:10.1016/j.bbi.2013.12.005 Kumar, P. S., . . . Bailey, M. T. (2014). Exposure to a
De Vadder, F., Grasset, E., Holm, L. M., Karsenty, G., social stressor disrupts the community structure of the
Macpherson, A. J., Olofsson, L. E., & Bäckhed, F. colonic mucosa-associated microbiota. BMC Microbiology,
(2018). Gut microbiota regulates maturation of the adult 14, 189. doi:10.1186/1471-2180-14-189
enteric nervous system via enteric serotonin networks. Gareau, M. G., Jury, J., MacQueen, G., Sherman, P. M., &
Proceedings of the National Academy of Sciences, USA, Perdue, M. H. (2007). Probiotic treatment of rat pups
115, 6458–6463. doi:10.1073/pnas.1720017115 normalises corticosterone release and ameliorates colonic
Dickerson, F., Severance, E., & Yolken, R. (2017). The micro- dysfunction induced by maternal separation. Gut, 56,
biome, immunity, and schizophrenia and bipolar disorder. 1522–1528. doi:10.1136/gut.2006.117176
Brain, Behavior, and Immunity, 62, 46–52. doi:10.1016/j Gareau, M. G., Wine, E., Rodrigues, D. M., Cho, J. H., Whary,
.bbi.2016.12.010 M. T., Philpott, D. J., . . . Sherman, P. M. (2011). Bacterial
Dinan, T. G., & Cryan, J. F. (2017). Gut instincts: Microbiota infection causes stress-induced memory dysfunction in
as a key regulator of brain development, ageing and neu- mice. Gut, 60, 307–317. doi:10.1136/gut.2009.202515
rodegeneration. The Journal of Physiology, 595, 489–503. Geisler, F. C. M., Vennewald, N., Kubiak, T., & Weber, H.
doi:10.1113/JP273106 (2010). The impact of heart rate variability on subjective
Dinan, T. G., Stanton, C., & Cryan, J. F. (2013). Psychobiotics: well-being is mediated by emotion regulation. Personality
A novel class of psychotropic. Biological Psychiatry, 74, and Individual Differences, 49, 723–728. doi:10.1016/j
720–726. doi:10.1016/j.biopsych.2013.05.001 .paid.2010.06.015
Dominguez-Bello, M., Costello, E. K., Contreras, M., Magris, M., Gildersleeve, K. A., Haselton, M. G., Larson, C. M., &
Hidalgo, G., Fierer, N., & Knight, R. (2010). Delivery Pillsworth, E. G. (2012). Body odor attractiveness as a
mode shapes the acquisition and structure of the initial cue of impending ovulation in women: Evidence from a
microbiota across multiple body habitats in newborns. study using hormone-confirmed ovulation. Hormones and
Proceedings of the National Academy of Sciences, USA, Behavior, 61, 157–166. doi:10.1016/j.yhbeh.2011.11.005
107, 11971–11975. doi:10.1073/pnas.1002601107 Golubeva, A. V., Crampton, S., Desbonnet, L., Edge, D.,
Engen, P. A., Green, S. J., Voigt, R. M., Forsyth, C. B., & O’Sullivan, O., Lomasney, K. W., . . . Cryan, J. F. (2015).
Keshavarzian, A. (2015). The gastrointestinal microbiome: Prenatal stress-induced alterations in major physiologi-
Alcohol effects on the composition of intestinal micro- cal systems correlate with gut microbiota composition
biota. Alcohol Research, 37, 223–236. in adulthood. Psychoneuroendocrinology, 60, 58–74.
Erny, D., Angelis, H. D., Jaitin, D., Wieghofer, P., Staszewski, O., doi:10.1016/j.psyneuen.2015.06.002
David, E., . . . Prinz, M. (2015). Host microbiota constantly Grimaldi, R., Gibson, G. R., Vulevic, J., Giallourou, N., Castro-
control maturation and function of microglia in the CNS. Mejía, J. L., Hansen, L. H., . . . Costabile, A. (2018). A
Nature Neuroscience, 18, 965–977. doi:10.1038/nn.4030 prebiotic intervention study in children with autism spec-
Fellows, R., Denizot, J., Stellato, C., Cuomo, A., Jain, P., Stoyanova, trum disorders (ASDs). Microbiome, 6, 133. doi:10.1186/
E., . . . Blackburn, H. (2018). Microbiota derived short chain s40168-018-0523-3
fatty acids promote histone crotonylation in the colon Guigoz, Y., Doré, J., & Schiffrin, E. J. (2008). The inflammatory
through histone deacetylases. Nature Communications, 9, status of old age can be nurtured from the intestinal environ-
Article 105. doi:10.1038/s41467-017-02651-5 ment. Current Opinion in Clinical Nutrition and Metabolic
Fernández, L., Langa, S., Martín, V., Maldonado, A., Jiménez, E., Care, 11, 13–20. doi:10.1097/MCO.0b013e3282f2bfdf
Martín, R., & Rodríguez, J. M. (2013). The human milk Hemmings, S. M. J., Malan-Müller, S., van den Heuvel, L. L.,
microbiota: Origin and potential roles in health and dis- Demmitt, B. A., Stanislawski, M. A., Smith, D. G., . . .
ease. Pharmacological Research, 69, 1–10. doi:10.1016/j Lowry, C. A. (2017). The microbiome in posttraumatic
.phrs.2012.09.001 stress disorder and trauma-exposed controls: An explor-
Forsythe, P., Bienenstock, J., & Kunze, W. A. (2014). Vagal atory study. Psychosomatic Medicine, 79, 936–946.
pathways for microbiome-brain-gut axis communication. doi:10.1097/PSY.0000000000000512
Microbes and the Mind 413

Hinde, K., & German, J. B. (2012). Food in an evolutionary antipsychotic induced weight gain. European Archives
context: Insights from mother’s milk. Journal of the Science of Psychiatry and Clinical Neuroscience, 268, 3–15.
of Food and Agriculture, 92, 2219–2223. doi:10.1002/ doi:10.1007/s00406-017-0820-z
jsfa.5720 Kato-Kataoka, A., Nishida, K., Takada, M., Kawai, M., Kikuchi-
Hoban, A. E., Stilling, R. M., Ryan, F. J., Shanahan, F., Dinan, Hayakawa, H., Suda, K., . . . Matsuki, T. (2016). Fermented
T. G., Claesson, M. J., . . . Cryan, J. F. (2016). Regulation milk containing Lactobacillus casei strain shirota preserves
of prefrontal cortex myelination by the microbiota. the diversity of the gut microbiota and relieves abdominal
Translational Psychiatry, 6, e774. doi:10.1038/tp.2016.42 dysfunction in healthy medical students exposed to aca-
Hollander, D. (1999). Intestinal permeability, leaky gut, and demic stress. Applied and Environmental Microbiology,
intestinal disorders. Current Gastroenterology Reports, 1, 82, 3649–3658. doi:10.1128/AEM.04134-15
410–416. Kelly, J. R., Allen, A. P., Temko, A., Hutch, W., Kennedy, P. J.,
Hollister, E. B., Riehle, K., Luna, R. A., Weidler, E. M., Rubio- Farid, N., . . . Dinan, T. G. (2017). Lost in translation?
Gonzales, M., Mistretta, T., . . . Versalovic, J. (2015). The potential psychobiotic Lactobacillus rhamnosus (JB-
Structure and function of the healthy pre-adolescent 1) fails to modulate stress or cognitive performance in
pediatric gut microbiome. Microbiome, 3, Article 36. healthy male subjects. Brain, Behavior, and Immunity,
doi:10.1186/s40168-015-0101-x 61, 50–59. doi:10.1016/j.bbi.2016.11.018
Hopkins, M. J., Sharp, R., & Macfarlane, G. T. (2002). Variation Kielstein, H., Suntharalingam, M., Perthel, R., Song, R.,
in human intestinal microbiota with age. Digestive and Schneider, S. M., Martens-Lobenhoffer, J., . . . Kielstein,
Liver Disease, 34(Suppl. 2), S12–S18. J. T. (2015). Role of the endogenous nitric oxide inhibitor
Huang, R., Wang, K., & Hu, J. (2016). Effect of probiotics asymmetric dimethylarginine (ADMA) and brain-derived
on depression: A systematic review and meta-analysis of neurotrophic factor (BDNF) in depression and behav-
randomized controlled trials. Nutrients, 8(8), Article 483. ioural changes: Clinical and preclinical data in chronic
doi:10.3390/nu8080483 kidney disease. Nephrology Dialysis Transplantation, 30,
Iyer, N., & Vaishnava, S. (2016). Alcohol lowers your (intes- 1699–1705. doi:10.1093/ndt/gfv253
tinal) inhibitions. Cell Host & Microbe, 19, 131–133. Kim, H., Yun, Y., Ryu, S., Chang, Y., Kwon, M., Cho, J.,
doi:10.1016/j.chom.2016.01.014 . . . Kim, H. (2018). Correlation between gut microbiota
Jackson, M. L., Butt, H., Ball, M., Lewis, D. P., & Bruck, D. and personality in adults: A cross-sectional study. Brain,
(2015). Sleep quality and the treatment of intestinal micro- Behavior, and Immunity, 69, 374–385. doi:10.1016/j.bbi
biota imbalance in chronic fatigue syndrome: A pilot study. .2017.12.012
Sleep Science (Sao Paulo, Brazil), 8, 124–133. doi:10.1016/j Kimura, I., Ozawa, K., Inoue, D., Imamura, T., Kimura, K.,
.slsci.2015.10.001 Maeda, T., . . . Tsujimoto, G. (2013). The gut microbiota
Janik, R., Thomason, L. A. M., Stanisz, A. M., Forsythe, P., suppresses insulin-mediated fat accumulation via the short-
Bienenstock, J., & Stanisz, G. J. (2016). Magnetic reso- chain fatty acid receptor GPR43. Nature Communications,
nance spectroscopy reveals oral Lactobacillus promotion 4, Article 1829. doi:10.1038/ncomms2852
of increases in brain GABA, N-acetyl aspartate and gluta- Kinross, J. M., von Roon, A. C., Holmes, E., Darzi, A., &
mate. NeuroImage, 125, 988–995. doi:10.1016/j.neuroim Nicholson, J. K. (2008). The human gut microbiome:
age.2015.11.018 Implications for future health care. Current Gastro­enter­
Jašarević , E., Howerton, C. L., Howard, C. D., & Bale, T. L. ology Reports, 10, 396–403.
(2015). Alterations in the vaginal microbiome by maternal Kleckner, I. R., Zhang, J., Touroutoglou, A., Chanes, L., Xia, C.,
stress are associated with metabolic reprogramming of the Simmons, W. K., . . . Barrett, L. F. (2017). Evidence for a
offspring gut and brain. Endocrinology, 156, 3265–3276. large-scale brain system supporting allostasis and intero-
doi:10.1210/en.2015-1177 ception in humans. Nature Human Behaviour, 1, Article
Jašarević, E., Rodgers, A. B., & Bale, T. L. (2015). A novel role 0069. doi:10.1038/s41562-017-0069
for maternal stress and microbial transmission in early Kligman, A. M., & Strauss, J. S. (1956). The bacteria respon-
life programming and neurodevelopment. Neurobiology sible for apocrine odor. The Journal of Investigative
of Stress, 1, 81–88. doi:10.1016/j.ynstr.2014.10.005 Dermatology, 27, 67–71.
Jiménez, E., Marín, M. L., Martín, R., Odriozola, J. M., Kok, B. E., Coffey, K. A., Cohn, M. A., Catalino, L. I.,
Olivares, M., Xaus, J., . . . Rodríguez, J. M. (2008). Is Vacharkulksemsuk, T., Algoe, S. B., . . . Fredrickson, B. L.
meconium from healthy newborns actually sterile? (2013). How positive emotions build physical health:
Research in Microbiology, 159, 187–193. doi:10.1016/j Perceived positive social connections account for the
.resmic.2007.12.007 upward spiral between positive emotions and vagal tone.
Jiménez, E., Villar-Tajadura, M., Marín, M., Fontecha, J., Psychological Science, 24, 1123–1132. doi:10.1177/09567976
Requena, T., Arroyo, R., . . . Rodríguez, J. M. (2012). 12470827
Complete genome sequence of Bifidobacterium breve Kong, J., Fang, J., Park, J., Li, S., & Rong, P. (2018). Treating
CECT 7263, a strain isolated from human milk. Journal depression with transcutaneous auricular vagus nerve stim-
of Bacteriology, 194, 3762–3763. doi:10.1128/JB.00691-12 ulation: State of the art and future perspectives. Frontiers
Kanji, S., Fonseka, T. M., Marshe, V. S., Sriretnakumar, V., in Psychiatry, 9, Article 20. doi:10.3389/fpsyt.2018.00020
Hahn, M. K., & Müller, D. J. (2018). The microbiome- Koren, O., Goodrich, J. K., Cullender, T. C., Spor, A., Laitinen,
gut-brain axis: Implications for schizophrenia and K., Helene, K. B., . . . Ley, R. E. (2012). Host remodeling of
414 Smith, Wissel

the gut microbiome and metabolic changes during preg- Maes, M., Kubera, M., Leunis, J., Berk, M., Geffard, M., &
nancy. Cell, 150, 470–480. doi:10.1016/j.cell.2012.07.008 Bosmans, E. (2013). In depression, bacterial transloca-
Kort, R., Caspers, M., van de Graaf, A., van Egmond, W., tion may drive inflammatory responses, oxidative and
Keijser, B., & Roeselers, G. (2014). Shaping the oral micro- nitrosative stress (O&NS), and autoimmune responses
biota through intimate kissing. Microbiome, 2, Article 41. directed against O&NS-damaged neoepitopes. Acta
doi:10.1186/2049-2618-2-41 Psychiatrica Scandinavica, 127, 344–354. doi:10.1111/
Kramer, M. S., Aboud, F., Mironova, E., Vanilovich, I., Platt, j.1600-0447.2012.01908.x
R. W., Matush, L., . . . Shapiro, S. (2008). Breastfeeding Magnusson, K. R., Hauck, L., Jeffrey, B. M., Elias, V.,
and child cognitive development: New evidence from a Humphrey, A., Nath, R., . . . Bermudez, L. E. (2015).
large randomized trial. Archives of General Psychiatry, 65, Relationships between diet-related changes in the gut
578–584. doi:10.1001/archpsyc.65.5.578 microbiome and cognitive flexibility. Neuroscience, 300,
Kramer, P., & Bressan, P. (2015). Humans as superorganisms: 128–140. doi:10.1016/j.neuroscience.2015.05.016
How microbes, viruses, imprinted genes, and other selfish Malan-Muller, S., Valles-Colomer, M., Raes, J., Lowry, C. A.,
entities shape our behavior. Perspectives on Psychological Seedat, S., & Hemmings, S. M. J. (2018). The gut micro-
Science, 10, 464–481. doi:10.1177/1745691615583131 biome and mental health: Implications for anxiety- and
Lencner, A. A., Lencner, C. P., Mikelsaar, M. E., Tjuri, M. E., trauma-related disorders. OMICS, 22, 90–107. doi:10.1089/
Toom, M. A., Väljaots, M. E., . . . Reznikov, I. M. (1984). omi.2017.0077
Die quantitative Zusammensetzung der Lactoflora des Martin, F. J., Rezzi, S., Peré-Trepat, E., Kamlage, B., Collino,
Verdauungstrakts vor und nach kosmischen Flügen unter- S., Leibold, E., . . . Kochhar, S. (2009). Metabolic effects
schiedlicher Dauer. [The quantitative composition of the of dark chocolate consumption on energy, gut microbi-
intestinal lactoflora before and after space flights of dif- ota, and stress-related metabolism in free-living subjects.
ferent lengths]. Nahrung, 28, 607–613. Journal of Proteome Research, 8, 5568–5579. doi:10.1021/
Levkovich, T., Poutahidis, T., Smillie, C., Varian, B. J., Ibrahim, pr900607v
Y. M., Lakritz, J. R., . . . Erdman, S. E. (2013). Probiotic Martin-Subero, M., Anderson, G., Kanchanatawan, B., Berk,
bacteria induce a ‘glow of health.’ PLOS ONE, 8(1), Article M., & Maes, M. (2016). Comorbidity between depression
e53867. doi:10.1371/journal.pone.0053867 and inflammatory bowel disease explained by immune-
Li, W., Dowd, S. E., Scurlock, B., Acosta-Martinez, V., & Lyte, M. inflammatory, oxidative, and nitrosative stress; tryptophan
(2009). Memory and learning behavior in mice is tempo- catabolite; and gut-brain pathways. CNS Spectrums, 21,
rally associated with diet-induced alterations in gut bac- 184–198. doi:10.1017/S1092852915000449
teria. Physiology & Behavior, 96, 557–567. doi:10.1016/j Mason, M. R., Nagaraja, H. N., Camerlengo, T., Joshi, V., &
.physbeh.2008.12.004 Kumar, P. S. (2013). Deep sequencing identifies ethnic-
Liu, C. M., Hungate, B. A., Tobian, A. A. R., Ravel, J., Prodger, J. L., ity-specific bacterial signatures in the oral microbiome.
Serwadda, D., . . . Gray, R. H. (2015). Penile microbiota PLOS ONE, 8(10), Article e77287. doi:10.1371/journal
and female partner bacterial vaginosis in Rakai, Uganda. .pone.0077287
mBio, 6(3), e00589–e00615. doi:10.1128/mBio.00589-15 Matamoros, S., Gras-Leguen, C., Le Vacon, F., Potel, G., de
Liu, R. T. (2017). The microbiome as a novel paradigm in La & Cochetiere, M. F. (2013). Development of intestinal
studying stress and mental health. American Psychologist, microbiota in infants and its impact on health. Trends in
72, 655–667. Microbiology, 21, 167–173. doi:10.1016/j.tim.2012.12.001
Logsdon, A. F., Erickson, M. A., Rhea, E. M., Salameh, T. S., Matthews, D. M., & Jenks, S. M. (2013). Ingestion of
& Banks, W. A. (2018). Gut reactions: How the blood- Mycobacterium vaccae decreases anxiety-related behav-
brain barrier connects the microbiome and the brain. ior and improves learning in mice. Behavioural Processes,
Experimental Biology and Medicine, 243, 159–165. doi:10 96, 27–35. doi:10.1016/j.beproc.2013.02.007
.1177/1535370217743766 McLean, P. G., Bergonzelli, G. E., Collins, S. M., & Bercik, P.
Luczynski, P., McVey Neufeld, K., Oriach, C. S., Clarke, G., (2012). Targeting the microbiota-gut-brain axis to modu-
Dinan, T. G., & Cryan, J. F. (2016). Growing up in a late behavior: Which bacterial strain will translate best to
bubble: Using germ-free animals to assess the influence of humans? Proceedings of the National Academy of Sciences,
the gut microbiota on brain and behavior. International USA, 109(4), Article e174. doi:10.1073/pnas.1118626109
Journal of Neuropsychopharmacology, 19(8), Article Meadow, J. F., Altrichter, A. E., Bateman, A. C., Stenson, J.,
pyw020. doi:10.1093/ijnp/pyw020 Brown, G. Z., Green, J. L., & Bohannan, B. J. M. (2015).
Luna, R. A., Savidge, T. C., & Williams, K. C. (2016). The brain- Humans differ in their personal microbial cloud. PeerJ,
gut-microbiome axis: What role does it play in autism 3, Article e1258. doi:10.7717/peerj.1258
spectrum disorder? Current Developmental Disorders Messaoudi, M., Lalonde, R., Violle, N., Javelot, H., Desor, D.,
Reports, 3, 75–81. doi:10.1007/s40474-016-0077-7 Nejdi, A., . . . Cazaubiel, J. (2011). Assessment of psychotro-
Lydiard, R. B. (2003). The role of GABA in anxiety disorders. pic-like properties of a probiotic formulation (Lactobacillus
Journal of Clinical Psychiatry, 64(Suppl. 3), 21–27. helveticus R0052 and Bifidobacterium longum R0175) in
Maes, M., Kubera, M., Leunis, J., & Berk, M. (2012). Increased rats and human subjects. British Journal of Nutrition, 105,
IgA and IgM responses against gut commensals in chronic 755–764. doi:10.1017/S0007114510004319
depression: Further evidence for increased bacterial trans- Mika, A., Gaffney, M., Roller, R., Hills, A., Bouchet, C. A.,
location or leaky gut. Journal of Affective Disorders, 141, Hulen, K. A., . . . Fleshner, M. (2018). Feeding the devel-
55–62. doi:10.1016/j.jad.2012.02.023 oping brain: Juvenile rats fed diet rich in prebiotics and
Microbes and the Mind 415

bioactive milk fractions exhibit reduced anxiety-related paraprobiotic Lactobacillus gasseri CP2305 ameliorates
behavior and modified gene expression in emotion cir- chronic stress-associated symptoms in Japanese medi-
cuits. Neuroscience Letters, 677, 103–109. doi:10.1016/j cal students. Journal of Functional Foods, 36, 112–121.
.neulet.2018.01.052 doi:10.1016/j.jff.2017.06.031
Mika, A., Will, V. T., González, A., Herrera, J. J., Knight, R., & Nowakowski, M. E., McCabe, R., Rowa, K., Pellizzari, J.,
Fleshner, M. (2015). Exercise is more effective at altering Surette, M., Moayyedi, P., & Anglin, R. (2016). The gut
gut microbial composition and producing stable changes in microbiome: Potential innovations for the understanding
lean mass in juvenile versus adult male F344 rats. PLOS ONE, and treatment of psychopathology. Canadian Psychology,
10(5), Article e0125889. doi:10.1371/journal.pone.0125889 57(2), 67–75. doi:10.1037/cap0000038
Misra, S., & Mohanty, D. (2017). Psychobiotics: A new O’Hagan, C., Li, J. V., Marchesi, J. R., Plummer, S., Garaiova, I.,
approach for treating mental illness? Critical Reviews in & Good, M. A. (2017). Long-term multi-species
Food Science and Nutrition. Advance online publication. Lactobacillus and Bifidobacterium dietary supplement
doi:10.1080/10408398.2017.1399860 enhances memory and changes regional brain metabo-
Montiel-Castro, A. J., Augusto, J., Báez-Yáñez Mario, G., & lites in middle-aged rats. Neurobiology of Learning and
Pacheco-López, G. (2014). Social neuroeconomics: The Memory, 144, 36–47. doi:10.1016/j.nlm.2017.05.015
influence of microbiota in partner-choice and sociality. O’Mahony, S. M., Felice, V. D., Nally, K., Savignac, H.
Current Pharmaceutical Design, 20, 4774–4783. doi:10.2 M., Claesson, M. J., Scully, P., . . . Cryan, J. F. (2014).
174/1381612820666140130210631 Disturbance of the gut microbiota in early-life selec-
Mueller, N. T., Bakacs, E., Combellick, J., Grigoryan, Z., tively affects visceral pain in adulthood without impact-
& Dominguez-Bello, M. (2015). The infant microbi- ing cognitive or anxiety-related behaviors in male rats.
ome development: Mom matters. Trends in Molecular Neuroscience, 277, 885–901. doi:10.1016/j.neurosci
Medicine, 21, 109–117. doi:10.1016/j.molmed.2014.12.002 ence.2014.07.054
Mueller, N. T., Whyatt, R., Hoepner, L., Oberfield, S., O’Toole, P. W., & Claesson, M. J. (2010). Gut microbiota:
Dominguez-Bello, M., Widen, E. M., . . . Rundle, A. Changes throughout the lifespan from infancy to elderly.
(2015). Prenatal exposure to antibiotics, cesarean sec- International Dairy Journal, 20, 281–291. doi:10.1016/j
tion and risk of childhood obesity. International Journal .idairyj.2009.11.010
of Obesity, 39, 665–670. doi:10.1038/ijo.2014.180 Palmer, C., Bik, E. M., DiGiulio, D. B., Relman, D. A., &
Muhtadie, L., Koslov, K., Akinola, M., & Mendes, W. B. (2015). Brown, P. O. (2007). Development of the human infant
Vagal flexibility: A physiological predictor of social sensi- intestinal microbiota. PLOS Biology, 5(7), Article e177.
tivity. Journal of Personality and Social Psychology, 109, doi:10.1371/journal.pbio.0050177
106–120. doi:10.1037/pspp0000016 Paolicelli, R. C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M.,
Mutlu, E., Keshavarzian, A., Engen, P., Forsyth, C. B., Panzanelli, P., . . . Gross, C. T. (2011). Synaptic pruning
Sikaroodi, M., & Gillevet, P. (2009). Intestinal dysbiosis: by microglia is necessary for normal brain development.
A possible mechanism of alcohol-induced endotoxemia Science, 333, 1456–1458. doi:10.1126/science.1202529
and alcoholic steatohepatitis in rats. Alcoholism: Clinical Parashar, A., & Udayabanu, M. (2016). Gut microbiota reg-
and Experimental Research, 33, 1836–1846. doi:10.1111/ ulates key modulators of social behavior. European
j.1530-0277.2009.01022.x Neuropsychopharmacology, 26, 78–91. doi:10.1016/j
Nemeroff, C. B. (2003). The role of GABA in the patho- .euroneuro.2015.11.002
physiology and treatment of anxiety disorders. Psy­ Patriquin, M. A., Scarpa, A., Friedman, B. H., & Porges, S. W.
chopharmacology Bulletin, 37, 133–146. (2013). Respiratory sinus arrhythmia: A marker for posi-
Neufeld, K. M., Kang, N., Bienenstock, J., & Foster, J. A. tive social functioning and receptive language skills in
(2011). Reduced anxiety-like behavior and central neuro- children with autism spectrum disorders. Developmental
chemical change in germ-free mice. Neurogastroenterology Psychobiology, 55, 101–112. doi:10.1002/dev.21002
and Motility, 23, 255–264. doi:10.1111/j.1365-2982.2010 Perez-Burgos, A., Wang, B., Mao, Y., Mistry, B., Neufeld, K. M.,
.01620.x Bienenstock, J., & Kunze, W. (2013). Psychoactive bac-
Neufeld, K. M., Luczynski, P., Oriach, C. S., Dinan, T. G., teria Lactobacillus rhamnosus (JB-1) elicits rapid fre-
& Cryan, J. F. (2016). What’s bugging your teen? The quency facilitation in vagal afferents. American Journal
microbiota and adolescent mental health. Neuroscience of Physiology: Gastrointestinal and Liver Physiology, 304,
and Biobehavioral Reviews, 70, 300–312. doi:10.1016/j G211–G220. doi:10.1152/ajpgi.00128.2012
.neubiorev.2016.06.00 Petra, A. I., Panagiotidou, S., Hatziagelaki, E., Stewart, J. M.,
Nicholson, B. (1984). Does kissing aid human bonding by Conti, P., & Theoharides, T. C. (2015). Gut-microbiota-
semiochemical addiction? British Journal of Dermatology, brain axis and its effect on neuropsychiatric disorders with
111, 623–627. suspected immune dysregulation. Clinical Therapeutics,
Nieto, R., Kukuljan, M., & Silva, H. (2013). BDNF and schizo- 37, 984–995. doi:10.1016/j.clinthera.2015.04.002
phrenia: From neurodevelopment to neuronal plasticity, Phaf, R. H., Mohr, S., Rotteveel, M., & Wicherts, J. M. (2014).
learning, and memory. Frontiers in Psychiatry, 4, Article Approach, avoidance, and affect: A meta-analysis of
45. doi:10.3389/fpsyt.2013.00045 approach-avoidance tendencies in manual reaction time
Nishida, K., Sawada, D., Kuwano, Y., Tanaka, H., Sugawara, T., tasks. Frontiers in Psychology, 5, Article 378. doi:10.3389/
Aoki, Y., . . . Rokutan, K. (2017). Daily administration of fpsyg.2014.00378
416 Smith, Wissel

Pinto-Sanchez, M. I., Ford, A. C., Avila, C. A., Verdu, E. F., Salazar, N., Arboleya, S., Valdés, L., Stanton, C., Ross, P., Ruiz, L.,
Collins, S. M., Morgan, D., . . . Bercik, P. (2015). Anxiety . . . de Los Reyes-Gavilán, C. G. (2014). The human
and depression increase in a stepwise manner in parallel intestinal microbiome at extreme ages of life. Dietary
with multiple FGIDs and symptom severity and frequency. intervention as a way to counteract alterations. Frontiers
American Journal of Gastroenterology, 110, 1038–1048. in Genetics, 5, Article 406. doi:10.3389/fgene.2014.00406
doi:10.1038/ajg.2015.128 Satokari, R., Grönroos, T., Laitinen, K., Salminen, S., & Isolauri, E.
Pinto-Sanchez, M. I., Hall, G. B., Ghajar, K., Nardelli, A., (2009). Bifidobacterium and Lactobacillus DNA in the
Bolino, C., Lau, J. T., . . . Bercik, P. (2017). Probiotic human placenta. Letters in Applied Microbiology, 48, 8–12.
Bifidobacterium longum NCC3001 reduces depression doi:10.1111/j.1472-765X.2008.02475.x
scores and alters brain activity: A pilot study in patients Saunders, P. R., Santos, J., Hanssen, N. P., Yates, D., Groot,
with irritable bowel syndrome. Gastroenterology, 153, J. A., & Perdue, M. H. (2002). Physical and psychological
448–459. doi:10.1053/j.gastro.2017.05.003 stress in rats enhances colonic epithelial permeability
Poutahidis, T., Kleinewietfeld, M., Smillie, C., Levkovich, T., via peripheral CRH. Digestive Diseases and Sciences, 47,
Perrotta, A., Bhela, S., . . . Erdman, S. E. (2013). Microbial 208–215. doi:10.1136/gut.2009.202515
reprogramming inhibits western diet-associated obesity. Savignac, H. M., Tramullas, M., Kiely, B., Dinan, T. G., &
PLOS ONE, 8(7), Article e68596. doi:10.1371/journal Cryan, J. F. (2015). Bifidobacteria modulate cognitive pro-
.pone.0068596 cesses in an anxious mouse strain. Behavioural Brain
Poutahidis, T., Springer, A., Levkovich, T., Qi, P., Varian, B. J., Research, 287, 59–72. doi:10.1016/j.bbr.2015.02.044
Lakritz, J. R., . . . Erdman, S. E. (2014). Probiotic microbes Schafer, D., Lehrman, E., Kautzman, A., Koyama, R., Mardinly, A.,
sustain youthful serum testosterone levels and testicu- Yamasaki, R., . . . Stevens, B. (2012). Microglia sculpt
lar size in aging mice. PLOS ONE, 9(1), Article e84877. postnatal neural circuits in an activity and complement-
doi:10.1371/journal.pone.0084877 dependent manner. Neuron, 74, 691–705. doi:10.1016/j
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K. S., .neuron.2012.03.026
Manichanh, C., . . . Wang, J. (2010). A human gut micro- Schele, E., Grahnemo, L., Anesten, F., Hallen, A., Baeckhed, F.,
bial gene catalogue established by metagenomic sequenc- & Jansson, J. (2013). The gut microbiota reduces leptin
ing. Nature, 464, 59–65. doi:10.1038/nature08821 sensitivity and the expression of the obesity-suppressing
Quintana, D. S., Guastella, A. J., Outhred, T., Hickie, I. B., neuropeptides proglucagon (Gcg) and brain-derived neu-
& Kemp, A. H. (2012). Heart rate variability is associated rotrophic factor (Bdnf) in the central nervous system.
with emotion recognition: Direct evidence for a relation- Endocrinology, 154, 3643–3651. doi:10.1210/en.2012-2151
ship between the autonomic nervous system and social Schmidt, K., Cowen, P. J., Harmer, C. J., Tzortzis, G., Errington,
cognition. International Journal of Psychophysiology, 86, S., & Burnet, P. W. J. (2015). Prebiotic intake reduces the
168–172. doi:10.1016/j.ijpsycho.2012.08.012 waking cortisol response and alters emotional bias in
Rao, A. V., Bested, A. C., Beaulne, T. M., Katzman, M. A., Iorio, C., healthy volunteers. Psychopharmacology, 232, 1793–1801.
Berardi, J. M., & Logan, A. C. (2009). A randomized, doi:10.1007/s00213-014-3810-0
double-blind, placebo-controlled pilot study of a probi- Sender, R., Fuchs, S., & Milo, R. (2016). Revised estimates
otic in emotional symptoms of chronic fatigue syndrome. for the number of human and bacteria cells in the body.
Gut Pathogens, 1(1), Article 6. doi:10.1186/1757-4749-1-6 PLOS Biology, 14(8), Article e1002533. doi:10.1371/journal
Rautava, S., Collado, M. C., Salminen, S., & Isolauri, E. .pbio.1002533
(2012). Probiotics modulate host-microbe interaction in Severance, E. G., Yolken, R. H., & Eaton, W. W. (2016).
the placenta and fetal gut: A randomized, double-blind, Autoimmune diseases, gastrointestinal disorders and
placebo-controlled trial. Neonatology, 102, 178–184. the microbiome in schizophrenia: More than a gut feel-
doi:10.1159/000339182 ing. Schizophrenia Research, 176, 23–35. doi:10.1016/j
Ravel, J., Gajer, P., Abdo, Z., Schneider, G. M., Koenig, S. S. K., .schres.2014.06.027
McCulle, S. L., . . . Forney, L. J. (2011). Vaginal micro- Shahrestani, S., Stewart, E. M., Quintana, D. S., Hickie, I. B.,
biome of reproductive-age women. Proceedings of the & Guastella, A. J. (2015). Heart rate variability during
Na­tional Academy of Sciences, USA, 108, 4680–4687. doi: adolescent and adult social interactions: A meta-analysis.
10.1073/pnas.1002611107 Biological Psychology, 105, 43–50. doi:10.1016/j.biopsycho
Reigstad, C. S., Salmonson, C. E., Rainey, J. F., Szurszewski, J. H., .2014.12.012
Linden, D. R., Sonnenburg, J. L., . . . Kashyap, P. C. Sharon, G., Segal, D., Ringo, J. M., Hefetz, A., Zilber-Rosenberg, I.,
(2015). Gut microbes promote colonic serotonin pro- & Rosenberg, E. (2010). Commensal bacteria play a role
duction through an effect of short-chain fatty acids on in mating preference of drosophila melanogaster. Pro­
enterochromaffin cells. FASEB Journal, 29, 1395–1403. ceedings of the National Academy of Sciences, USA, 107,
doi:10.1096/fj.14-259598 20051–20056. doi:10.1073/pnas.1009906107
Roger, L. C., Costabile, A., Holland, D. T., Hoyles, L., & McCartney, Shimizu, E., Hashimoto, K., Okamura, N., Koike, K., Komatsu, N.,
A. L. (2010). Examination of faecal Bifidobacterium popula- Kumakiri, C., . . . Iyo, M. (2003). Alterations of serum levels
tions in breast- and formula-fed infants during the first 18 of brain-derived neurotrophic factor (BDNF) in depressed
months of life. Microbiology, 156, 3329–3341. doi:10.1099/ patients with or without antidepressants. Biological
mic.0.043224-0 Psychiatry, 54, 70–75. doi:10.1016/S0006-3223(03)00181-1
Microbes and the Mind 417

Siegel, E. H., Wormwood, J. B., Quigley, K. S., & Barrett, L. F. adolescent academic achievement. Child Development,
(2018). Seeing what you feel: Affect drives visual percep- 83, 743–757. doi:10.1111/j.1467-8624.2011.01721.x
tion of structurally neutral faces. Psychological Science, Tucker-Drob, E. M., Rhemtulla, M., Harden, K. P.,
29, 496–505. doi:10.1177/0956797617741718 Turkheimer, E., & Fask, D. (2011). Emergence of a Gene ×
Singh, D., & Bronstad, P. M. (2001). Female body odour is Socioeconomic Status interaction on infant mental ability
a potential cue to ovulation. Proceedings of the Royal between 10 months and 2 years. Psychological Science,
Society of London, Series B: Biological Sciences, 268, 797– 22, 125–133. doi:10.1177/0956797610392926
801. doi:10.1098/rspb.2001.1589 United Nations. (2015). World population ageing 2015 (ST/
Slyepchenko, A., Maes, M., Machado-Vieira, R., Anderson, G., ESA/SER.A/390). New York, NY: Author. Retrieved from
Solmi, M., Sanz, Y. F., . . . Carvalho, A. (2016). Intestinal http://www.un.org/en/development/desa/population/
dysbiosis, gut hyperpermeability and bacterial transloca- publications/pdf/ageing/WPA2015_Report.pdf
tion: Missing links between depression, obesity and type Vaishampayan, P. A., Kuehl, J. V., Froula, J. L., Morgan, J. L.,
2 diabetes. Current Pharmaceutical Design, 22, 6087– Ochman, H., & Francino, M. P. (2010). Comparative
6106. doi:10.2174/1381612822666160922165706 metagenomics and population dynamics of the gut
Song, Y., Liu, C., & Finegold, S. M. (2004). Real-time PCR micr­obiota in mother and infant. Genome Biology and
quantitation of clostridia in feces of autistic children. Evolution, 2, 53–66. doi:10.1093/gbe/evp057
Applied and Environmental Microbiology, 70, 6459–6465. van de Wouw, M., Boehme, M., Lyte, J. M., Wiley, N., Strain, C.,
doi:10.1128/AEM.70.11.6459-6465.2004 O’Sullivan, O., . . . Cryan, J. F. (2018). Short-chain
Stakenborg, N., Di Giovangiulio, M., Boeckxstaens, G. E., fatty acids: Microbial metabolites that alleviate stress-
& Matteoli, G. (2013). The versatile role of the vagus induced brain-gut axis alterations. Journal of Physiology.
nerve in the gastrointestinal tract. EMJ Gastroenterology, doi:10.1113/JP276431
1, 106–114. Van den Bergh, B. R. H. (1990). The influence of maternal
Stilling, R. M., Dinan, T. G., & Cryan, J. F. (2016). The brain’s emotions during pregnancy on fetal and neonatal behav-
Geppetto-microbes as puppeteers of neural function ior. Journal of Prenatal & Perinatal Psychology & Health,
and behaviour? Journal of Neurovirology, 22, 14–21. 5(2), 119–130.
doi:10.1007/s13365-015-0355-x Varian, B. J., Levkovich, T., Poutahidis, T., Ibrahim, Y. M.,
Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X., Perrotta, A., Alm, E. J., & Erdman, S. E. (2016). Beneficial
. . . Koga, Y. (2004). Postnatal microbial colonization pro- dog bacteria up-regulate oxytocin and lower risk of obesity.
grams the hypothalamic-pituitary-adrenal system for stress Journal of Probiotics and Health, 4(3), Article 149. doi: 10
response in mice. Journal of Physiology, 558, 263–275. .4172/2329-8901.1000149
doi:10.1113/jphysiol.2004.063388 Varian, B. J., Poutahidis, T., DiBenedictis, B. T., Levkovich, T.,
Svendsen, J. L., Osnes, B., Binder, P., Dundas, I., Visted, E., Ibrahim, Y., Didyk, E., . . . Erdman, S. E. (2017). Microbial
Nordby, H., . . . Sørensen, L. (2016). Trait self-compassion lysate upregulates host oxytocin. Brain, Behavior, and
reflects emotional flexibility through an association with Immunity, 61, 36–49. doi:10.1016/j.bbi.2016.11.002
high vagally mediated heart rate variability. Mindfulness, Wall, R., Cryan, J. F., Ross, R. P., Fitzgerald, G. F., Dinan, T. G.,
7, 1103–1113. doi:10.1007/s12671-016-0549-1 & Stanton, C. (2014). Bacterial neuroactive compounds
Thayer, J. F., Åhs, F., Fredrikson, M., Sollers, J. J., III, & Wager, T. D. produced by psychobiotics. Advances in Experimental
(2012). A meta-analysis of heart rate variability and neuro- Medicine and Biology, 817, 221–239. doi:10.1007/978-1-
imaging studies: Implications for heart rate variability as a 4939-0897-4_10
marker of stress and health. Neuroscience and Biobehavioral Wallace, C. J. K., & Milev, R. (2017). The effects of probiotics
Reviews, 36, 747–756. doi:10.1016/j.neubiorev.2011 on depressive symptoms in humans: A systematic review.
.11.009 Annals of General Psychiatry, 16, Article 14. doi:10.1186/
Theis, K. R., Dheilly, N. M., Klassen, J. L., Brucker, R. M., s12991-017-0138-2
Baines, J. F., Bosch, T. C., . . . Sapp, J. (2016). Getting the Watanabe, I. (1998). Masticatory function and life style
hologenome concept right: An eco-evolutionary frame- in aged. Japanese Journal of Geriatrics, 35, 194–200.
work for hosts and their microbiomes. mSystems, 1(2), doi:10.3143/geriatrics.35.194
Article e0002816. doi:10.1128/mSystems.00028-16 Waworuntu, R. V., Neufeld, K. A. M., O’Mahony, S. M., Pusceddu, M.,
Tillisch, K., Labus, J. S., Ebrat, B., Stains, J., Naliboff, B. D., Berg, B. M., Dinan, T. G., & Cryan, J. F. (2017). A combination
Guyonnet, D., . . . Mayer, E. A. (2012). 589 modulation of of milk fat globule membrane and prebiotics modulate pain
the brain-gut axis after 4-week intervention with a pro- hypersensitivity and cognitive responses to maternal separa-
biotic fermented dairy product. Gastroenterology, 142(5 tion stress. FASEB Journal, 31(Suppl. 1), 448–450. doi:10.1096/
Suppl. 1), S-115. doi:10.1016/S0016-5085(12)60435-1 fasebj.31.1_supplement.448.5
Tucker-Drob, E. M., & Bates, T. C. (2016). Large cross-national Weathers, F. W., Litz, B. T., Keane, T. M., Palmieri, P. A., Marx,
differences in Gene × Socioeconomic Status interaction B. P., & Schnurr, P. P. (2013). The PTSD checklist for
on intelligence. Psychological Science, 27, 138–149. doi:10 DSM-5 (PCL-5). Retrieved from https://www.ptsd.va.gov/
.1177/0956797615612727 professional/assessment/adult-sr/ptsd-checklist.asp
Tucker-Drob, E. M., & Harden, K. P. (2012). Intellectual inter- White, J., Murielle, R., Massot, M., & Meylan, S. (2011).
est mediates Gene × Socioeconomic Status interaction on Cloacal bacterial diversity increases with multiple mates:
418 Smith, Wissel

Evidence of sexual transmission in female common liz- Wissel, E. F. (2018). Bacterial strains that impact mood
ards. PLOS ONE, 6(7), Article e22339. doi:10.1371/journal [Organizational spreadsheet]. Retrieved from https://
.pone.0022339 docs.google.com/spreadsheets/d/1wgoYlOZP7uFXlbajj
White House Office of Science and Technology Policy. 1hu-Pa1O8qI6ZJP4-Y69qcsk0Y/edit?usp=sharing
(2016). Announcing the National Microbiome Initiative. Wren, A. M., & Bloom, S. R. (2007). Gut hormones and appe-
Washington, DC: Author. Retrieved from https://obam tite control. Gastroenterology, 132, 2116–2130. doi:10
awhitehouse.archives.gov/the-press-office/2016/05/12/ .1053/j.gastro.2007.03.048
fact-sheet-announcing-national-microbiome-initiative Yan, A. W., & Schnabl, B. (2012). Bacterial translocation and
Wikoff, W. R., Anfora, A. T., Liu, J., Schultz, P. G., Lesley, S. changes in the intestinal microbiome associated with
A., Peters, E. C., & Siuzdak, G. (2009). Metabolomics anal- alcoholic liver disease. World Journal of Hepatology, 4,
ysis reveals large effects of gut microflora on mammalian 110–118. doi:10.4254/wjh.v4.i4.110
blood metabolites. Proceedings of the National Academy Zigmond, A. S., & Snaith, R. P. (1983). The hospital anxiety
of Sciences, USA, 106, 3698–3703. doi:10.1073/pnas.08128 and depression scale. Acta Psychiatrica Scandinavica,
74106 67, 361–370.
Wiley, N. C., Dinan, T. G., Ross, R. P., Stanton, C., Clarke, G., Zijlmans, M. A., Korpela, K., Riksen-Walraven, J. M., de
& Cryan, J. F. (2017). The microbiota-gut-brain axis as a Vos, W. M., & de Weerth, C. (2015). Maternal prenatal
key regulator of neural function and the stress response: stress is associated with the infant intestinal microbiota.
Implications for human and animal health. Journal of Psychoneuroendocrinology, 53, 233–245. doi:10.1016/j
Animal Science, 95, 3225–3246. doi:10.2527/jas2016.1256 .psyneuen.2015.01.006

Das könnte Ihnen auch gefallen