You are on page 1of 20

Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


journal homepage: www.elsevier.com/locate/neubiorev

The connection between microbiome and schizophrenia T


a,b, a
Bogdana Golofast *, Karel Vales
a
National Institute of Mental Health, Topolova 748, 250 67 Klecany, Prague East, Czech Republic
b
Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic

A R T I C LE I N FO A B S T R A C T

Keywords: There has been an accumulation of knowledge about the human microbiome, some detailed investigations of the
Microbiome gastrointestinal microbiota and its functions, and the highlighting of complex interactions between the gut, the
Schizophrenia gut microbiota, and the central nervous system. That assumes the involvement of the microbiome in the pa-
Immune inflammatory response thogenesis of various CNS diseases, including schizophrenia. Given this information and the fact, that the gut
Prenatal environment
microbiota is sensitive to internal and environmental influences, we have speculated that among the factors that
Postnatal environment
Cesarean section
influence the formation and composition of gut microbiota during life, possible key elements in the schizo-
Probiotics phrenia development chain are hidden where gut microbiota is a linking component. This article aims to de-
Prebiotics scribe and understand the developmental relationships between intestinal microbiota and the risk of developing
schizophrenia.

1. Introduction et al., 2011), and pathogens can either be a reason of inflammatory


diseases of the central nervous system or protect from them (Ochoa-
The human microbiota is unequally distributed in the body and Repáraz et al., 2011). Epidemiological studies have shown a link be-
includes the aggregate of all microorganisms, that reside on or within tween microbial infections early in life and neurodevelopmental dis-
human tissues and biofluids along with the corresponding anatomical orders, including autism and schizophrenia (Finegold et al., 2002;
sites in which they inhabit (Marchesi et al., 2016). The most abundant Mittal et al., 2008). The gut microbiota can affect the immune response
microbiota in our body is gastrointestinal (gut) microbiota. It may by activating the immune system or through mediators that are able to
contain hundreds of species of different microorganisms but in adult penetrate the blood-brain barrier (BBB) or other chemicals-related
members, Actinobacteria, Bacteroidetes and Firmicutes predominate substances that have free access to the brain. The mechanisms about
(Rajilić-Stojanović et al., 2009). The gastrointestinal microbiota is how the gut microbiota may affect brain functions will be discussed in
better studied than other human bacterial communities. And studies detail here later on.
described below have precisely shown that the intestinal microbiota has The microorganisms count populating the gastrointestinal tract has
a great impact on the health of its host. been evaluated to transcend 1014, that corresponds of ten times more
The human gastrointestinal tract is a portrait of one of the biggest bacterial cells than the number of human cells and over 100 times the
associations between the host, environments, and antigens within the number of genomic composition (microbiome) as the human genome
human body. During the average human lifetime, around 60 tons of (Bäckhed, 2005; Gill et al., 2006). However, a revised assessment has
food with plenty of microorganisms from the ambient environment indicated that the ratio of human cells over bacterial cells is closer to
transfer through the gastrointestinal tract, which can pose a big threat equal and their total weight is about 200 g (Sender et al., 2016). The
on the intestinal entirety (Bengmark, 1998). Intestinal microbes play a interrelation with the human host and the bacterial colonizers are that
pivotal part in keeping up the resistance and the metabolic homeostasis these relationships benefit both parties: the microbiome is granted with
effective and are safeguarding against pathogenic microbes. The colony an environment to live in and an easy existing nutriments source
of bacteria, archaea and eukarya populating the gastrointestinal tract is (Wexler, 2007) and the microbiota offers numerous bonuses to the host,
designated by «gut microbiota» and has developed together with the due to physiological capacities such as harvesting energy (den Besten
host for an extended period to create a complicated and useful liaison et al., 2013), strengthening gut integrity or shaping the intestinal epi-
(Bäckhed, 2005; Neish, 2009). Gut microbiota is an important mod- thelium (Natividad and Verdu, 2013), protecting against pathogens
ulator of brain development and subsequent adult behavior (Diaz Heijtz (Bäumler and Sperandio, 2016) and regulating host immunity


Corresponding author.
E-mail address: bogdana.golofast@nudz.cz (B. Golofast).

https://doi.org/10.1016/j.neubiorev.2019.12.011
Received 30 August 2019; Received in revised form 1 December 2019; Accepted 6 December 2019
Available online 09 December 2019
0149-7634/ © 2019 Elsevier Ltd. All rights reserved.
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

(Gensollen et al., 2016). Microorganisms which live inside our digestive Some of the important functions of the intestinal microbiota are
tract are beneficial and essential for the proper development of the syntheses of specific lipopolysaccharides and certain vitamins and
central nervous system (CNS), for brain response (Cryan and O’Mahony, amino acids (Lloyd-Price et al., 2016), short-chain fatty acids, and of
2011; Dinan and Cryan, 2013; Qin et al., 2010) and the regulation of course the degradation of polysaccharides. Genes coding for 3200 un-
host physiology. The ecosystem in the human gut is dynamic, there is ique chemical reactions have been updated through a recent metabolic
potential for these processes to be disordered because of a transformed genome reconstruction of 773 genome members of the human intestinal
microbial composition, known as dysbiosis. With progressively in- microbiota, which would mean that this community encodes hundreds
novative techniques to characterize ecosystems being developed, a or even thousands of metabolic pathways (Magnúsdóttir et al., 2017).
potential role of gut microbiota for intestinal and extraintestinal dis-
eases has become clear (Chang and Lin, 2016; Schroeder and Bäckhed, 2.2. Microbiota-gut-brain axis. Ways of interaction. An overview of the gut
2016). microbiota in behavior
This review presents data on the development and configuration of
the human gastrointestinal microbiota, its main functions and interac- Studies on germ-free (GF) rodents, gastrointestinal tract infections,
tion mechanisms in the microbiota-gut-brain axis. This article has been antibiotics, probiotics, and bacteriotherapy have shown that intestinal
made through investigations studying the relationship between gut microorganisms regulate the brain, behaviors, and stress responses by
microbiota and neuropsychiatric conditions, behavioral traits in order the creation of the microbiota-gut-brain (MGB) axis (Cryan and Dinan,
to detect possible correlations between gut microbiota and schizo- 2015; Sherwin et al., 2016a, 2016b; Kim and Shin, 2018). Gut micro-
phrenia. We assume that this review may allow identifying new areas biota supports two-way communication with major parts of the CNS
for the study of this severe mental disorder and its treatment. directly and indirectly. Commensal bacteria (indigenous microbiota) in
the gastrointestinal tract communicate with the CNS and manage brain
2. Microbiome neurochemistry and behavior in various ways. These pathways include
the production of bacterial metabolites, such as short-chain fatty acids
Microorganisms have been found throughout the human body, for (SCFA) and immune mediators, such as cytokines, and also signaling to
the most part on the internal and external surfaces, among are the the brain directly by the longest cranial nerve (vagus nerve) (El Aidy
gastrointestinal tract, skin, oral mucosa, saliva, and conjunctiva. et al., 2014; Dinan et al., 2015; Sherwin et al., 2016a, b). The MGB axis
According to estimates by Sender et al., the bacterial content of the is part of a physiological network containing the immune system (cy-
colon transcends all other organs in the human body by at least two tokines, chemokines), the endocrine system (hypothalamic-pituitary-
orders of magnitude. In that context, it is important to note, within the adrenal axis), the autonomic nervous system (ANS), and the enteric
gastrointestinal tract, the colon is the only significant donator to the nervous system (ENS). Gut microbiota is considered to be affecting the
total bacterial population, while the small intestine and stomach are hypothalamic-pituitary-adrenal (HPA) axis and the vagus nerve by
nonessential sources (Sender and Fuchs, 2016). bacterial metabolites in tryptophan metabolism (Kim and Shin, 2018).
The enteric nervous system (ENS) and the central nervous system
2.1. Main functions of the gut microbiota (CNS) are connected by the vagus nerve. The study by Parashar and
Udayabanu has shown that gut microbiota directly in contact with the
The human being cannot digest all the molecules that make up his ENS allows modulating sensory neurons excitability and consequently
diet in his small intestine, he can only produce enzymes to break down information relayed to the brain (Parashar and Udayabanu, 2016).
proteins, starch, and fatty acids into smaller absorbable molecules, such Some studies have reported producing some neurotransmitters by gut
as acids. amines and monosaccharides. Complex proteins and complex microorganisms specifically, that Lactobacillus and Bifidobacterium
carbohydrates, such as fibers and other plant-derived polysaccharides, species can produce GABA (Barrett et al., 2012); the species Escherichia
cannot be broken down by human enzymes because the human genome coli, Bacillus and Saccharomyces produce norepinephrine; Candida,
does not encode their manufacture. It is, therefore, the intestinal mi- Streptococcus, Escherichia and Enterococcus produce serotonin, and
crobiota (Flint et al., 2012) that will digest these almost intact mole- Bacillus and Serratia synthesize dopamine (Lyte, 2011). Recently evi-
cules when they reach the colon. In addition, the ability to degrade dence has shown that gut microbiota affects the serotonergic system
complex carbohydrates can be dictated by the diet of the human po- and regulates the synthesis and secretion serotonin (Kelly et al., 2015a;
pulation. Desbonnet et al., 2015; O’Mahony et al., 2015; Clarke et al., 2013).
The human microbiome plays a major role in the distal intestine Microbiota influence this system by altering tryptophan availability in
because it extracts energy from these otherwise non-digestible food the plasma (Rackers et al., 2018). These neurotransmitters are capable
components (Flint et al., 2012). And many metabolic processes of colon of stimulating enteric cells that can modulate communication between
lumen are dedicated to this task. The reconstitution of the metabolic ENS and CNS.
pathways of the different body sites sampled in the human microbiome Through the gut-brain axis (Rea et al., 2016; Dinan and Cryan,
project (HMP) consortium showed site-specific metabolic profiles 2017a), the gut microbiota can interact with the central nervous system
(Human Microbiome Project Consortium, 2012). Glycosaminoglycan through communication with the host's immune, neuroendocrine and
degradation, which was rare or absent in other body site profiles, neural pathways. For example, during infections, inflammation, and
characterized human intestinal metabolic profiles as determined by autoimmunity on the surface of the intestinal mucosa, modulation of
metagenomic sequencing. Although significant interindividual varia- immune responses by the microbiota of the intestinal tract. Besides, the
tions in microbial species composition can be seen (Lozupone et al., intestinal microbiota may influence microglial cell maturation, mor-
2012), this functionality was remarkably similar in the intestinal spe- phology, and immune function as evidenced by studies of microglia
cimens of all HMP individuals. Some studies show population-specific dysfunction in various preclinical animal models (Rea et al., 2016). The
variations, yet the interindividual metabolic capabilities are overall gut microbiota is thought to modulate a psychiatric illness because
very similar. It is interesting to note, that Bacteroides plebeius strains of many psychiatric disorders are related to inflammation and immune
Japanese subjects have genetic material encoding porphyranases and dysregulation. By direct communication with the neural and endocrine
agarose (Hehemann et al., 2010). The environment can act as a selec- pathways, the microbiota can also influence brain activity, behavior,
tion force on the functional potential of the human microbiota, since and development of mental disorders.
these genes do not exist in other populations and would come from B. Several studies had as a subject of study the relationship between
plebeius by horizontal gene transfer from marine bacteria through the intestinal microbiota with the neuropsychiatric state, the behavioral
consumption. seaweed. traits, and the temperament. Behavioral traits have even been suggested

713
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

to influence the intestinal microbiota. For example, reduced anxiety effects had a long-term impact on the central nervous system (CNS) of
phenotypes and a relatively higher amount in the hippocampus of mice adulthood. Tillisch et al. (2013) have shown in humans that in-
serotonin and its metabolite, 5-hydroxyindolacetic acid (5HIAA), were formation processing of emotional material can be altered by probiotic
found in germ-free (GF) mice compared to colonized mice. Despite the treatment. Messaoudi et al. (2011) found that the values of the 24 -h
anxiety phenotypes had been reversible with colonization after free urinary cortisol test were lower after probiotic treatment. As one
weaning, the rise in serotonin levels in the hippocampus was not ob- study shows, depressed patients would have an increase in the number
served (Clarke et al., 2013). The study by Huo et al. (2017) also showed of Bacteroides, Actinobacteria, Proteobacteria, but a decrease in the
that specific-pathogen-free (SPF) mice with intestinal microorganisms number of Firmicutes (Jiang et al., 2015). Contrary to this experience, a
noted increased anxiety-like behavior under the same pressure compare study by Naseribafrouei et al. (2014) showed that among depressed
to GF mice. Some research previously reported the opposite that GF patients and controls (Naseribafrouei et al., 2014), no difference in
F344 rats were more likely to have anxiety-like behavior than SPF rats microbial composition and diversity was found. Other pre-clinical stu-
(Crumeyrolle-Arias et al., 2014; Wong et al., 2016; Zheng et al., 2016, dies have involved the reversal of noradrenaline abnormalities in a
2017a, 2017b). Desbonnet et al. (2014) found significant social beha- depressive model in mice (Desbonnet et al., 2009), as well as probiotic
vior impairment in GF mice. These behavior shifts were normalized by treatment in the relief of depressive symptoms.
following colonization of the gut of GF mice. A later study by Desbonnet Moreover, bidirectional communication is reported between the
et al. (2015) in mice have shown antibiotic-treatment in adolescence central nervous system and the immune system. Received data by Bilbo
(21 postpartum day onwards) had depleted and reorganized intestinal et al. (2012) confirm the assumption that neonatal immune activation
microbiota composition, which in turn caused reduced anxiety, non- indirectly raises the risk of cognitive deficits, through the programming
spatial cognition deficits and worse performance in the social trans- of neuroimmune responses. Subsequently, that interferes with cognitive
mission of food preference test that had accompanied by modify dy- functions and emotional behavior. That is consistent with later studies
namics of the tryptophan metabolic pathway and significantly de- by Filiano et al. (2017) and Freytag et al. (2017). And these data are in
creasing brain-derived neurotrophic factor (BDNF), oxytocin and good agreement with the “two-hit” hypothesis of schizophrenia, which
vasopressin expression in the brain (Desbonnet et al., 2015). assumes that the combination of vulnerability (likely instantiated at
The other factors associated with behavior alteration are SCFAs younger age) and a later-life (typically young adult) precipitating factor
which are produced by gut microbiota through fermentation of poly- (such as, stress, infection) is required for the inducing the illness (Bilbo
saccharides (Rea et al., 2016; Dinan and Cryan, 2017b). These SCFAs et al., 2012; Choy et al., 2009; Pantelis et al., 2003; Maynard et al.,
are known to have neuroactive properties, to induce neuroinflamma- 2001; Keshavan, 1999; Keshavan and Hogarty, 1999).
tion, to be associated with behavioral alterations. Dietary fiber-derived Norris and Kipnis (2019) in their review have reported the inter-
SCFAs, such as lactic acid, acetic acid, butyric acid, and propionic acid esting suggestion that the immune system may be as a “sensory” arm of
(PPA), influence intestinal epithelial cells (IECs) and immune cells the brain, recognizing peripheral microorganisms and other menace
(Peng et al., 2009; Kelly et al., 2015b; Zheng et al., 2017a, 2017b; Feng and informing the brain about them using the cytokines (Kipnis, 2018),
et al., 2018). SCFAs that enter the circulation and cross the blood-brain as a supplement to the directly interactions through the vagus nerve
barrier (BBB) can directly influence the central nervous system (Chavan et al., 2017; Pavlov and Tracey, 2017).
(Macfarlane and Macfarlane, 2003; Shi et al., 2006; Flierl et al., 2007; Neurons of the central nervous system, microglia, and astrocytes
Kim et al., 2012; Maes et al., 2012). It was showed that PPA, that is a can be modulated by cytokine peripheral signaling (Kohman and
common preservative added to dairy products and refined wheat, ac- Rhodes, 2013). This occurs because of regions with leaks in the BBB,
tivates microglia of the hippocampus, white matter, cingulate, and such as active transport through transport molecules, HPA axis stimu-
neocortex (MacFabe et al., 2011) and can also modulate the balance lation at the anterior pituitary HPA axis or hypothalamus, and re-
between excitation and inhibition in neural circuitry by increased cruitment of activated cells such as monocytes / macrophages from
glutamatergic and decreased GABAergic transmission (MacFabe, 2012). periphery to brain, activation of cells lining the cerebral vasculature
PPA seems to be linked with autism spectrum disorder (ASD) sympto- (endothelial cells and perivascular macrophages), binding to cytokine
matology in humans. Higher autistic symptoms have noticed after receptors associated with the vagus nerve, and circumventricular or-
consuming food containing propionate, and an improvement has been gans (Haroon et al., 2012). It has been discovered that immune cells can
observed after the elimination of this product (Cenit et al., 2017b). communicate with the central nervous system through functional
There is now more and more evidence showing a relationship between lymphatic vessels lining the dural sinuses (Louveau et al., 2015).
certain behavioral traits and psychiatric illnesses, with the intestinal Therefore, neurogenesis, plasticity, and synapse formation can be
microbiota. modulated by peripheral cytokines (Hodes et al., 2015). There is evi-
Also, it seems that the immune and the endocrine system are sig- dence that cytokines can affect cognition and mood (Dowlati et al.,
nificant parts of the synergy between microbiota and the brain. Gut 2010; Udina et al., 2012; Valkanova et al., 2013; Khandaker et al.,
commensal can affect the development and regulation of the hypotha- 2014). Brain regions affected by the administration of inflammatory
lamic-pituitary-adrenal (HPA) axis. Huo and colleagues noticed sig- stimuli include the basal ganglia and the anterior dorsal cortex (anterior
nificant changes in HPA axis hormone levels with increased cortico- cingulate) (DACC), part of the limbic system, involved in cognitive and
trophin-releasing factor (CRF) expression, elevated adrenocorticotropic emotional processing (Harrison et al., 2009; Slavich et al., 2010;
hormone (ACTH), cortisol, and aldosterone levels in GF stressed mice Capuron et al., 2012; Felger and Miller, 2012; Felger et al., 2013; Miller
compared to SPF stressed mice (Huo et al., 2017). Although few human et al., 2013).
studies have been conducted on the gut microbiota and behavior, there It has been shown that an essential function of the gut microbiota is
seems to be a two-way relationship between microbial composition and the priming of the development of the neuroimmune system
stress. An example is Bailey et al. (2011), who found that stress led to a (Chistiakov et al., 2014; Francino, 2014; Olszak et al., 2012; Round and
decrease in the genus Bacteroides and Clostridium (Bailey et al., 2011). Mazmanian, 2009). The luminal surface of the intestine (O’Hara and
Several other studies on the same subject have found that the use of Shanahan, 2006) plays a major role in the process of predisposition to
probiotics can reduce stress-related behaviors (Desbonnet et al., 2009) immune disorders, by changes in the signature of the intestinal mi-
and corticosterone levels due to stress (Gareau et al., 2011). Moya- crobiota at the beginning of immune life (Fujimura et al., 2016; Penders
Pérez et al. (2017b) showed that Bifidobacterium (B. pseudocatenu- et al., 2007). The hygiene hypothesis first proposed in the late 1980s
latum CECT 7765) consumption had beneficially changed the con- (Patel et al., 2017; Strachan, 1989) and reconceptualized as an "old
sequences of maternal separation (MS), had induced chronic stress on friends hypothesis" (Williamson et al., 2015; Rook et al., 2003) sug-
the HPA axis and had down-regulated intestinal inflammation. These gests, that an encounter with less exposure to immunoregulatory

714
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

microorganisms contributes to the increase in chronic inflammatory the innervation of the esophagus, which usually starts after 13 weeks
disorders, as well as stress-related psychiatric disorders (Klerman and (Trahair, 2001). In the third trimester of pregnancy, the human fetus
Weissman, 1989; Guarner et al., 2006; Rook and Lowry, 2008; swallows up the amniotic fluid to 1 L per day (Blakelock et al., 1998;
Turnbaugh et al., 2009; Hidaka, 2012; Rook et al., 2013, 2014; Kostic Gitlin et al., 1972). This makes it possible to introduce bacteria and
et al., 2015; Reber et al., 2016; Stein et al., 2016; Kelly et al., 2017). bacterial products (e.g., RNA and DNA, glycoproteins) into the devel-
Stress leads to excessive inflammation by disrupting homeostasis be- oping gut (Chong et al., 2018).
tween the microbiota and the host. Reber and colleagues showed that Previous studies have ascertained the possibility of the infiltration
repeated immunization of mice with a heat-killed preparation of an of maternal microbes to the amniotic fluid (Collado et al., 2016;
immunoregulatory environmental microorganism (Mycobacterium Jiménez et al., 2008) and the placenta (Epstein et al., 2000). But the
vaccae), averts stress-induced pathology. These data support a strategy transmission of microbes from the mother to the fetus in utero, which
of “reintroducing” humans to their old friends to protect against ne- leads to active colonization and the effect on developmental, remains
gative stress-related outcomes (Reber et al., 2016). unproven (Chong et al., 2018).
In this part of the review, the concept of gut microbiota and its main The developmental trajectory of the gut microbiota is consistent
functions were considered. It has also been shown that the gut micro- with the concepts of early life psychiatry as a vulnerable phase for the
biota has been viewed as a crucial regulator of bidirectional commu- later emergence of psychopathology in adulthood (O’Mahony et al.,
nication between the gut and the brain (gut-brain axis), which implies 2017). At the beginning of life, during the first days, the gut microbiota
the impact in many neurodevelopmental and neurodegenerative dis- is not very diversified and unstable, its composition will evolve during
orders and may affect behavior (de Theije et al., 2011; Cenit et al., the first years of the child to resemble that of an adult when he reaches
2017b). Several studies have shown that the modifications in the the age of 3 years (Voreades et al., 2014). It is becoming increasingly
composition of microorganisms inhabiting the GI tract have linked to popular to study the effect of the mode of delivery and the implications
different neuropsychiatric disorders, including mood disorders, autism for the development of the central nervous system host (Clarke et al.,
spectrum disorder (ASD), Parkinson’s disease (PD), and schizophrenia 2014; Dominguez-Bello et al., 2010; Adlerberth and Wold, 2009). There
(Cenit et al., 2017b). Besides, some studies have shown that many pa- is a difference in bacterial colonization between vaginal and cesarean
tients affected by gastrointestinal problems were more likely to develop delivery. In fact, vaginal delivery infants are colonized by the mother's
mental illness (Mussell et al., 2008; Lee et al., 2016). Thus, dysbiosis of vaginal and faecal bacteria, such as Lactobacilli, while infants born
the gut microbiota may facilitate the pathogenesis of mental health from cesarean delivery are colonized by other bacteria, such as mother's
disorders, confirming the hypothesis of a pathologic process of bidir- skin and environmental sources such as other newborns, medical
ectional communication between the gut and the brain (Huang et al., equipment, medical staff, or even air (Borre et al., 2014). The trajectory
2019). Many factors can have an impact on microbiota composition in of microbiota acquisition may be due to many other factors, such as
early life, including the mode of birth delivery, antibiotic treatment, exposure to family members and domestic animals (Dominguez-Bello
nutritional habits, environmental stressors, and host genetics. For a et al., 2010; Marques et al., 2010; Fujimura et al., 2010; Penders et al.,
better understanding of this aspect and possible risk factors for the 2006), diet (Thum et al., 2012; Koenig et al., 2011), gestational age
development of mental health disorders, this review has considered the (Barrett et al., 2013) or the use of antibiotics (Persaud et al., 2014). It is
development of microbiota and the elements that influence it still unclear what relative importance all these factors have in de-
throughout life. termining the final profile of a stable microbiota.
Evidence from recent studies indicates that prenatal stress will in-
3. Development of the microbiota during the life fluence gut microbiota, with physiological consequences on offspring
(Golubeva et al., 2015). Nevertheless, it is still difficult to draw defi-
3.1. Gut microbiota: development nitive conclusions about the relationship between prenatal stress and
intestinal microbiota from clinical studies.
The confidence that the gut is sterile before birth turned out to be a
mistake. That has been proven by the results of several studies looking 4. Human microbiota during pregnancy
at bacteria, bacterial DNA, or bacterial products in meconium (Nagpal
et al., 2017; Wampach et al., 2017; Jiménez et al., 2008), amniotic fluid 4.1. Gut microbiota
(Collado et al., 2016; DiGiulio et al., 2008), and the placenta (Collado
et al., 2016; Friedrich, 2013). Despite the limited evidence of live Dynamic changes in the composition that accompany changes in
bacterial culture from placental and amniotic fluid samples, these dis- metabolism during pregnancy in the human microbiota (Priyadarshini
coveries raise the ability that intrauterine human gut development is et al., 2014; Koren et al., 2012; Santacruz et al., 2010; Collado et al.,
affected by the development of stage-specific microbiota that begins in 2008) are observed. As gestation progresses, the diversity of the gut
utero (Collado et al., 2016). microbiota, and especially the Actinobacteria and Proteobacteria, in-
Ensuring the influence of microbiota on the development of the creases (Koren et al., 2012). This is why, already at the end of the third
gastrointestinal tract in utero could provide a mechanism that produces trimester, the composition of the intestinal microbiota is significantly
the selection and exposure of the conforming microbial population or different from that at the beginning of pregnancy (Koren et al., 2012).
factors. The environment for such a system is the amniotic fluid that Intestinal microbial diversity has also been shown to increase post-
surrounds the developing fetus. The fact that the composition of the partum in vaginal and vaginal infants (Jakobsson et al., 2014; Koren
amniotic fluid is transformed during pregnancy is noteworthy et al., 2012; Kurokawa et al., 2007). Note that germ-free mice that
(Mennella and Beauchamp, 1998). Amniotic fluid contains mainly fetal received a human microbiota transplant in the third trimester had the
urine, with contributions from secreted lung liquid, buccal secretions, following metabolic syndrome symptoms: inflammation and decreased
and transmembrane flow (Trahair, 2001). And also contains hormones glucose tolerance, and increased adiposity (Koren et al., 2012;
and growth regulators (Bagci et al., 2016), immune modulating pro- Santacruz et al., 2010; Collado et al., 2008). Short-chain fatty acids,
teins, and microbial components (Collado et al., 2016). The pathway for which are essential intestinal microbial metabolites, have been shown
the selection of particular microbes in the amniotic fluid is still unclear. to be associated with metabolic changes during pregnancy and may be
One of the probable way of influencing the developing GI to microbes / partly responsible for the effects mentioned above (Priyadarshini et al.,
microbial products within the amniotic fluid is through swallowing. 2014). It is important to note that neurodevelopment may be affected
The human fetus begins to swallow amniotic fluid 10 weeks after by microbiota modulation during the second and third trimesters, as
conception (Bagci et al., 2016). Which corresponds to the beginning of most changes in the gut microbiota occur at these times.

715
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

4.2. Vaginal microbiota et al., 2018). These results are consistent with Offenbacher et al.
(1998), who first reported the association between maternal period-
The human vaginal microbiome is composed of several types of ontal disease and preterm birth (Escobar-Arregoces et al., 2018). A
stable community states that become destabilized during pregnancy study of the effect of periodontal treatment on the reduction of pro-
(Romero et al., 2014; Ravel et al., 2011). It is most abundant in Lac- inflammatory markers and its association with preterm labor report
tobacillus spp. before pregnancy, but this species may be reduced conflicting data. Offenbacher et al. (2006) report that periodontal
during pregnancy (DiGiulio et al., 2015; MacIntyre et al., 2015). But treatment showed a 3.8-fold reduction in preterm birth rates and a
there are only studies that show a simultaneous reduction in overall decrease in IL1β and IL6. Penova-Veselinovic, found that in the peri-
bacterial diversity during pregnancy (Aagaard et al., 2012), while odontal treatment group there was a significant decrease in the levels of
others indicate the opposite (DiGiulio et al., 2015; MacIntyre et al., IL1β, IL10, IL-12p70 and IL6 compared to the control group. Da Silva
2015). In addition, it was observed that postpartum bacterial diversity et al. (2017) reported a decrease in inflammatory biomarkers in gin-
has increased (MacIntyre et al., 2015). Among different ethnic groups, gival crevicular fluid and serum after non-surgical periodontal therapy
there are differences in the composition of the vaginal microbiome in pregnant women. Despite this, the treatment did not affect the level
during pregnancy and postpartum, indicating that host genetics, the of inflammatory biomarkers in umbilical cord blood and without re-
environment, and bacterial colonization interact together (MacIntyre ducing the incidence of adverse gestational outcomes.
et al., 2015). The mother's vaginal microbiome, which initially colo- However, studies such as Michalowicz et al. (2009); Fiorini et al.
nizes the infant, is extremely similar to the infant's microbiome, this (2013) and Pirie et al. (2013) report that non-surgical periodontal
corresponds to patterns of vertical transmission across the animal therapy during pregnancy did not lead to a decrease in markers of
kingdom (Funkhouser and Bordenstein, 2013; Dominguez-Bello et al., systemic inflammation. It is worth nothing that, thanks to the devel-
2010; Ley et al., 2006). opment of newer epidemiological methods to assess bias caused by the
truncated outcome, Merchant et al. (2018) re-analyzed data from a
4.3. Oral microbiota study by Michalowicz et al., reporting that treatment of periodontitis
during pregnancy does not significantly change stakes of low birth
An important part of human microbiota is oral microbiota, which weight or preterm birth (Michalowicz et al., 2006). Through the use of
performs an important protective function against the colonization of Merchant and al. the survivor average causal effect (SACE), also called
external bacteria that can hit systematic health. At the same time, the the principal strata effect, to correct potential bias resulting from un-
most extended oral health disease, caries, gingivitis, and periodontitis equal survival of fetuses in the treatment and control group of the
are microorganisms based (Arweiler and Netuschil, 2016). Obstetrics and Periodontal Therapy (OPT) study, differences in risk for
In the study by Dasanayake et al. (2005) noted that throughout preterm birth reached statistical significance and showed a beneficial
pregnancy elevated levels of Actinomyces naeslundii genospecies (gsp) effect of periodontal treatment.
2 and Lactobacillus casei in saliva are respectively negatively and po-
sitively associated with gestational age at delivery and birth weight. 5. Prenatal environment impacts on microbiota
This conclusion matches with the hypothesis that oral organisms in-
fluence the delivery process via the orogenital contact or through op- A real serious contact with the world of microorganisms occurs after
portunity penetration the uterine environment from the bloodborne birth, and in many respects the future health of a person depends on
route. The paradigm that most intra-uterine infections, which are as- how this meeting will happen. There is evidence that the first colonizers
sociated with preterm birth, originate in the lower genital tract and are inhabiting the human body even before his birth. Although con-
ascend into an otherwise sterile uterus is outdated and may need to be troversial, some evidence exists to support the hypothesis of a prenatal
reconsidered (Wassenaar and Panigrahi, 2014). microbiome, contrary to what is generally thought, namely that initial
Actinomyces naeslundii gsp 2 is not considered part of the period- intestinal colonization occurs at birth during vaginal delivery (Perez-
ontitis pathogenic complex (Socransky et al., 1998). Despite this, A. Muñoz et al., 2017; Willyard, 2018). A microbiome of the placenta close
naeslundii gsp 2 may also be regarded as pathogenic through they have to the microbiome of the mouth has been demonstrated morphologi-
been caused periodontal infections in gnotobiotic rodents and have cally and by sequencing, from bacteria isolated in human meconium,
been related with actinomycosis and gingivitis (Ellen, 1982; Slack and feces that form in the fetus before birth, and others (Jiménez et al.,
Gerencser, 1975; Jordan and Hammond, 1972). 2008; Stout et al., 2013; Ardissone et al., 2014; Aagaard et al., 2014).
It has been observed that the reason why infants were born with low The development of the fetus may be influenced by a placental mi-
weight or prematurely would be periodontal disease (as an independent crobiome, even if it is not necessarily transferred to the fetus in utero.
risk). It has been observed that in the homogenized cord (Wright et al., But placental colonization can occur during delivery due to breaks in
1994) and the amniotic fluid of this case of preterm delivery (PTD) with the placental barrier (Willyard, 2018). Although bacteria have been
chorioamnionitis (Dixon et al., 1994), periodontal pathogens are found. isolated from the umbilical cord of neonates born from cesarean um-
In the oral cavity of some women who have given birth prematurely to bilicals (Jiménez et al., 2005), many of these studies have been criti-
low weight infants, high levels of pathogens have been observed cized for lack of adequate controls for contamination, for evidence of
(Offenbacher et al., 2001, 1998). In some studies preterm low birth the presence of bacteria viable rather than searching for bacterial genes
weight (PLBW) has been associated with periodontal disease (Lopez by sequencing or inappropriate molecular approaches to detect the
et al., 2002; Jeffcoat et al., 2001; Offenbacher et al., 2001; Dasanayake, bacterium (Perez-Muñoz et al., 2017). Moreover, there is additional
1998; Offenbacher et al., 1996), however, some other studies have not evidence against the prenatal microbiome, and it comes from the ex-
reached the same conclusions (Davenport et al., 2002; Mitchell-Lewis istence of germ-free mouse models (Perez-Muñoz et al., 2017).
et al., 2001; Offenbacher et al., 1998). Ambiguous data on the re-
lationship between PLBW and Ig level are specific for the serum peri- 5.1. Maternal diet and the microbiota
odontal pathogen. According to Dasanayake et al. (2001) this re-
lationship is positive, while Madianos et al. (2001) claim negative Maternal nutritional status before and during pregnancy will in-
association with PLBW. fluence the outcomes for the mother and the baby. The importance of
Recent studies analyzing the relationship between maternal peri- an adequate diet during fetal life for long-term physical health is well
odontal disease and the risk of preterm delivery have shown that pa- described (Barker, 1998; Harding, 2001). Recent studies of diet in hu-
tients with a high risk of preterm delivery had higher levels of in- mans, in addition to poor cardiovascular health or obesity, have an
flammatory cytokines (Escobar-Arregoces et al., 2018; Latorre Uriza impact on neurocognitive development in humans and rodents (Monk

716
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

et al., 2013). However, some of these studies have been criticized be- 5.3. Maternal stress. Maternal immune activation model
cause inappropriate controls are often selected and may introduce
confounding factors that complicate the interpretation of truly intri- It has been proven that maternal stress is modulated by the HPA axis
guing results (Pellizzon, Ricci, 2018; Almeida-Suhett et al., 2017). If the and that it affects this axis in the offspring. Maternal stress has resulted
mouse was fed a high-fat maternal diet, then there could be a greater in children's altered resilience of different strains of rodents (Hiroi
increase in the number of bacteria in the mother's gut microbiome et al., 2016; Lee et al., 2016; Golubeva et al., 2015; Rana et al., 2015;
during pregnancy, but also differences in the composition's change from Bale, 2015), increased anxiety, increased serum corticosterone levels,
animals receiving a control regime (Gohir et al., 2015). The composi- and increased social unrest. Maternal stress can also alter the intestinal
tion of the intestinal metabolome and microbiota in the macaque, and vaginal microbiota during pregnancy, disrupt glucose metabolism
mouse, and rat has been altered by prenatal exposure and adolescence in mice (Jašarević et al., 2017), and reduce the diversity of maternal
to a high-fat diet (Oberbach et al., 2017; Buffington et al., 2016; Gohir intestinal microbiota. The vaginal microbiota-related changes in preg-
et al., 2015; Ma et al., 2014). It has also been shown that the compo- nancy-related mice were interrupted by a variety of prenatal stress, and
sition of the gut microbiota can be modified by a western prenatal diet, this stress also had an impact on the protein content of the vaginal
which results in an increase in the Firmicutes / Bacteroides ratio as well mucosa, which could possibly have contributed to an alteration of the
as gender differences in the expression of the genes of the colon vaginal mucosa. the abundance of Firmicutes and Bacteroidetes in the
(Steegenga et al., 2017). In addition, unhealthy diets in mice also lead intestinal microbiota of their offspring (Jašarević et al., 2015). In the
to social deficits (Buffington et al., 2016; Graf et al., 2016), change in case of humans, persistent maternal stress (including prenatal and
hypothalamic stress response (Grissom et al., 2017), inflammation postnatal stress) would lead to mental health problems in adult off-
(Grissom et al., 2017; Du et al., 2012) in offspring, and differences in spring (Betts et al., 2015) and influence the development of the off-
expression sex ratio (Edlow et al., 2016; Graf et al., 2016). spring microbiota during the first 110 days after birth (Zijlmans et al.,
Researches about nutrients that affect microbiota have shown that 2015).
some of them can have a positive influence. In particular, the study in The maternal immune activation (MIA) model is a controversial
mice by Patterson and colleagues, have revealed that a high-fat diet model in which maternal infections during pregnancy could alter psy-
supplemented with omega-3 polyunsaturated fatty acids increased the chiatric outcomes in children (Estes and McAllister, 2016). MIA mod-
diversity of the microbiota and significantly raised Bifidobacterium at eling in mice commonly administers a polyinosinic mimetic viral
the genus level (Patterson et al., 2014). Gibson et al. have noted that polycytidylic acid or bacterial lipopolysaccharide to produce psychia-
prebiotics help the growth of beneficial gut bacteria and that they in- tric endophenotypes in offspring. Specifically, viral mimetic poly-
clude nondigestible fibers that are fermented by bacteria in the colon inosinic polycytidylic acid administered in embryonic day 12.5 mice
which produce short-chain fatty acids and play an important role in the changed the composition of the gut microbiota and increased intestinal
body's functions (Gibson et al., 2017). Concerning the probiotics that loss by reducing claudin expression while also increasing intestinal
are beneficial strains of bacteria, they bring health benefits to the host cytokine levels, including including interleukin 6, in offspring (Hsiao
and their administration during pregnancy in humans may reduce the et al., 2013). This model also improved bacterial production of 4-
risk of atopy but not other immune-related diseases such as asthma ethylphenylsulfate, resulting in anxiety-like symptoms in wild-type
(Elazab et al., 2013; Azad et al., 2013a). A study in rats by Hsu and mice (Hsiao et al., 2013). But there is a heterogeneity in the time of
colleagues, reports that maternal gut microbiota-targeted therapies administration of polyinosinic viral mimetic polycytidylic acid, which
(with probiotic Lactobacillus casei or prebiotic inulin) could be repro- can lead to the development of different biomarkers or behaviors
gramming strategies to protect from the development of hypertension common to different disorders, including schizophrenia (Juckel et al.,
caused by maternal fructose-rich diet (Hsu et al., 2018). 2011; Li et al., 2009) and spectrum disorders of autism (Malkova et al.,
2012). Bacterial mimetic lipopolysaccharide can also induce behavioral
phenotypes for depression, for anxiety, for alterations in hippocampal
5.2. Drugs and the microbiota development and neurogenesis (Escobar et al., 2011; Romero et al.,
2007), for an increase in postnatal inflammation (Oskvig et al., 2012),
Some medications can have a direct effect on the composition of the and for autism spectrum disorders in offspring (Depino, 2015; Oskvig
microbiota. The most evident class of medicine to affect the microbiota, et al., 2012) although there is currently no study on its impact on the
are antibiotics that have a well-described impact on the gut microbiota microbiota. In mice, persistent fetal intestinal lesions in adulthood, as
with long-standing consequences for the host (Davey et al., 2013; de well as placental lesions, are the result of maternal inflammation in-
Theije et al., 2014; Keeney et al., 2014; Degroote et al., 2016; Langdon duced by lipopolysaccharide (Fricke et al., 2018).
et al., 2016; Le Bastard et al., 2018). But non-antibiotic prescription
drugs have a significant influence on the overall structure of the gut 6. Cesarean section, neurodevelopment, and health
microbiota too (Le Bastard et al., 2018). Despite all this, only a few
studies describe a correlation between the microbiota and the prenatal Now we will consider in more detail how the processes of coloni-
effect of drugs, and below are given some examples. For example, data zation and the formation of the immune response occur during and
by Kuperman and Koren have revealed that antibiotics (which are the after birth. We will focus on the C-section since it has been shown that
most commonly prescribed drugs during pregnancy) reduce the di- delivery by cesarean section can have an immediate impact on psy-
versity and bacterial burden of the microbiome (Kuperman and Koren, chological health in future life. There are now a large number of studies
2016). Besides, negative effects of different classes of antidepressants, showing a marked effect of mode of delivery on the gut microbiome
including selective serotonin (Munoz-Bellido et al., 2000) inhibitors, (Jakobsson et al., 2014; Dominguez-Bello et al., 2010; Mueller et al.,
ketamine (Yang et al., 2017), and tricyclic antidepressants (Csiszar and 2015; Biasucci et al., 2008; Bäckhed et al., 2015; Madan et al., 2016;
Molnar, 1992), can be seen in the growth of bacteria. That may matter, Azad et al., 2013b; Salminen et al., 2004; Brumbaugh et al., 2016;
given that the depression is common during pregnancy, about 14–23% Dogra et al., 2015; Grześkowiak et al., 2015; Martin et al., 2016; Hill
of pregnant women have symptoms of depression (Yonkers et al., 2009; et al., 2017; Tun et al., 2018), although some studies have found less of
Jimenez-Solem, 2014), and the antidepressants use during pregnancy an influence than others (Chu et al., 2017). Infants born via cesarean
has increased over the past 20 years (Cooper et al., 2007; Wichman section (CS) had a gut microbiota more similar to the maternal skin
et al., 2008). microbiota than the vagina (Jakobsson et al., 2014; Dominguez-Bello
et al., 2010; Mueller et al., 2015; Biasucci et al., 2008), delayed Bac-
teroides colonization (Mueller et al., 2015; Biasucci et al., 2008),

717
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

delayed Lactobacillus colonization (Dominguez-Bello et al., 2010), disease than naturally born newborns, conforming to the data from
lower circulating chemokines (Jakobsson et al., 2014), and a higher risk LISA study by Negele et al. (2004).
of vertical obesity transmission from their mother (Tun et al., 2018). As reported by the conclusions drawn from a systematic review,
Despite the fact that recovery of the microbiota in C-section infants Rutayisire et al. (2016), cesarean section is associated with a lower
using a vaginal swab has been tested, the long-term consequences of abundance of Actinobacteria and Bacteroidetes phyla, Bifidobacterium
such an intervention have yet to be studied (Dominguez-Bello et al., and Bacteroides, the most affected genera, and lower microbial di-
2016). versity. A recent study (Liu et al., 2019) noticed that the compared with
The relative contribution of such disorders to brain health is less the vaginally delivered and cesarean delivered infants was character-
clear, although epidemiology and animal researches are beginning to ized by a decrease in phylum Actinobacteria, class Actinobacteria, order
reveal some clear links (O’Neill et al., 2016; Fond et al., 2017; Moya- Bifidobacteria, and family Bifidobacteriaceae, as well as an increase in
Pérez et al., 2017; Martinez et al., 2017; Curran et al., 2017). phylum Firmicutes, classes Negativicutes and Clostridia, orders Sele-
The biochemical landscape in the maternal body can be completely nomonadales and Clostridiales, and family Lactobacillaceae. Four
different if mothers give birth by cesarean section and if there is no genera were significantly different: Bifidobacterium was enriched in the
labor. In fact, the labor is responsible for a change in contractile and group vaginally delivered, and Lactobacillus, Veillonella, and Klebsiella
endocrine inflammatory factors. The creation of the neonatal micro- were enriched in the group cesarean delivered. It was concluded by a
biome and the change of maternal microbiome could be affected by systematic review of Rutayisire et al. (2016) that cesarean section was
these modifications. In most cases, fetal membranes break during labor, associated with a lower abundance of Actinobacteria and Bacteroidetes
exposing the fetus to maternal vaginal bacteria (Stinson et al., 2018). phyla, Bifidobacterium and Bacteroides (being the most affected
The initial environment is shaped by the pioneer species present in the genera), as well as lower microbial diversity. Moreover, these authors,
gut of the newborn. This influences the immune cascades and the dy- in good agreement with Milani et al. (2017), reported that the Firmi-
namic succession of microbes. In the intestine, the microbial commu- cutes phylum, mainly represented by Clostridium and Lactobacillus,
nity is modulated by the pH gradient, digestive processes, gases, and was increased more in CS infants than in neonates with vaginally de-
nutrients. These attributes also see their characteristics influenced livered (VD) from birth to the third month of life. It is observed that
(McKeen et al., 2019). So, in utero, a microbiota resembling an oral Lactobacillus is very abundant in the vagina of the mother and specific
microbiota present in the mother will then begin to colonize the in- to it (Chu et al., 2017). The extent of infant acquisition of Lactobacillus
testine of the newborn, for example, low-abundance commensal bac- in the gastrointestinal tract is a good example of the effect of birth mode
teria such as Escherichia Coli, Prevotella, and Neisseria (Aagaard et al., on the gut microbiota. As reported by Nagpal et al. (2016), unlike in-
2014). However, the delivery mode is considered to be the first con- fants born in the vagina, who had higher detection rates of Lactobacilli
firmed major event that allows the microbiome to be seeded in infants in the intestinal tract throughout the first six months, cesarean infants
with sustainable colonizers (Bäckhed et al., 2015). persisted during this period to have a low detection rate in Lactobacilli.
The bacteria delivered vaginally are mainly colonized by However, by the age of three, this difference in Lactobacilli detection
Bacteroides, Escherichia/Shigella, Parabacteroides, and rates has disappeared. These data prove that colonization of the gut
Bifidobacterium, among which several are obligatory anaerobes. C- microbiota can begin already before birth, that is to say in utero. Be-
section infants are fortified with Staphylococcus, Streptococcus, sides, relatively different proportions of facultative and obligatory
Veilonella, Haemophilus, and Enterobacter, which are associated with anaerobes in the meconium of CS babies and delayed/inferior coloni-
environmental, oral and cutaneous species (Bäckhed et al., 2015), with zation of Lactobacilli indicate that from the first day of life, the ele-
much of it being aerobic. In the first year of life in infants, differences in ments of intestinal dysbiosis associated with the mode of delivery can
gene content (i.e., the metagenome), and the structure of the microbial begin to accumulate (Nagpal et al., 2016). Birth mode also affects
community decrease between those delivered vaginally and those de- bacterial levels in the Bacteroides and Clostridium genera in the in-
livered by C-section. However, observing until the age of 2, differences dividual's microbiota (Jakobsson et al., 2014; Penders et al., 2006;
in innate and adaptive immunity remain detectable. If these infants Wampach et al., 2017; Martin et al., 2016; Bäckhed et al., 2015; Fallani
were delivered by C-section, lower levels of IgM, IgA, and IgG secreting et al., 2010; Biasucci et al., 2010). As reported by a study of 24 infants
cells were found, indicating a reduced adaptive immune response, a in south-east Sweden, at the phylum, if neonates are born by cesarean
lower CD4 + T lymphocyte response, and lower levels of chemokines section and not born vaginally, then we find that low abundance of
supporting Th1, IFNy and IL-8 (Amenyogbe et al., 2017). Bacteroidetes (Jakobsson et al., 2014). It has been observed that this
The pioneer facultative anaerobic species with metabolic flexibility phenomenon persists even after the first 2 years of birth (Jakobsson
in the presence of oxygen, modify the environment in the first weeks of et al., 2014). This is consistent with previous reports that report late
life in favor of obligate anaerobic species through the use of oxygen to establishment in the first six months of Bacteroides group members in
create a more anaerobic environment (Penders et al., 2006), and by cesarean infants (Martin et al., 2016) and one year of life (Adlerberth
reducing the substrates of light by redox-dependent (oxygen)-depen- et al., 2006).
dent genetic pathways that produce metabolites, such as acetate, which The link between development, gastrointestinal microbiota trans-
may be obligatory and stimulate anaerobes (Appleman and Wormser, mission and birth mode (vaginal or cesarean section) has not been
2005). The meconium of newborns is rich in facultative anaerobes such demonstrated by all studies. For example, according to one study,
as E. coli, but the fecal microbiota becomes more diversified with the during the first 3 months after birth, 21 births were not affected by the
appearance of obligate anaerobic such as Bifidobacterium and Clos- birth mode (Arboleya et al., 2012). In this study, this could be explained
tridium from the first week (Jiménez et al., 2008). by the incorporation of premature infants (gestational age 30–35
For infants born from a C-section, recent research shows that if there weeks), since another study of premature infants also revealed that the
was a period of work before the operation, then the microbiota of in- mode of delivery was not associated with significantly to the compo-
fants resembles that of infants born in the vagina, and if not, the mi- sition of the microbiome (Stewart et al., 2017). In summary, studies
crobiota resembles that of the maternal skin (Chu et al., 2017). Early indicate that if infants are born by cesarean section, then they tend to
microbial disruption is due in part to cesarean section, and during in- have: a less diverse microbiota (Martin et al., 2016; Jakobsson et al.,
fancy this disruption of microbial colonization affects the microbial- 2014; Biasucci et al., 2008), a lower number of anaerobes (e.g., Bac-
host interaction, leading to long-term metabolic consequences in the teroidetes), that they contract atopic diseases (Biasucci et al., 2008) and
host (Dominguez-Bello et al., 2016; Cox et al., 2014; Dominguez-Bello metabolic disorders (Dominguez-Bello et al., 2016) more often than
et al., 2010). Besides, it has been shown that during the first two years children born from unassisted vaginal delivery, and there is delayed
after birth, neonates of cesareans will be more likely to contract atopic colonization of the microbial population. However, these studies are

718
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

complicated by the differences in analytical methodology and ethnic prepared to face future insults if there has been birth stress due to va-
and geographical diversity (Chong et al., 2018). ginal birth (Cho and Norman, 2013).
Chu et al. studied the potential metabolic function of neonatal and The fact that the elective CS is normally programmed between 37
early infant microbiota and taxonomic composition in several body and 39 weeks of gestation (as opposed to complete 40-week gestation to
sites (vaginal introitus, oral cavity, nostrils, skin, posterior fornix, stool) avoid spontaneous labor) may be another possible explanation for the
and concluded that the infant microbiota undergoes significant re- impact of childbirth on psychological development (Tita et al., 2009).
organization the first 6 weeks of life, mainly motivated by the body site The last few weeks may be important for brain development. Therefore,
and not by the mode of delivery (Chu et al., 2017). At the age of 6 and not being born at term may lead to an increased risk of psychological
12 months, Rutayisire et al. (2016) found no differences in the micro- problems (Curran et al., 2015). A study done in 2010 shows that a birth
biota because of the mode of delivery, this was also observed by other between 37 and 39 weeks or early-term birth implied a need for special
authors, even up to the age of 7 years (However differences at this age education (Mackay et al., 2013). In addition, the gut microbiota affects
were less pronounced than in neonates) (Milani et al., 2017; Salminen brain physiology through regulation of microglia development, ma-
et al., 2004). Different confounding factors could be the partial cause of turation and function of microglia, synaptogenesis, neurotransmitters
the differences in the results obtained by different authors, but also regulation and regulation of neurotrophic factors such as BDNF (Abdel-
these divergences can be explained by confusions that are not always Haq et al., 2019; Erny et al., 2015; Stilling et al., 2015; Clarke et al.,
correctly identified, for example, factors related to experimental tech- 2013; McVey Neufeld et al., 2011; Heijtz et al., 2011; Sudo et al., 2004).
niques used (dependent and independent culture techniques, DNA ex- In spite of everything, any association may be motivated by factors
traction methods, etc.), neonatal exposures (such as intrapartum anti- independently associated with both psychological development and CS
biotic prophylaxis), or racial or geographical differences (Arboleya and not only CS itself.
et al., 2018). Cerebral physiology could be affected, according to researches on
It is important to know that in terms of the risk of childhood and the main mechanisms of neuropsychiatric disorders, by the gut micro-
maternal illness, the abuse of C-section is not without risk. It has been biota (Zhuang et al., 2019). Brain physiology affects behavior through
shown that, unlike women who give birth by VD, women with CS are the pathways of gut–brain connection (neural and humoral), thus
more likely to develop a urinary tract infection (Gundersen et al., showing that the microbial community in the intestine is an important
2018). Subcutaneous delivery is the main risk factor for maternal element (Cryan and Dinan, 2015). Also, other studies have shown that
postpartum infection (Smaill and Grivell, 2014). Because of antibiotics gene expression associated with learning, memory, neuronal plasticity,
introducing antimicrobial agents during the perinatal period to reduce neurogenesis and disorders such as schizophrenia, cognitive dysfunc-
the risk of infection for these women, this poses an additional risk for tion, depression, could be influenced by changes at the level of the host
the development of the infant and maternal microbiota. Besides, the epigenome because of gut microbiota activity (Majnik and Lane, 2015;
differences between CS and VD babies are not limited to the gut mi- Stilling et al., 2014). Conforming to recent studies for the prevention of
crobiota; Infants born after CS have been found to have reduced levels neurodevelopmental and neurodegenerative disorders, the intestinal
of immune mediators such as cytokines (Malamitsi-Puchner et al., microbiota is essential for maintaining the healthy functional condition
2005). As a result, there were unsurprising reports that CS had both of microglia (Erny et al., 2015; Harry, 2013). As reported by some
short- and long-term consequences for infants, and that in comparison studies, the intestinal microbiota of children with ASD presents dif-
with VD it was associated with a very poor health outcome. As can be ferent shares of Firmicutes and Bacteroidetes (Tomova et al., 2015;
seen in the Apgar scores (measuring neonatal health), the differences Finegold et al., 2010). Besides, attention-deficit /hyperactivity disorder
between CS and VD babies are already observable from the first mo- (ADHD) and schizophrenia have also been evaluated to be related to gut
ments of life (Costa-Ramón et al., 2018). Subsequently, a higher risk of microbes (Cenit et al., 2017a; Dinan et al., 2014). With a study by
developing celiac disease (Decker et al., 2010), allergic diseases Rutayisire et al. (2017), it was shown that cesarean section had a real
(Wopereis et al., 2014; Bager et al., 2008; Thavagnanam et al., 2008), impact on the total scores of the Strengths and Difficulty Questionnaire
obesity (Mueller et al., 2017; Pei et al., 2014), hypertension in young (SDQ), which corroborates previous studies reporting CS associations
adults (Horta et al., 2013), and type 1 diabetes (Cardwell et al., 2008), with ASD (Curran et al., 2015) and ADHD (Silva et al., 2014). However,
was observed in subjects delivered by CS. no such association had been shown by two previous studies. Al Khalaf
Some epidemiological studies show that cesarean delivery results in et al. (2015) noted no significant association between total SDQ scores
a slight increase in some neuropsychiatric disorders, such as autism and CS in three-year-old children and Curran et al. (2016) detected no
spectrum disorders (ASD), attention deficit hyperactivity disorder and association between abnormal SDQ scores and CS in seven-year-old
bipolar disorder (Curran et al., 2015; Chudal et al., 2014). But other children. The results of Rutayisire et al. (2017) for the SDQ subscales
definitive studies have not found links with attention deficit/ hyper- indicate that cesarean delivery is not associated with hyperactivity,
activity disorder, autism or psychosis (Curran et al., 2016; O’Neill et al., emotional problems or conduct problems, but with behavioral problems
2016). Recently, even after controlling for common family confounders in childhood. Exposure to microbiota at early-life is one of the alleged
and measured covariates, a higher risk of obsessive-compulsive disorder biological mechanism that links the development of behavioral pro-
was associated with a variety of perinatal risk factors, including the blems and CS. It has been suggested that the child development and risk
mode of delivery for birth (Brander et al., 2016). of unfavorable behavior may be due to postpartum complications in his
Regarding psychological development, there are several hypotheses mother after cesarean section. Concerning psychological impact factors,
showing a potential connection with CS childbirth (Curran et al., 2015). the appearance of negative attitude and behavior in the mother could
With cesarean section, brain disorders later in life, as well as neuro- have effects on peer and behavior problems of the child. And for chil-
developmental disorders could appear due to changes in the normal dren born via cesarean section, this could be the explanation for the
developmental trajectories of the microbiota-brain-gut axis as well as increase in the rate of neurobehavioral problems (Rutayisire et al.,
the disruption of its normal maturing (McVey Neufeld et al., 2016; 2017). The study by Polidano et al. (2017) suggested that obesity, at-
Sampson and Mazmanian, 2015; Borre et al., 2014; Foster and McVey tention deficit disorder (ADD), and breastfeeding were important
Neufeld, 2013; Collins et al., 2012; Cryan and Dinan, 2012; Dinan and mediators for the relationship between child cognitive outcomes and of
Cryan, 2012; Rhee et al., 2009). Exposure to a bacterium at birth is a cesarean delivery. By the 1 st stage of the mediating analysis of this
critical event in establishing a stable intestinal microbiota, which is study, it was also shown that higher rates of obesity, lower rates of
altered when infants are delivered by cesarean section (Moya-Pérez breastfeeding, and higher rates of ADD are associated with a cesarean
et al., 2017). We understand more and more that the immune system birth. According to the 2nd stage, higher cognitive performance is
and the hypothalamic-pituitary-adrenal (HPA) axis will be better significantly associated with breastfeeding, whereas ASD, ADD and

719
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

obesity are significantly associated with lower cognitive performance concern premature infants, and to the impacts of newborns’ health fu-
(Polidano et al., 2017). ture. Treatment with enteral probiotics, prebiotics, and synbiotics
during postnatal life seems to be a possible way of transforming the
7. Postnatal environment impacts on microbiota infant's microbiome. Intestinal colonization by beneficial bacteria is
important for the establishment and maintenance of the mucosal bar-
The microbiome and metabolic reactions of the human infant gut rier, therefore protecting the newborn from enteric pathogens and in-
are distinct from those of the adult gut. Colonization of the baby’s in- flammation (Sohn and Underwood, 2017). Studies of the use of com-
testine begins immediately after birth, in a process that is believed to positions for bottle-feeding, with the addition of oligosaccharides,
include the initial seeding with vaginal and skin microbes obtained indicate the achievement of a prebiotic effect (Fanaro et al., 2005; Rao
from the mother, which are gradually replaced during the first months et al., 2009; Srinivasjois et al., 2013), which confirms their effectiveness
of life with strains obtained from other sources with large shifts com- comparable to that of breastfeeding. The international study including
positions during weaning or antibiotic treatment (Mueller et al., 2015). 440 healthy on time born babies had shown that the formula with a
Exposure to antibiotics within the first 3 years of life in humans de- combination of neutral oligosaccharides and pectin-derived acidic oli-
creased microbiome stability and diversity while it transiently in- gosaccharides is effective as prime prevention in low developing atopic
creased transcription of antibiotic-resistant genes (Yassour et al., 2016) dermatitis risk infants (Grüber et al., 2010). As for probiotics, oral in-
and increased adiposity in male individuals during childhood (Azad troduced probiotic strains are not able to stay in the intestines for a long
et al., 2014). time, since the composition of the indigenous (constant) microbiota is
largely determined genetically and is based on subtle immune inter-
7.1. Postnatal diet and the microbiota actions with a host (Methé et al., 2012; Huttenhower et al., 2012).
However, the positive effects of probiotics have been proven (Rijkers
Multiple investigations have determined that nutrition and medi- et al., 2010), including mental health. Studies by Huang et al. (2016)
cation use are the main factors affecting gut microbiota diversity (Zeevi report a reduced risk of depression and studies by Pärtty et al. (2015)
et al., 2015; Falony et al., 2016; Goodrich et al., 2016; Turpin et al., and Gilbert et al. (2013) indicate a reduction in the risk of autism.
2016; Wang et al., 2016; Zhernakova et al., 2016; Jackson et al., 2018; When breastfeeding is stopped and solid foods are introduced, the
Rothschild et al., 2018). The important factor in the establishment of infant's intestinal microbiota begins to change in the direction of a more
commensal microbiota of the newborn is the nature of nutrition. Breast adult composition characterized by an increase in the number of
milk is an optimally suited nutrition for infants, ensuring its optimal Bacteroides, Clostridium, Faecalibacterium and Ruminococcus
development (Butte et al., 2002). Human milk is very rich in lactose, (Bäckhed et al., 2015; Mueller et al., 2015), the composition of the
fatty acids and hundreds of different types of oligosaccharides con- intestinal microbiota in children continues to resemble more and more
sisting of different combinations of sugar moieties bonded through a the composition of adults until it reaches maturity at the age of 3–4
variety of glycosidic bonds, some of which are sialylated (Smilowitz years (Yatsunenko et al., 2012).
et al., 2014). Oligosaccharide glycoside of milk and other bonds cannot Besides nutrition, lifestyle may also affect the intestinal microbiota
be lysed by enzymes that encode the human genome, like dietary fiber composition and diversity. A recent investigation by Mitchell and col-
in the gastrointestinal tract of an adult, and the infant relies on bacteria leagues has described that doing exercise was linked with modifications
to digest these compounds. It is believed that the microbes necessary for in gut microbial composition and with an increase in butyrate produ-
their digestion are transmitted from mother to baby vertically through cing bacteria regardless of dietary in rodents and humans. However, the
milk (Mueller et al., 2015). These bacteria, in particular Bifido- overall quality of evidence in the studies in humans was low (Mitchell
bacterium infantis, Bacteroides thetaiotaomicron, and Bacteroides fra- et al., 2019).
gilis, are present in the intestinal microbiota of most children who are
exclusively breastfed in the first months of life (Yatsunenko et al., 2012; 7.2. Postnatal stress and the microbiota
Bäckhed et al., 2015). The genomes of these species are well equipped
for cleavage of oligosaccharides present in human milk, encoding sev- Events in early childhood, both positive and negative, have a pro-
eral receptors, intracellular and extracellular enzymes that can digest gramming effect on the subsequent development of the individual, on
many of the sugar components of human milk oligosaccharides (Sela its behavior, physiological and neurobiological functions (Ladd et al.,
et al., 2011). 2000; Bondar and Merkulova, 2016; Silberman et al., 2016). Clinical
Under the postnatal period, many women suffer from acute and studies have shown that stress in early childhood in humans leads to a
chronic health conditions like cough/colds, infections, mastitis, in- decrease in cognitive functions and emotional disturbances that can
testines problems, headache, migraine, back pain, hypertension, de- persist throughout life (Gershon et al., 2013; Pesonen et al., 2013).
pression (Glazener et al., 1995; Brown and Lumley, 1998; Yokoe et al., Studies in rodents also show that stress in early life is associated with
2001; Amir et al., 2007; Declercq et al., 2008; Ahnfeldt-Mollerup et al., behavioral and cognitive abnormalities in adulthood (Sánchez et al.,
2012; Woolhouse et al., 2012), and need medications. Most of the 2001; Pryce and Feldon, 2003; Kosten et al., 2012).
commonly used drugs are compatible with breastfeeding and relatively Early postnatal stress affects the hypothalamic-pituitary-adrenal
safe for nursing infants. The dose obtained by babies through drug axis and promotes to the programming of brain health later in life
excretion into breast milk, is mostly small and much less than the (Heim and Nemeroff, 2001). Different types of early postnatal stress
known medication safe doses used in newborns and infants (Hotham (social isolation and maternal separation) change the intestinal micro-
and Hotham, 2015). Nevertheless, in humans, antidepressants have biota composition and metabolism in rats (Farshim et al., 2016;
been detected to transfer through breast milk, and some can reach a Doherty et al., 2018; O’Mahony et al., 2009) and their inflammatory
clinically significant concentration in the infant’s serum (Sachs and profiles (Doherty et al., 2018; O’Mahony et al., 2009). Social isolation
Drugs, 2013), although their effects on the infant have not yet been also impaired memory and learning in rats (Doherty et al., 2018).
established (Glover and Clinton, 2016). Some pharmaceutical drugs Studies with germ-free mice noticed that were more vulnerable to re-
may cause side effects in breastfed babies (Chaves and Lamounier, straint stress-resulting in higher corticosterone and adrenocorticotropic
2004) or can interfere with the let-down reflex and reduce milk supply hormone in plasma (Sudo et al., 2004; Clarke et al., 2013), a reduction
(Chaves and Lamounier, 2004; U.S. Department of Health & Human in glucocorticoid receptor messenger RNA, and an increased stress re-
Services, 2018). sponse (Sudo et al., 2004). Notably, these effects were rescued with
Dysbiosis of the neonatal intestine makes a significant contribution microbiota transplantation during adolescence but not during adult-
to developing colic in the term infant, to the disease processes which hood (Sudo et al., 2004).

720
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

7.3. Host genetics studies suggest that the disruption of the “microbiota-gut-brain” (MGB)
axis may promote the development of schizophrenia (Zheng et al.,
Diet and medications only explain small part variations of the mi- 2019). Studies in mice by Desbonnet and colleagues have shown the
crobiome in the human populace (Scepanovic et al., 2019). Host ge- microbiota modulates the programming of social behavior and cogni-
netics has also been suggested, as a member of affecting relative gut tion, attributes that are known to be violated in schizophrenia
microbiota abundance (Khachatryan et al., 2008; Goodrich et al., (Desbonnet et al., 2015).
2014). Even so, variations in the microbiome between individuals can A recent study has found that unmedicated and medicated patients
be considerable, the microbiome of each individuals can not stay stable with schizophrenia had a decreased microbiome α-diversity index and
(David et al., 2014; Ding and Schloss, 2014; Oh et al., 2016). That have noticed alterations of gut microbial composition compare to
suggests that the host genetic is one factor maintaining the human healthy controls. Some unique bacterial taxa (such as Veillonellaceae
microbial communities' composition (Kolde et al., 2018). and Lachnospiraceae) were related to the severity of schizophrenia
Host genetics plays a critical role in human diseases, but just re- (Zheng et al., 2019). Clinical studies have allowed for the role of change
cently has science come to understand how the microbiota cooperates in the digestive and digestive microbiota in schizophrenia. For example,
with host genetics. For example, metabolic disorders that have a genetic non-compliance with the regulation of inflammatory processes is in-
component (Herbert et al., 2006; Frayling et al., 2007) are also asso- volved in schizophrenia (Al-Asmari and Khan, 2014; Chase et al., 2015)
ciated with a specific gut microbiota structure (Turnbaugh et al., 2009; and it is expected that the intestinal microbiota modulates immune
Qin et al., 2012; Karlsson et al., 2013), suggesting that impaired host processes (Lopetuso et al., 2014), it is expected that the microbiome-
bacterial regulation is a possible mechanism for origination and de- intestine-brain axis has the ability to act on schizophrenia using im-
velopment of a disease. On the way to the understanding of possible munological devices.
interactions between the risks of developing schizophrenia and the When the oropharyngeal microbiome was compared to patients
composition of the host microbiota, it is important to note that genetics with schizophrenia and healthy people, Castro-Nallar and his collea-
form a strong risk factor for schizophrenia (Henriksen et al., 2017) with gues found that certain types of bacteria significantly predominate in
heritability between 64 and 81 % (Sullivan et al., 2003; Lichtenstein patients suffering from this mental state. In addition, differences in the
et al., 2009). number of species and their distribution were recorded, and they also
observed completely different metabolic pathways. In patients with
8. The microbiome in schizophrenia schizophrenia, lactic acid bacteria and the system of metabolite trans-
port prevailed (Castro-Nallar et al., 2015). But this study provides early
Schizophrenia is a destructive illness, it is one of the top 15 leading confirmations of differences in the oral microbiota between patients
causes of disability worldwide in 2016 (GBD, 2016; Disease and Injury with schizophrenia and the control group; there are no confirmations
Incidence and Prevalence Collaborators, 2017). Previous investigations regarding the connection of the oropharyngeal component with the
have focused on the analysis of the human genome to determine the digestive microbiome, encompassing effects of the former on the gut-
pathogenesis of SCZ (The Schizophrenia Psychiatric GWAS Consortium, brain axis. Yuan et al. (2018) studied microbiota changes in 41 patients
2011). Still, the known interactions are probably estimate just for about with schizophrenia in the first episode after 24 weeks of treatment with
4 % of the variance in schizophrenia. Consequently, for a better un- risperidone. In patients with schizophrenia compared with healthy
derstanding of this disease, affecting approximately 0.3–0.7 % of the controls, there was significantly less fecal Bifidobacterium, Escherichia
general population global (Saha et al., 2005; Moreno-Küstner et al., coli and Lactobacillus. Conversely, a significantly high amount of feces
2018), there should be researches also to recognize the role of non- of Clostridium coccoides. After 24 weeks of treatment with risperidone,
human genetic factors in the inception of schizophrenia (Zheng et al., there was a significant increase in the amount of fecal Bifidobacterium
2019). An epidemiological study has shown that prenatal microbial and E. Coli, and there was a significant decrease in the amount of feces
infection in the early stages of pregnancy, has increased the risk of of Clostridium coccoides and Lactobacillus. The authors concluded that
developing schizophrenia and other schizophrenia spectrum disorders patients with schizophrenia in the first episode of the disease, suffering
by 10–20 times in offspring (Babulas et al., 2006). from schizophrenia, have anomalies in the composition of the micro-
Several studies have investigated the intestinal microbiota in asso- biota, and noted that significant changes in fecal bacteria were due to
ciation to schizophrenia (Clarke et al., 2013; Sudo et al., 2004; treatment with risperidone. In addition, they suggested that these
Christian et al., 2015; Hsiao et al., 2013; Desbonnet et al., 2010; Castro- changes were associated with metabolic changes caused by risperidone.
Nallar et al., 2015; Desbonnet et al., 2015; Bercik et al., 2011; Bailey The differences in fecal microbiota investigated by Schwarz et al.
et al., 2011; Desbonnet et al., 2009; Gareau et al., 2011; Tillisch et al., (2018) between 28 individuals diagnosed with first-episode psychosis
2013; Messaoudi et al., 2011; Jiang et al., 2015; Naseribafrouei et al., (half of whom received a diagnosis of schizophrenia by the study 1-year
2014; Severance et al., 2013; Dickerson et al., 2014; Davey et al., 2012, follow up appraisal) and 16 health controls. They found an increased
2013; Morgan et al., 2014; Bahr et al., 2015a, 2015b). It is difficult to amount of Lactobacillus bacteria in psychotic patients. Interesting to
determine the mechanism by which intestinal microbiota can con- note that, after up to 12 months of treatment, the subgroup of patients
tribute to the development of schizophrenia, given the limitations as- that showed the strongest differences in microbiota coincided with the
sociated with the current lack of an animal model of schizophrenia, subjects who had a lower response (Schwarz et al., 2018).
which would cover all the complexities of the disease. (Al-Asmari and Shen et al. (2018) evaluated the distinction in gut microbiota be-
Khan, 2014; Powell and Miyakawa, 2006). However, as described tween 64 patients with schizophrenia and 53 healthy controls and
earlier, the gut microbiota could modulate brain function and behaviors found in patients with schizophrenia a higher number of Proteo-
through the “microbiota-gut-brain” (MGB) axis. Recent evidence has bacteria, Succinivibrio, Megasphaera, Collinsella, Clostridium, Kleb-
shown that the gut microbiota have been reported to be associated with siella and Methanbrevibacter and a lower number of Blautia, Copro-
alterations in anxiety (Diaz Heijtz et al., 2011), memory (Gareau et al., coccus, Roseburia compared to healthy controls. Interestingly, the
2011), cognition (Desbonnet et al., 2014), and locomotor activity authors noted the possibility of using 12 microbiotas as diagnostic
(Sampson et al., 2016). Nonetheless, some studies have evaded to the factors for distinguishing patients with schizophrenia from the control
role of the gut microbiome in schizophrenia behaviors. In animal stu- group. Yolken et al. (2015) tested the association between genomes
dies, intestinal microbiota have played a key role in postnatal devel- schizophrenia and bacteriophage. 41 persons with schizophrenia and
opment and maturation of neural, immune, and endocrine systems 33 healthy controls have participated in this study. Patients with
(Clarke et al., 2013), and these behavioral and physiological processes schizophrenia had in the oropharynx significantly more common Lac-
are often disturbed in schizophrenia patients (Miller et al., 2011). These tobacillus phage phiadh. The existence of this microorganism also

721
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

correlated with immune disorders and valproate administration in the outcomes by Severance et al. (2017) had shown biological improve-
study group. ment attributable to probiotic treatment and proposed that manipula-
Severance et al. (2013) evaluated serological surrogate markers of tion of the intestinal microbiota with probiotics may regulate immune
bacterial translocation - soluble CD14 (sCD14) and lipopolysaccharide responses and leads us to the conclusion that microbial therapy could
binding proten (LBP). LBP and sCD14 levels were significantly inter- be a potential strategy to improve psychiatric symptoms and relieve
connected, as accord with their coordinated roles in activating the in- gastrointestinal comorbidities.
nate immunity. Both these markers in persons with schizophrenia had
significantly correlated with C-reactive protein (CRP), indicating a 10. Studies about probiotic and prebiotics use, with reference to
common pathway of inflammation. Although coherence these markers, schizophrenia
the study had detected notably elevated sCD14 level in schizophrenia
compared to controls, which was not matched by raise LBP. Con- Considering the latest data on the relationship between intestinal
sidering bacterial translocation might partially promote to inflamma- microbiota and schizophrenia and the role of environmental factors in
tion psychiatric disorder-associated, other forms of innate immunity the development of this disease, it is possible to use probiotics for the
dysregulation proper to the disease may also explain these results. treatment of inflammatory processes of the gastrointestinal tract, with a
Critchley and Harrison (2013) tested the impact of visceral homeostasis positive effect of schizophrenia symptoms. Such treatment with “psy-
on both physiological and mental capacities of the brain and added to chobiotics” (live bacteria, which when ingested in adequate amounts,
the studies showing that microbiota might influence gut-brain axis at confer mental health benefits) could become a breakthrough in the
any age, leading to neurodevelopmental or neurodegenerative condi- management of mental illnesses (Saulnier et al., 2013; Sarkar et al.,
tions (Dinan and Cryan, 2017b). 2016; Deans, 2017). Severance and colleagues explored the possible
Investigations of the fungal composition of the human gut-the relationship between food antigen-associated immune activation in
Mycobiome are also rising (Suhr and Hallen-Adams, 2015). A case- patients with schizophrenia and gastrointestinal inflammation. They
control cohort study with 261 individuals with schizophrenia, 270 with recorded 193 subjects with non-recent and 67 with recent beginning of
bipolar disorder, and 277 non-psychiatric controls has been done. No schizophrenia, while there were 207 persons in the control group. They
differences were found when analyzing Candida albicans exposure at uncovered food antigen antibodies and gastrointestinal inflammation in
the group level. However, there has been an increase in the likelihood both schizophrenia groups (Severance et al., 2012).
of schizophrenia in men when conducting statistical distribution by In a study published in 2015, the same authors explored the link
gender (Severance et al., 2016a). The same group conducted a rando- between dietary agents (wheat gluten and bovine milk casein) and
mized, double-blind, placebo-controlled, 14-week period probiotic trial, immune response in blood and sample of cerebrospinal fluid (CSF) in
and demonstrated that probiotic treatment significantly reduced C. al- 105 patients with first episode of schizophrenia and 61 persons in the
bicans antibodies in males only, and a trend toward improvement in control group. In the experimental group of schizophrenic patients, the
positive psychiatric symptoms in seronegative males (Severance et al., levels of IgG response to dietary proteins were significantly higher in
2017). Both groups had antipsychotic treatment, but antipsychotic both serum and CSF (Severance et al., 2015). Preliminary yet inter-
medication regimes were not different between probiotic and placebo esting information is emerging from clinical trials with probiotics in the
groups. treatment of schizophrenia (Bruce-Keller et al., 2018). Microbiome
transplants from donor mice fed with high-fat diet showed that high fat-
9. The association between schizophrenia and immune shaped microbiota disrupted cognitive, exploratory, and stereotypical/
inflammatory response impulsive behaviors (Bruce-Keller et al., 2015). Other studies involving
animal models demonstrated that probiotics may improve cognition,
Several clinical studies have investigated blood-based biomarkers of mood, anxiety, while improving neural activity and signaling (Sudo
microbial translocation (Castro-Nallar et al., 2015; Yolken et al., 2015), et al., 2004; Desbonnet et al., 2010; Bravo et al., 2011; Smith et al.,
Severance et al. (2012, 2013, 2016, 2017)). Patients with schizophrenia 2014; Bruce-Keller et al., 2018). Also, mice studies have shown the
exhibited higher serum antibody levels to fungal pathogens Sacchar- ability of probiotics to promote hypothalamic synaptic plasticity and
omyces cerevisiae and Candida albicans (Severance et al., 2012, 2016) prevent decreases in hippocampal neurogenesis induced by stress (Ait-
and soluble CD14 (Severance et al., 2013), a protein marker of bacterial Belgnaoui et al., 2014). Dietary trans and saturated fats, may increase
translocation. Studies of the effects of psychotropic drugs on these intestinal inflammation (Deopurkar et al., 2010; Okada et al., 2013),
biomarkers showed that levels of antibodies to Saccharomyces cerevi- which results in a decrease of commensal Bacteroidetes and increase of
siae were higher in patients with psychosis of the first episode who did pathogenic Enterobacteriaceae and Proteobacteria (Lupp et al., 2007;
not receive antipsychotic treatment, compared with patients receiving Stecher et al., 2007; Pédron and Sansonetti, 2008).
antipsychotics (Severance et al., 2012). Otherwise, antibodies to C. al- Karakuła-Juchnowicz et al. (2016) reviewed the role of the food
bicans, sCD14, and LBP did not differ in patients who take various types antigens in schizophrenia, the use of diet modification, as well as an-
of psychiatric medicines (including lithium, valproate or anti- tibiotics and probiotics as the possible treatment solutions. Probiotics
psychotics) (Severance et al., 2016, 2013). are microorganisms, usually Lactobacilli and/or Bifidobacteria
The increased level of these serological biomarkers suggests in- (Messaoudi, 2011; Tillisch et al., 2013; Steenbergen, 2015; Sarkar et al.,
creased permeability of the intestinal lumen or “leaky gut” through the 2016). Prebiotics are nondigestible carbohydrates that increase bene-
process of microbial translocation, and denotes inflammation of the ficial microbiota. Prebiotics may improve emotional affect and mod-
intestine (Dickerson et al., 2017). Hsiao et al. (2013) in a preclinical ulate stress responses (Schmidt et al., 2015). Randomized trials have
model of autism detected that microbial dysbiosis led to intestinal shown the efficacy of probiotics on mood (Messaoudi, 2011;
permeability and elevated cytokine levels. Thereby, the gut may be- Steenbergen, 2015) as well as the ability to reduce responses to stress
come a cause of autointoxication. Emaciation or dysbiosis of microbes (Kato-Kataoka, 2016). However, other studies have produced con-
that promote development of the immune system may be at the reason troversial results and therefore more trials are needed to completely
of a chronic inflammatory state in severe mental illnesses (SMI), which demonstrate efficacy, to identify the specific strains that are most
can affect the function of the central nervous system (Dickerson et al., beneficial, as well as the correct dose and treatment duration (Doron
2007; Frodl and Amico, 2014; Pedersen et al., 2008). Actually, the and Snydman, 2015; Bruce-Keller et al., 2018). Similarly, large, con-
memory performance was significantly reduced in women with schi- trolled and well-powered studies about the efficacy of prebiotics are
zophrenia who were seropositive for C. albicans, compared to their warranted (Bruce-Keller et al., 2018).
respective seronegative counterparts (Severance et al., 2016). The Tomasik et al. studied probiotic in schizophrenia and found their

722
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

significant impact in reducing von Willebrand factor (VWF) and in- References
creasing BDNF, monocyte chemotactic protein-1 (MCP-1), T-cell-spe-
cific protein RANTES (regulated on activation, normal T cell expressed Aagaard, K., Ma, J., Antony, K.M., Ganu, R., Petrosino, J., Versalovic, J., 2014. The
and secreted), and macrophage inflammatory protein-1 beta (MIP-1) placenta harbors a unique microbiome. Sci. Transl. Med. 6, 237–265.
Aagaard, K., Riehle, K., Ma, J., Segata, N., Mistretta, T.A., Coarfa, C., et al., 2012. A
beta. Also, they found that probiotics were related to the regulation of metagenomic approach to characterization of the vaginal microbiome signature in
intestinal immune and epithelial cells and suggested that supple- pregnancy. PLoS One 7, e36466.
mentation of probiotics may improve control of gastrointestinal leakage Abdel-Haq, R., Schlachetzki, J.C.M., Glass, C.K., Mazmanian, S.K., 2019. Microbiome-
microglia connections via the gut-brain axis. J. Exp. Med. 216 (1), 41–59 ISSN: 1540-
in patients with schizophrenia (Tomasik et al., 2015). 9538.
Dickerson et al. (2014) performed a randomized, double-blind, Adlerberth, I., Lindberg, E., Åberg, N., Hesselmar, B., Saalman, R., Strannegård, I.L.,
placebo-controlled study and enrolled 65 patients with schizophrenia Wold, A.E., 2006. Reduced enterobacterial and increased staphylococcal colonization
of the infantile bowel: an effect of hygienic lifestyle? Pediatr. Res. 59, 96–101.
who were first treated with double-blind probiotic or placebo for 14 Adlerberth, I., Wold, A.E., 2009. Establishment of the gut microbiota in Western infants.
weeks. Although no significant differences between probiotics and Acta Paediatr. 98 (2), 229–238.
placebo groups were found in terms of changing schizophrenia symp- Ahnfeldt-Mollerup, P., Petersen, L.K., Kragstrup, J., Christensen, R.D., Sorensen, B., 2012.
Postpartum infections: occurrence, healthcare contacts and association with breast-
toms severity, probiotics reduced the likelihood to develop severe
feeding. Acta Obstet. Gynecol. Scand. 91, 1440–1444.
bowel difficulty throughout the trial. Afterward, Severance et al. (2017) Ait-Belgnaoui, A., Colom, A., Braniste, V., Ramalho, L., Marrot, A., Cartier, C., et al.,
conducted a randomized, placebo-controlled, longitudinal pilot study 2014. Probiotic gut effect prevents the chronic psychological stress-induced brain
and explored the use of probiotics in the treatment of both yeast gut activity abnormality in mice. Neurogastroenterol. Motil. 26, 510–520.
Al Khalaf, S.Y., O’Neill, S.M., O’Keeffe, L.M., Henriksen, T.B., Kenny, L.C., Cryan, J.F.,
infection and psychiatric symptoms 56 patients with schizophrenia. et al., 2015. The impact of obstetric mode of delivery on childhood behavior. Soc.
Probiotics were associated with a decrease of Candida albicans anti- Psychiatry Psychiatr. Epidemiol. 50, 1557–1567.
body levels as well as a decrease in gastrointestinal symptoms in male Al-Asmari, A.K., Khan, M.W., 2014. Inflammation and schizophrenia: alterations in cy-
tokine levels and perturbation in antioxidative defense systems. Hum. Exp. Toxicol.
subjects and trends for improvement in positive schizophrenia symp- 33 (2), 115–122.
toms in males who received probiotics and were seronegative for C. Almeida-Suhett, C.P., Scott, J.M., Graham, A., Chen, Y., Deuster, P.A., 2017. Control diet
albicans. in a high-fat diet study in mice: regular chow and purified low-fat diet have similar
effects on phenotypic, metabolic, and behavioral outcomes. [published online ahead
of print Jul 19]. Nutr. Neurosci.
Amenyogbe, N., Kollmann, T.R., Ben-Othman, R., 2017. Early-life host-microbiome in-
11. Conclusions terphase: the key frontier for immune development. Front. Pediatr. 5, 111.
Amir, L.H., Forster, D.A., Lumley, J., McLachlan, H., 2007. A descriptive study of mastitis
in Australian breastfeeding women: incidence and determinants. BMC Public Health
Trough the knowledge we already have, the studies and the analysis 7, 62.
being done recently, we can draw some conclusions. The microbiota Appleman, M.D., Wormser, G.P., 2005. Strict and facultative anaerobes: medical and
environmental aspects. Clin. Infect. Dis. 41 (July (1)), 132–133.
community transmitted to the offspring may have plasticity. Dynamic
Arboleya, S., Binetti, A., Salazar, N., Fernández, N., Solís, G., Hernández-Barranco, A.,
changes in the maternal microbiota and the early offspring microbiota Margolles, A., los Reyes-Gavilán, C.G., Gueimonde, M., 2012. Establishment and
would occur due to the prenatal and postnatal environments, respec- development of intestinal microbiota in preterm neonates. FEMS Microbiol. Ecol. 79,
tively, as well as host genetics. Although these changes may be bene- 763–772.
Arboleya, S., Suárez, M., Fernández, N., Mantecón, L., Solís, G., Gueimonde, M., de los
ficial for immediate survival, they may result in immediate and later Reyes-Gavilán, C.G., 2018. C-section and the neonatal gut microbiome acquisition:
physiological and behavioral consequences. This field has the potential consequences for future health. Ann. Nutr. Metab. 73 (Supplement), 17–23.
for the development of new bioactive and dietary/lifestyle interven- Ardissone, A.N., de la Cruz, D.M., Davis-Richardson, A.G., Rechcigl, K.T., Li, N., Drew,
J.C., et al., 2014. Meconium microbiome analysis identifies bacteria correlated with
tions, that would modulate the microbiota during pregnancy to reduce premature birth. PLoS One 9, e90784.
the risk of disease later in life. However, there are few studies directly Arweiler, N.B., Netuschil, L., 2016. The Oral Microbiota. Microbiota of the Human Body:
assessing the impact of changes to the microbiota composition early in Implications in Health and Disease. ISSN 22148019. pp. 45–60.
Azad, M.B., Bridgman, S.L., Becker, A.B., Kozyrskyj, A.L., 2014. Infant antibiotic exposure
life on brain health outcomes during adulthood. Further research and the development of childhood overweight and central adiposity. Int. J. Obes.
should focus on studying the microbiota modulation to support early (Lond) 38, 1290–1298.
neurodevelopment and brain health later in life. Azad, M.B., Coneys, J.G., Kozyrskyj, A.L., Field, C.J., Ramsey, C.D., Becker, A.B., et al.,
2013a. Probiotic supplementation during pregnancy or infancy for the prevention of
Additional studies of the relationship between schizophrenia and
asthma and wheeze: systematic review and meta-analysis. BMJ 347, f6471.
changes in the immune system, as well as the intestinal microbiota have Azad, M.B., Konya, T., Maughan, H., Guttman, D.S., Field, C.J., Chari, R.S., et al., 2013b.
great potential. In addition, exploring the possibility that resource such Gut microbiota of healthy Canadian infants: profiles by mode of delivery and infant
diet at 4 months. CMAJ 185, 385–394.
as probiotics may contribute to the treatment of inflammatory processes
Babulas, V., Factor-Litvak, P., Goetz, R., Schaefer, C.A., Brown, A.S., 2006. Prenatal ex-
in the gastrointestinal system, and have a positive influence on the posure to maternal genital and reproductive infections and adult schizophrenia. Am.
symptoms of schizophrenia, may promote the development of new J. Psychiatry 163 (5), 927–929.
therapies to help the human with this disease and potentially reducing Bäckhed, F., Roswall, J., Peng, Y., Feng, Q., Jia, H., Kovatcheva-Datchary, P., Li, Y., Xia,
Y., Xie, H., Zhong, H., et al., 2015. Dynamics and stabilization of the human gut
the adverse metabolic side effects of current treatments. microbiome during the first year of life. Cell Host Microbe 17, 690–703.
It will also be significant to research the effect of antibiotic treat- Bäckhed, F., 2005. Host-bacterial mutualism in the human intestine. Science 307,
ment on neuropsychiatric conditions through the modification of the 1915–1920.
Bagci, S., Brosens, E., Tibboel, D., De Klein, A., Ijsselstijn, H., Wijers, C.H.W., Roeleveld,
intestinal microbiota. In addition, the study of this area can lead to the N., de Blaauw, I., Broens, P.M., van Rooij, I.A.L.M., et al., 2016. More than fetal urine:
creation of special dietary regimes, lifestyle interventions, which would enteral uptake of amniotic fluid as a major predictor for fetal growth during late
contribute to the modulation of the microbiota during pregnancy, di- gestation. Eur. J. Pediatr. 175, 825–831.
Bager, P., Wohlfahrt, J., Westergaard, T., 2008. Caesarean delivery and risk of atopy and
minishing the risk of developing the disease at a later age. allergic disease: meta-analyses. Clin. Exp. Allergy 38, 634–642.
Bahr, S.M., Tyler, B.C., Wooldridge, N., Butcher, B.D., Burns, T.L., Teesch, L.M., Oltman,
C.L., Azcarate Peril, M.A., Kirby, J.R., Calarge, C.A., 2015a. Use of the second-gen-
eration antipsychotic, risperidone, and secondary weight change are associated with
Acknowledgments
an altered gut microbiota in children. Transl. Psychiatry 5, e652.
Bahr, S.M., Weidemann, B.J., Castro, A.N., Walsh, J.W., deLeon, O., Burnett, C.M.L.,
This work was supported by the GACR 304/18-09296S, AZV CR 17- Pearson, N.A., et al., 2015b. Risperidone-induced weight gain is mediated through
shifts in gut microbiome and suppression of energy expenditure. EBioMedicine 2
31852A, Institutional support for NIMH-CZ was provided by the project
(11), 1725–1734.
"Sustainability for the National Institute of Mental Health" number Bailey, M.T., Dowd, S.E., Galley, J.D., Hufnagle, A.R., Allen, R.G., Lyte, M., 2011.
LO161, "PharmaBrain" CZ.02.1.01/0.0/0.0/16_025/0007444 and Third Exposure to a social stressor alters the structure of the intestinal microbiota: im-
Faculty of Medicine, Charles University in Prague. We would like to plications for stressor-induced immunomodulation. Brain Behav. Immun. 25,
397–407.
thank DanielBermejo-Rodriguez for language proofreading.

723
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Bale, T.L., 2015. Epigenetic and transgenerational reprogramming of brain development. Cho, C.E., Norman, M., 2013. Cesarean section and development of the immune system in
Nat. Rev. Neurosci. 16, 332–344. the offspring. Am. J. Obstet. Gynecol. 208, 249–254.
Barker, D.J.P., 1998. Mothers, Babies and Health in Later Life, 2nd ed. Churchill Chong, C.Y.L., Bloomfield, F.H., O’Sullivan, J.M., 2018. Factors affecting gastrointestinal
Livingstone, Edinburgh. microbiome development in neonates. Nutrients 10 (February (3)), 274 ISSN: 2072-
Barrett, E., Kerr, C., Murphy, K., O’Sullivan, O., Ryan, C.A., Dempsey, E.M., et al., 2013. 6643.
The individual-specific and diverse nature of the preterm infant microbiota. Arch. Choy, K.H., de Visser, Y.P., van den Buuse, M., 2009. The effect of ‘two hit’ neonatal and
Dis. Child. Fetal Neonatal Ed. 98 (4), F334–40. young-adult stress on dopaminergic modulation of prepulse inhibition and dopamine
Barrett, E., Ross, R.P., O’Toole, P.W., Fitzgerald, G.F., Stanton, C., 2012. γ-Aminobutyric receptor density. Br. J. Pharmacol. 156, 388–396.
acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. Christian, L.M., Galley, J.D., Hade, E.M., Schoppe-Sullivan, S., Dush, C.K., Bailey, M.T.,
113 (2), 411–417. 2015. Gut microbiome composition is associated with temperament during early
Bäumler, A.J., Sperandio, V., 2016. Interactions between the microbiota and pathogenic childhood. Brain Behav. Immun. 45, 118–127.
bacteria in the gut. Nature 535, 85–93. Chu, D.M., Ma, J., Prince, A.L., Antony, K.M., Seferovic, M.D., Aagaard, K.M., 2017.
Bengmark, S., 1998. Ecological control of the gastrointestinal tract. The role of probiotic Maturation of the infant microbiome community structure and function across mul-
flora. Gut 42, 2–7. tiple body sites and in relation to mode of delivery. Nat. Med. 23 (3), 314–326.
Bercik, P., Denou, E., Collins, J., 2011. The intestinal microbiota affect central levels of Chudal, R., Sourander, A., Polo-Kantola, P., et al., 2014. Perinatal factors and the risk of
brain-derived neurotropic factor and behavior in mice. Gastroenterology 141. bipolar disorder in Finland. J. Affect. Disord. 155, 75–80.
Betts, K.S., Williams, G.M., Najman, J.M., Alati, R., 2015. The relationship between Clarke, G., Grenham, S., Scully, P., Fitzgerald, P., Moloney, R.D., Shanahan, F., Dinan,
maternal depressive, anxious, and stress symptoms during pregnancy and adult off- T.G., Cryan, J.F., 2013. The microbiome-gut-brain axis during early life regulates the
spring behavioral and emotional problems. Depress. Anxiety 32, 82–90. hippocampal serotonergic system in a sex-dependant manner. Mol. Psychiatry 18,
Biasucci, G., Benenati, B., Morelli, L., Bessi, E., Boehm, G., 2008. Cesarean delivery may 666–673.
affect the early biodiversity of intestinal bacteria. J. Nutr. 138, 1796S–1800S. Clarke, G., O’Mahony, S.M., Dinan, T.G., Cryan, J.F., 2014. Priming for health: gut mi-
Biasucci, G., Rubini, M., Riboni, S., Morelli, L., Bessi, E., Retetangos, C., 2010. Mode of crobiota acquired in early life regulates physiology, brain and behaviour. Acta
delivery affects the bacterial community in the newborn gut. Early Hum. Dev. 86 Paediatr. 103 (8), 812–819.
(Suppl 1), 13–15. Collado, M.C., Isolauri, E., Laitinen, K., Salminen, S., 2008. Distinct composition of gut
Bilbo, S.D., Smith, S.H., Schwarz, J.M., 2012. A lifespan approach to neuroinflammatory microbiota during pregnancy in overweight and normal-weight women. Am. J. Clin.
and cognitive disorders: a critical role for glia. J. Neuroimmune Pharmacol. 7, 24–41. Nutr. 88, 894–899.
Blakelock, R., Upadhyay, V., Kimble, R., Pease, P., Kolbe, A., Harding, J., 1998. Is a Collado, M.C., Rautava, S., Aakko, J., Isolauri, E., Salminen, S., 2016. Human gut colo-
normally functioning gastrointestinal tract necessary for normal growth in late ge- nisation may be initiated in utero by distinct microbial communities in the placenta
station? Pediatr. Surg. Int. 13, 17–20. and amniotic fluid. Sci. Rep. 6, 23129.
Bondar, N.P., Merkulova, T.I., 2016. Brain-derived neurotrophic factor and early-life Collins, S.M., Surette, M., Bercik, P., 2012. The interplay between the intestinal micro-
stress: multifaceted interplay. J. Biosci. 41 (4), 751–758. biota and the brain. Nat Rev Micro. 10 (11), 735–742.
Borre, Y.E., O’Keeffe, G.W., Clarke, G., Stanton, C., Dinan, T.G., Cryan, J.F., 2014. Cooper, W.O., Willy, M.E., Pont, S.J., Ray, W.A., 2007. Increasing use of antidepressants
Microbiota and neurodevelopmental windows: implications for brain disorders. in pregnancy. Am. J. Obstet. Gynecol. 196 (6), 544 e1-5.
Trends Mol. Med. 20 (9), 509–518. Costa-Ramón, A.M., Rodríguez-González, A., Serra-Burriel, M., Campillo-Artero, C., 2018.
Brander, G., Rydell, M., Kuja-Halkola, R., et al., 2016. Association of perinatal risk factors It’s about time: cesarean sections and neonatal health. J. Health Econ. 59, 46–59.
with obsessive-compulsive disorder: a population-based birth cohort, sibling control Cox, L.M., Yamanishi, S., Sohn, J., Alekseyenko, A.V., Leung, J.M., Cho, I., Kim, S.G., Li,
study. JAMA Psychiatry 73, 1135–1144. H., Gao, Z., Mahana, D., et al., 2014. Altering the intestinal microbiota during a
Bravo, J.A., Forsythe, P., Chew, M.V., Escaravage, E., Savignac, H.M., Dinan, T.G., et al., critical developmental window has lasting metabolic consequences. Cell 158,
2011. Ingestion of Lactobacillus strain regulates emotional behavior and central 705–721.
GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. U. S. Critchley, H.D., Harrison, N., 2013. A visceral influences on brain and behavior. Neuron.
A. 108, 16050–16055. 77, 624–638.
Brown, S., Lumley, J., 1998. Maternal health after childbirth: results of an Australian Crumeyrolle-Arias, M., Jaglin, M., Bruneau, A., Vancassel, S., Cardona, A., Daugé, V.,
population based survey. Br. J. Obstet. Gynaecol. 105, 156–161. Naudon, L., Rabot, S., 2014. Absence of the gut microbiota enhances anxiety-like
Bruce-Keller, A.J., Salbaum, J.M., Berthoud, H.R., 2018. Harnessing gut microbes for behavior and neuroendocrine response to acute stress in rats.
mental health: getting from here to there. Biol. Psychiatr. 83, 214–223. Psychoneuroendocrinology. 42, 207–217.
Bruce-Keller, A.J., Salbaum, J.M., Luo, M., Blanchard, E.T., Taylor, C.M., Welsh, D.A., Cryan, J.F., Dinan, T.G., 2012. Mind-altering microorganisms: the impact of the gut mi-
et al., 2015. Obese-type gut microbiota induce neurobehavioral changes in the ab- crobiota on brain and behaviour. Nat. Rev. Neurosci. 13, 701–712.
sence of obesity. Biol. Psychiatr. 77, 607–615. Cryan, J.F., Dinan, T.G., 2015. More than a gut feeling: the microbiota regulates neuro-
Brumbaugh, D.E., Arruda, J., Robbins, K., Ir, D., Santorico, S.A., Robertson, C.E., et al., development and behavior. Neuropsychopharmacology 40, 241–242.
2016. Mode of delivery determines neonatal pharyngeal bacterial composition and Cryan, J.F., O’Mahony, S.M., 2011. The microbiome-gut-brain axis: from bowel to be-
early intestinal colonization. J. Pediatr. Gastroenterol. Nutr. 63, 320–328. havior. Neurogastroenterol. Motil. 23, 187–192.
Buffington, S.A., Di Prisco, G.V., Auchtung, T.A., Ajami, N.J., Petrosino, J.F., Costa- Csiszar, K., Molnar, J., 1992. Mechanism of action of tricyclic drugs on Escherichia coli
Mattioli, M., 2016. Microbial reconstitution reverses maternal diet-induced social and and Yersinia enterocolitica plasmid maintenance and replication. Anticancer Res. 12,
synaptic deficits in offspring. Cell 165, 1762–1775. 2267–2272.
Butte, N., Lopez-Alarcon, M., Garza, C., 2002. Nutrient Adequacy of Exclusive Curran, E.A., Cryan, J.F., Kenny, L.C., Dinan, T.G., Kearney, P.M., Khashan, A.S., 2016.
Breastfeeding for the Term Infant During the First Six Months of Life. World Health Obstetrical mode of delivery and childhood behavior and psychological development
Organization, Geneva 47 p. in a British cohort. J. Autism Dev. Disord. 46, 603–614.
Capuron, L., Pagnoni, G., Drake, D.F., Woolwine, B.J., Spivey, J.R., Crowe, R.J., et al., Curran, E.A., Kenny, L.C., Dalman, C., Kearney, P.M., Cryan, J.F., Dinan, T.G., et al.,
2012. Dopaminergic mechanisms of reduced basal ganglia responses to hedonic re- 2017. Birth by caesarean section and school performance in Swedish adolescents—a
ward during interferon alfa administration. Arch. Gen. Psychiatry 69, 1044–1053. population-based study. BMC Pregnancy Childbirth 17, 121.
Cardwell, C.R., Stene, L.C., Joner, G., Cinek, O., Svensson, J., Goldacre, M.J., Parslow, Curran, E.A., O’Neill, S.M., Cryan, J.F., Kenny, L.C., Dinan, T.G., Khashan, A.S., Kearney,
R.C., Pozzilli, P., Brigis, G., Stoyanov, D., Urbonaite, B., Sipetic, S., Schober, E., P.M., 2015. Research Review: birth by caesarean section and development of autism
Ionescu-Tirgoviste, C., Devoti, G., de Beaufort, C.E., Buschard, K., Patterson, C.C., spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review
2008. Caesarean section is associated with an increased risk of childhood-onset type 1 and meta-analysis. J. Child Psychol. Psychiatry 56 (5), 500–508.
diabetes mellitus: a meta-analysis of observational studies. Diabetologia 51, 726–735. Da Silva, H.E.C., Stefani, C.M., de Santos Melo, N., de Almeida de Lima, A., et al., 2017.
Castro-Nallar, E., Bendall, M.L., Perez-Losada, M., Sabuncyan, S., Severance, E.G., Effect of intrapregnancy nonsurgical periodontal therapy on inflammatory bio-
Dickerson, F.B., Schroeder, J.R., Yolken, R.H., Crandall, K.A., 2015. Composition, markers and adverse pregnancy outcomes: a systematic review with metaanalysis.
taxonomy and functional diversity of oropharynx microbiome in individuals with Syst. Rev. 6 (October), 197.
schizophrenia and controls. PeerJ 3, e1140. Dasanayake, A.P., Boyd, D., Madianos, P.N., Offenbacher, S., Hills, E., 2001. The asso-
Cenit, M.C., Nuevo, I.C., Codoner-Franch, P., Dinan, T.G., Sanz, Y., 2017a. Gut microbiota ciation between Porphyromonas gingivalis-specific maternal serum IgG and low birth
and attention deficit hyperactivity disorder: new perspectives for a challenging weight. J. Periodontol. 72, 1491–1497.
condition. Eur. Child Adolesc. Psychiatry 26, 1081–1092. Dasanayake, A.P., Li, Y., Wiener, H., Ruby, J.D., Lee, M.J., 2005. Salivary Actinomyces
Cenit, M.C., Sanz, Y., Codoñer-Franch, P., 2017b. Influence of gut microbiota on neu- naeslundii genospecies 2 and Lactobacillus casei levels predict pregnancy outcomes.
ropsychiatric disorders. World J. Gastroenterol. 23 (30), 5486–5498. J. Periodontol. 76 (2), 171–177.
Chang, C., Lin, H., 2016. Dysbiosis in gastrointestinal disorders. Best Pract. Res. Clin. Dasanayake, A.P., 1998. Poor periodontal health of the pregnant woman as a risk factor
Gastroenterol. 30, 3–15. for low birth weight. Ann. Periodontol. 3, 206–212.
Chase, K.A., Rosen, C., Gin, H., Bjorkquist, O., Feiner, B., Marvin, R., Conrin, S., Sharma, Davenport, E.S., Williams, C.E., Sterne, J.A., Murad, S., Sivapathasundram, V., Curtis,
R.P., 2015. Metabolic and inflammatory genes in schizophrenia. Psychiat. Res. 225 M.A., 2002. Maternal periodontal disease and preterm low birthweight: case-control
(1–2), 208–211. study. J. Dent. Res. 81, 313–318.
Chavan, S.S., Pavlov, V.A., Tracey, K.J., 2017. Mechanisms and therapeutic relevance of Davey, J.K., O’Mahoney, S.M., Schellekens, H., O’Sullivan, O., Bienenstock, J., Cotter,
neuro-immune communication. Immunity 46, 927–942. P.D., Dinan, T.G., Cryan, J.F., 2012. Genderdependent consequences of chronic
Chaves, R.G., Lamounier, J.A., 2004. Breastfeeding and maternal medications. J. Pediatr. olanzapine in the rat: effect on body weight, inflammatory, metabolic and microbiota
80 (5), 189–198. parameters. Psychopharmacology 221 (1), 155–169.
Chistiakov, D.A., Bobryshev, Y.V., Kozarov, E., Sobenin, I.A., Orekhov, A.N., 2014. Davey, K.J., Cotter, P.D., O’Sullivan, O., Crispie, F., Dinan, T.G., Cryan, J.F., et al., 2013.
Intestinal mucosal tolerance and impact of gut microbiota to mucosal tolerance. Antipsychotics and the gut microbiome: olanzapine induced metabolic dysfunction is
Front. Microbiol. 5, 781. attenuated by antibiotic administration in the rat. Transl. Psychiatry 3, e309.

724
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

David, L.A., Materna, A.C., Friedman, J., Campos-Baptista, M.I., Blackburn, M.C., 11971–11975.
Perrotta, A., Erdman, S.E., Alm, E.J., 2014. Host lifestyle affects human microbiota on Dominguez-Bello, M.G., De Jesus-Laboy, K.M., Shen, N., Cox, L.M., Amir, A., Gonzalez,
daily timescales. Genome Biol. 15 (7), R89. A., Bokulich, N.A., Song, S.J., Hoashi, M., Rivera-Vinas, J.I., et al., 2016. Partial re-
de Theije, C.G., Wopereis, H., Ramadan, M., van Eijndthoven, T., Lambert, J., Knol, J., storation of the microbiota of cesarean-born infants via vaginal microbial transfer.
et al., 2014. Altered gut microbiota and activity in a murine model of autism spec- Nat. Med. 22, 250–253.
trum disorders. Brain Behav. Immun. 37, 197–206. Doron, S., Snydman, D.R., 2015. Risk and safety of probiotics. Clin. Infect. Dis. 60,
de Theije, C.G.M., Wu, J., Silva, S.L., Kamphuis, P.J., Garssen, J., Korte, S.M., Kraneveld, S129–S134.
A.D., 2011. Pathways underlying the gut-to-brain connection in autism spectrum Dowlati, Y., Herrmann, N., Swardfager, W., Liu, H., Sham, L., Reim, E.K., et al., 2010. A
disorders as future targets for disease management. Eur. J. Pharmacol. 668, 570–580. meta-analysis of cytokines in major depression. Biol. Psychiatry 67, 446–457.
Deans, E., 2017. Microbiome and mental health in the modern environment. J. Physiol. Du, Y., Yang, M., Lee, S., Behrendt, C.L., Hooper, L.V., Saghatelian, A., et al., 2012.
Anthropol. 36, 1. Maternal Western diet causes inflammatory milk and TLR2/4- dependent neonatal
Decker, E., Engelmann, G., Findeisen, A., Gerner, P., Laass, M., Ney, D., Posovszky, C., toxicity. Genes Dev. 26, 1306–1311.
Hoy, L., Hornef, M.W., 2010. Cesarean delivery is associated with celiac disease but Edlow, A.G., Guedj, F., Pennings, J.L., Sverdlov, D., Neri, C., Bianchi, D.W., 2016. Males
not inflammatory bowel disease in children. Pediatrics 125, e1433–e1440. are from Mars, and females are from Venus: sexspecific fetal brain gene expression
Declercq, E., Sakala, C., Corry, M., Applebaum, S., 2008. New Mothers Speak Out: signatures in a mouse model of maternal diet-induced obesity. Am. J. Obstet.
National Survey Results Highlight Women’s Postpartum Experiences. Childbirth Gynecol. 214 (623), e1–e623 e10.
Connection, New York. El Aidy, S., Dinan, T.G., Cryan, J.F., 2014. Immune modulation of the brain-gut-microbe
Degroote, S., Hunting, D.J., Baccarelli, A.A., Takser, L., 2016. Maternal gut and fetal brain axis. Front. Microbiol. 5, 146.
connection: increased anxiety and reduced social interactions in Wistar rat offspring Elazab, N., Mendy, A., Gasana, J., Vieira, E.R., Quizon, A., Forno, E., 2013. Probiotic
following peri-conceptional antibiotic exposure. Prog. Neuropsychopharmacol. Biol. administration in early life, atopy, and asthma: a metaanalysis of clinical trials.
Psychiatry 71, 76–82. Pediatrics 132, e666–e676.
den Besten, G., van Eunen, K., Groen, A.K., Venema, K., Reijngoud, D.-J., Bakker, B.M., Ellen, R., 1982. Oral colonization by Gram-positive bacteria significant to periodontal
2013. The role of short-chain fatty acids in the interplay between diet, gut micro- disease. In: Genco, R., Mergenhagen, S. (Eds.), Host-Parasite Interactions in
biota, and host energy metabolism. J. Lipid Res. 54, 2325–2340. Periodontal Diseases. ASM, Washington, pp. 98–111.
Deopurkar, R., Ghanim, H., Friedman, J., Abuaysheh, S., Sia, C.L., Mohanty, P., et al., Epstein, F.H., Goldenberg, R.L., Hauth, J.C., Andrews, W.W., 2000. Intrauterine infection
2010. Differential effects of cream, glucose, and orange juice on inflammation, en- and preterm delivery. N. Engl. J. Med. 342, 1500–1507.
dotoxin, and the expression of Toll-like receptor- 4 and suppressor of cytokine sig- Erny, D., Hrab_e de Angelis, A.L., Jaitin, D., Wieghofer, P., Staszewski, O., David, E., et al.,
naling-3. Diabetes Care 33, 991–997. 2015. Host microbiota constantly control maturation and function of microglia in the
Depino, A.M., 2015. Early prenatal exposure to LPS results in anxiety and depression- CNS. Nat. Neurosci. 18, 965–977.
related behaviors in adulthood. Neuroscience. 299, 56–65. Escobar, M., Crouzin, N., Cavalier, M., Quentin, J., Roussel, J., Lanté, F., et al., 2011.
Desbonnet, L., Clarke, G., Shanahan, F., Dinan, T.G., Cryan, J.F., 2014. Microbiota is Early, time-dependent disturbances of hippocampal synaptic transmission and plas-
essential for social development in the mouse. Mol. Psychiatry 19 (2), 146–148. ticity after in utero immune challenge. Biol. Psychiatry 70, 992–999.
Desbonnet, L., Clarke, G., Traplin, A., O’Sullivan, O., Crispie, F., Moloney, R.D., Cotter, Escobar-Arregoces, F., Latorre-Uriza, C., Velosa-Porras, J., Roa-Molina, N., Ruiz, A., Silva,
P.D., Dinan, T.G., Cryan, J.F., 2015. Gut microbiota depletion from early adolescence J., Arias, E., Echeverri, J., 2018. Inflamatory response in pregnant women with high
in mice: implications for brain and behaviour. Brain Behav. Immun. 48, 165–173. risk of preterm delivery and its relationship with periodontal disease. A pilot study.
Desbonnet, L., Garett, L., Clarke, G., Bienenstock, J., Dinan, T.G., 2009. The probiotic Acta Odontol. Latinoam. 31 (June (1)), 53–57 ISSN: 1852-4834.
Bifidobacteria infantis: an assessment of potential antidepressant properties in the Estes, M.L., McAllister, A.K., 2016. Maternal immune activation: implications for neu-
rat. J. Psychiat Res. 43, 164–174. ropsychiatric disorders. Science 353, 772–777.
Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryan, J.F., Dinan, T.G., 2010. Effects of Fallani, M., Young, D., Scott, J., Norin, E., Amarri, S., Adam, R., Aguilera, M., Khanna, S.,
the probiotic Bifidobacterium infantis in the maternal separation model of depres- Gil, A., Edwards, C.A., et al., 2010. Intestinal microbiota of 6-week-old infants across
sion. Neuroscience. 170, 1179–1188. Europe: geographic influence beyond delivery mode, breast-feeding, and antibiotics.
Diaz Heijtz, R., Wang, S., Anuar, F., Qian, Y., Björkholm, B., Samuelsson, A., Hibberd, J. Pediatr. Gastroenterol. Nutr. 51, 77–84.
M.L., Forssberg, H., Pettersson, S., 2011. Normal gut microbiota modulates brain Falony, G., Joossens, M., Vieira-Silva, S., Wang, J., Darzi, Y., Faust, K., Kurilshikov, A.,
development and behavior. Proc. Natl. Acad. Sci. U. S. A. 108 (7), 3047–3052. Bonder, M.J., Valles-Colomer, M., Vandeputte, D., Tito, R.Y., Chaffron, S., Rymenans,
Dickerson, F., Severance, E., Yolken, R., 2017. The microbiome, immunity, and schizo- L., Verspecht, C., De, S.L., Lima-Mendez, G., D’hoe, K., Jonckheere, K., Homola, D.,
phrenia and bipolar disorder. Brain Behav. Immun. 62, 46–52. Garcia, R., Tigchelaar, E.F., Eeckhaudt, L., Fu, J., Henckaerts, L., Zhernakova, A.,
Dickerson, F., Stallings, C., Origoni, A., Boronow, J., Yolken, R., 2007. C-reactive protein Wijmenga, C., Raes, J., 2016. Population-level analysis of gut microbiome variation.
is associated with the severity of cognitive impairment but not of psychiatric symp- Science 352, 560–564.
toms in individuals with schizophrenia. Schizophr. Res. 93 (1–3), 261–265. Fanaro, S., Boehm, G., Garssen, J., Knol, J., Mosca, F., Stahl, B., Vigi, V., 2005. Galacto-
Dickerson, F.B., Stallings, C., Origoni, A., Katsafanas, E., Savage, C.L., Schweinfurth, L.A., oligosaccharides and long-chain fructo-oligosaccharides as prebiotics in infant for-
Goga, J., Khushalani, S., Yolken, R.H., 2014. Effect of probiotic supplementation on mulas: a review. Acta Paediatr. Suppl. 94 (449), 22–26.
schizophrenia symptoms and association with gastrointestinal functioning: a rando- Farshim, P., Walton, G., Chakrabarti, B., Givens, I., Saddy, D., Kitchen, I., et al., 2016.
mized, placebo-controlled trial. Prim. Care Companion CNS Disord. 16 (1). Maternal weaning modulates emotional behavior and regulates the gut-brain axis.
DiGiulio, D.B., Callahan, B.J., McMurdie, P.J., Costello, E.K., Lyell, D.J., Robaczewska, A., Sci. Rep. 6, 21958.
et al., 2015. Temporal and spatial variation of the human microbiota during preg- Felger, J.C., Miller, A.H., 2012. Cytokine eff ;ects on the basal ganglia and dopamine
nancy. Proc. Natl. Acad. Sci. U. S. A. 112, 11060–11065. function: the subcortical source of inflammatory malaise. Front. Neuroendocrinol. 33,
DiGiulio, D.B., Romero, R., Amogan, H.P., Kusanovic, J.P., Bik, E.M., Gotsch, F., Kim, C.J., 315–327.
Erez, O., Edwin, S., Relman, D.A., 2008. Microbial prevalence, diversity and abun- Felger, J.C., Mun, J., Kimmel, H.L., Nye, J.A., Drake, D.F., Hernandez, C.R., et al., 2013.
dance in amniotic fluid during preterm labor: a molecular and culture-based in- Chronic interferon-alpha decreases dopamine 2 receptor binding and striatal dopa-
vestigation. PLoS One 3 (8), e3056. mine release in association with anhedonia-like behavior in nonhuman primates.
Dinan, T.G., Cryan, J.F., 2017b. Gut instincts: microbiota as a key regulator of brain Neuropsychopharmacology 38, 2179–2187.
development, ageing and neurodegeneration. J. Physiol. 595 (2), 489–503. Feng, Y., Wang, Y., Wang, P., Huang, Y., Wang, F., 2018. Short-chain fatty acids manifest
Dinan, T.G., Cryan, J.F., 2017a. Gut-brain axis in 2016: brain-gut-microbiota axis - mood, stimulative and protective effects on intestinal barrier function through the inhibition
metabolism and behaviour. Nat. Rev. Gastroenterol. Hepatol. 14 (2), 69–70. of NLRP3 inflammasome and autophagy. Cell. Physiol. Biochem. 49, 190–205.
Dinan, T.G., Cryan, J.F., 2013. Melancholic microbes: a link between gut microbiota and Filiano, A.J., Gadani, S.P., Kipnis, J., 2017. How and why do T cells and their derived
depression? Neurogastroenterol. Motil. 25, 713–719. cytokines aff ;ect the injured and healthy brain? Nat. Rev. Neurosci. 18, 375–384.
Dinan, T.G., Cryan, J.F., 2012. Regulation of the stress response by the gut microbiota: Finegold, S.M., Dowd, S.E., Gontcharova, V., Liu, C., Henley, K.E., Wolcott, R.D., et al.,
implications for psychoneuroendocrinology. Psychoneuroendocrinology. 37, 2010. Pyrosequencing study of fecal microflora of autistic and control children.
1369–1378. Anaerobe 16, 444–453.
Dinan, T.G., Borre, Y.E., Cryan, J.F., 2014. Genomics of schizophrenia: time to consider Finegold, S.M., Molitoris, D., Song, Y., Liu, C., Vaisanen, M.L., Bolte, E., McTeague, M.,
the gut microbiome? Mol. Psychiatry 19, 1252–1257. Sandler, R., Wexler, H., Marlowe, E.M., Collins, M.D., Lawson, P.A., Summanen, P.,
Dinan, T.G., Stilling, R.M., Stanton, C., Cryan, J.F., 2015. Collective unconscious: how gut Baysallar, M., Tomzynski, T.J., Read, E., Johnson, E., Rolfe, R., Nasir, P., Shah, H.,
microbes shape human behavior. J Psychiatr Res. Elsevier Ltd. 63, 1–9. Haake, D.A., Manning, P., Kaul, A., 2002. Gastrointestinal microflora studies in late-
Ding, T., Schloss, P.D., 2014. Dynamics and associations of microbial community types onset autism. Clin. Infect. Dis. 35 (1), 6–16.
across the human body. Nature 509, 357–360. Fiorini, T., Susin, C., da Rocha, J.M., Weidlich, P., et al., 2013. Effect of nonsurgical
Dixon, N.G., Ebright, D., Defrancesco, M.A., Hawkins, R.E., 1994. Orogenital contact: A periodontal therapy on serum and gingival crevicular fluid cytokine levels during
cause of chorioamnionitis? Obstet. Gynecol. 84 (Pt. 2), 654–655. pregnancy and postpartum. J. Periodontal Res. Suppl. 48, 126–133.
Dogra, S., Sakwinska, O., Soh, S.E., Ngom-Bru, C., Brück, W.M., Berger, B., et al., 2015. Flierl, M.A., Rittirsch, D., Nadeau, B.A., Chen, A.J., Sarma, J.V., Zetoune, F.S., McGuire,
Dynamics of infant gut microbiota are influenced by delivery mode and gestational S.R., List, R.P., Day, D.E., Hoesel, L.M., Gao, H., Van Rooijen, N., Huber-Lang, M.S.,
duration and are associated with subsequent adiposity. MBio 6 e02419–14. Neubig, R.R., Ward, P.A., 2007. Phagocyte-derived catecholamines enhance acute
Doherty, F.D., O’Mahony, S.M., Peterson, V.L., O’Sullivan, O., Crispie, F., Cotter, P.D., inflammatory injury. Nature. 449 (7163), 721–725.
et al., 2018. Post-weaning social isolation of rats leads to long-term disruption of the Flint, H.J., Scott, K.P., Duncan, S.H., Louis, P., Forano, E., 2012. Microbial degradation of
gut microbiota-immune-brain axis. Brain Behav. Immun. 68, 261–273. complex carbohydrates in the gut. Gut Microbes 3, 289–306.
Dominguez-Bello, M.G., Costello, E.K., Contreras, M., Magris, M., Hidalgo, G., Fierer, N., Fond, G., Bulzacka, E., Boyer, L., Llorca, P.M., Godin, O., Brunel, L., et al., 2017. Birth by
Knight, R., 2010. Delivery mode shapes the acquisition and structure of the initial cesarean section and schizophrenia: results from the multicenter FACE-SZ data-set.
microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. 107 (26), Eur. Arch. Psychiatry Clin. Neurosci. 267, 587–594.

725
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Foster, J.A., McVey Neufeld, K.A., 2013. Gut-brain axis: how the microbiome influences nationwide cohort study. BMJ Open 8, e018479.
anxiety and depression. Trends Neurosci. 36, 305–312. Harding, J.E., 2001. The nutritional basis of the fetal origins of adult disease. Int. J.
Francino, M.P., 2014. Early development of the gut microbiota and immune health. Epidemiol. 30, 15–23.
Pathogens 3, 769–790. Haroon, E., Raison, C.L., Miller, A.H., 2012. Psychoneuroimmunology meets neu-
Frayling, T.M., Timpson, N.J., Weedon, M.N., Zeggini, E., Freathy, R.M., et al., 2007. A ropsychopharmacology: translational implications of the impact of inflammation on
common variant in the FTO gene is associated with body mass index and predisposes behavior. Neuropsychopharmacology 37, 137–162.
to childhood and adult obesity. Science 316 (5826), 889–894. Harrison, N.A., Brydon, L., Walker, C., Gray, M.A., Steptoe, A., Critchley, H.D., 2009.
Freytag, V., Carrillo-Roa, T., Milnik, A., 2017. A peripheral epigenetic signature of im- Inflammation causes mood changes through alterations in subgenual cingulate ac-
mune system genes is linked to neocortical thickness and memory. Nat. Commun. 8, tivity and mesolimbic connectivity. Biol. Psychiatry 66, 407–414.
15193. Harry, G.J., 2013. Microglia during development and aging. Pharmacol. Ther. 139,
Fricke, E.M., Elgin, T.G., Gong, H., Reese, J., Gibson-Corley, K.N., Weiss, R.M., et al., 313–326.
2018. Lipopolysaccharide-induced maternal inflammation induces direct placental Hehemann, J.-H., Correc, G., Barbeyron, T., Helbert, W., Czjzek, M., Michel, G., 2010.
injury without alteration in placental blood flow and induces a secondary fetal in- Transfer of carbohydrate-active enzymes from marine bacteria to Japanese gut mi-
testinal injury that persists into adulthood. Am. J. Reprod. Immunol. 79, e12816. crobiota. Nature 464, 908–912.
Friedrich, M.J., 2013. Genomes of microbes inhabiting the body offer clues to human Heijtz, R.D., Wang, S., Anuar, F., et al., 2011. Normal gut microbiota modulates brain
health and disease. JAMA 309, 1447. development and behavior. Proc. Natl. Acad. Sci. U. S. A. 108, 3047–3052.
Frodl, T., Amico, F., 2014. Is there an association between peripheral immune markers Heim, C., Nemeroff, C.B., 2001. The role of childhood trauma in the neurobiology of
and structural/functional neuroimaging findings? Prog. Neuro Psychopharmacol. mood and anxiety disorders: preclinical and clinical studies. Biol. Psychiatry 49,
Biol. Psychiatr. 48, 295–303. 1023–1039.
Fujimura, K.E., Slusher, N.A., Cabana, M.D., Lynch, S.V., 2010. Role of the gut microbiota Henriksen, M.G., Nordgaard, J., Jansson, L.B., 2017. Genetics of schizophrenia: overview
in defining human health. Expert Rev. Anti. Ther. 8 (4), 435–454. of methods, findings and limitations. Front. Hum. Neurosci. 11, 322.
Fujimura, K.E., Sitarik, A.R., Havstad, S., Lin, D.L., Levan, S., Fadrosh, D., et al., 2016. Herbert, A., Gerry, N.P., McQueen, M.B., Heid, I.M., Pfeufer, A., Illig, T., Wichmann, H.E.,
Neonatal gut microbiota associates with childhood multisensitized atopy and T cell Meitinger, T., Hunter, D., Hu, F.B., Colditz, G., Hinney, A., Hebebrand, J., Koberwitz,
diff ;erentiation. Nat. Med. 22, 1187–1191. K., Zhu, X., Cooper, R., Ardlie, K., Lyon, H., Hirschhorn, J.N., Laird, N.M., Lenburg,
Funkhouser, L.J., Bordenstein, S.R., 2013. Mom knows best: the universality of maternal M.E., Lange, C., Christman, M.F., 2006. A common genetic variant is associated with
microbial transmission. PLoS Biol. 11, e1001631. adult and childhood obesity. Science. 312 (5771), 279–283.
Gareau, M.G., Wine, E., Rodrigues, D.M., Cho, J.H., Whary, M.T., Philpott, D.J., Hidaka, B.H., 2012. Depression as a disease of modernity: explanations for increasing
Macqueen, G., Sherman, P.M., 2011. Bacterial infection causes stress-induced prevalence. J. Affect. Disord. 140, 205–214.
memory dysfunction in mice. Gut. 60 (3), 307–317. Hill, C.J., Lynch, D.B., Murphy, K., Ulaszewska, M., Jeffery, I.B., O’Shea, C.A., et al., 2017.
GBD, 2016. Disease and Injury Incidence and Prevalence Collaborators. Global, regional, Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET
and national incidence, prevalence, and years lived with disability for 328 diseases cohort. Microbiome 5, 4.
and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Hiroi, R., Carbone, D.L., Zuloaga, D.G., Bimonte-Nelson, H.A., Handa, R.J., 2016. Sex-
Burden of Disease Study 2016. Lancet 390 (10100), 1211–1259. dependent programming effects of prenatal glucocorticoid treatment on the devel-
Gensollen, T., Iyer, S.S., Kasper, D.L., Blumberg, R.S., 2016. How colonization by mi- oping serotonin system and stress-related behaviors in adulthood. Neuroscience 320,
crobiota in early life shapes the immune system. Science 352, 539–544. 43–56.
Gershon, A., Sudheimer, K., Tirouvanziam, R., Williams, L.M., O’Hara, R., 2013. The long- Hodes, G.E., Kana, V., Menard, C., 2015. Neuroimmune mechanisms of depression. Nat.
term impact of early adversity on late-life psychiatric disorders. Curr. Psychiatry Rep. Neurosci. 18, 1386–1393.
15 (4), 352. Horta, B.L., Gigante, D.P., Lima, R.C., Barros, F.C., CG, Victora, 2013. Birth by caesarean
Gibson, G.R., Hutkins, R., Sanders, M.E., Prescott, S.L., Reimer, R.A., Salminen, S.J., et al., section and prevalence of risk factors for non-communicable diseases in young adults:
2017. Expert consensus document: the International Scientific Association for a birth cohort study. PLoS One 8, e74301.
Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of Hotham, N., Hotham, E., 2015. Drugs in breastfeeding. Aust. Prescr. 38 (5), 156–159.
prebiotics. Nat. Rev. Gastroenterol. Hepatol. 14, 491–502. Hsiao, E.Y., McBride, S.W., Hsien, S., Sharon, G., Hyde, E.R., McCue, T., Codelli, J.A.,
Gilbert, J.A., Krajmalnik-Brown, R., Porazinska, D.L., Weiss, S.J., Knight, R., 2013. Chow, J., Reisman, S.E., Petrosino, J.F., Patterson, P.H., Mazmanian, S.K., 2013.
Toward effective probiotics for autism and other neurodevelopmental disorders. Cell Microbiota modulate behavioral and physiological abnormalities associated with
155, 1446–1448. neurodevelopmental disorders. Cell. 155 (7), 1451–1463.
Gill, S.R., Pop, M., DeBoy, R.T., Eckburg, P.B., Turnbaugh, P.J., Samuel, B.S., et al., 2006. Hsu, C.N., Lin, Y.J., Hou, C.Y., Tain, Y.L., 2018. Maternal administration of probiotic or
Metagenomic analysis of the human distal gut microbiome. Science 312, 1355–1359. prebiotic prevents male adult rat offspring against developmental programming of
Gitlin, D., Kumate, J., Morales, C., Noriega, L., Arévalo, N., 1972. The turnover of am- hypertension induced by high fructose consumption in pregnancy and lactation.
niotic fluid protein in the human conceptus. Am. J. Obstet. Gynecol. 113, 632–645. Nutrients. 10 (9), 1229.
Glazener, C.M., Abdalla, M., Stroud, P., Naji, S., Templeton, A., Russell, I.T., 1995. Huang, R., Wang, K., Hu, J., 2016. Effect of probiotics on depression: a systematic review
Postnatal maternal morbidity: extent, causes, prevention and treatment. Br. J. Obstet. and meta-analysis of randomized controlled trials. Nutrients 8, e483.
Gynaecol. 102, 282–287. Huang, T.T., Lai, J.B., Du YL, X.Y., Ruan, L.M., Hu, S.H., 2019. Current understanding of
Glover, M.E., Clinton, S.M., 2016. Of rodents and humans: a comparative review of the gut microbiota in mood disorders: an update of human studies. Front. Genet. 10, 98.
neurobehavioral effects of early life SSRI exposure in preclinical and clinical research. Huo, R., Zeng, B., Zeng, L., et al., 2017. Microbiota modulate anxiety-like behavior and
Int. J. Dev. Neurosci. 51, 50–72. endocrine abnormalities in hypothalamic-pituitary-adrenal axis. Front. Cell. Infect.
Gohir, W., Whelan, F.J., Surette, M.G., Moore, C., Schertzer, J.D., Sloboda, D.M., 2015. Microbiol. 7, 489.
Pregnancy-related changes in the maternal gut microbiota are dependent upon the Huttenhower, C., Gevers, D., Knight, R., Abubucker, S., Badger, J.H., et al., 2012. Human
mother’s periconceptional diet. Gut Microbes 6, 310–320. microbiome project consortium. structure, function and diversity of the healthy
Golubeva, A.V., Crampton, S., Desbonnet, L., Edge, D., O’Sullivan, O., Lomasney, K.W., human microbiome. Nature. 486 (7402), 207–214.
et al., 2015. Prenatal stress-induced alterations in major physiological systems cor- Jackson, M.A., Verdi, S., Maxan, M.E., Shin, C.M., Zierer, J., Bowyer, R.C.E., Martin, T.,
relate with gut microbiota composition in adulthood. Psychoneuroendocrinology 60, Williams, F.M.K., Menni, C., Bell, J.T., Spector, T.D., Steves, C.J., 2018. Gut micro-
58–74. biota associations with common diseases and prescription medications in a popula-
Goodrich, J.K., Davenport, E.R., Beaumont, M., Jackson, M.A., Knight, R., Ober, C., tion-based cohort. Nat. Commun. 9, 2655.
Spector, T.D., Bell, J.T., Clark, A.G., Ley, R.E., 2016. Genetic determinants of the gut Jakobsson, H.E., Abrahamsson, T.R., Jenmalm, M.C., Harris, K., Quince, C., Jernberg, C.,
microbiome in UK twins. Cell Host Microbe 19, 731–743. et al., 2014. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation
Goodrich, J.K., Waters, J.L., Poole, A.C., Sutter, J.L., Koren, O., Blekhman, R., Beaumont, and reduced Th1 responses in infants delivered by caesarean section. Gut 63,
M., Van Treuren, W., Knight, R., Bell, J.T., Spector, T.D., Clark, A.G., Ley, R.E., 2014. 559–566.
Human genetics shape the gut microbiome. Cell. 159, 789–799. Jašarević, E., Howard, C.D., Misic, A.M., Beiting, D.P., Bale, T.L., 2017. Stress during
Graf, A.E., Lallier, S.W., Waidyaratne, G., Thompson, M.D., Tipple, T.E., Hester, M.E., pregnancy alters temporal and spatial dynamics of the maternal and offspring mi-
et al., 2016. Maternal high fat diet exposure is associated with increased hepcidin crobiome in a sex-specific manner. Sci. Rep. 7, 44182.
levels, decreased myelination, and neurobehavioral changes in male offspring. Brain Jašarević, E., Howerton, C.L., Howard, C.D., Bale, T.L., 2015. Alterations in the vaginal
Behav. Immun. 58, 369–378. microbiome by maternal stress are associated with metabolic reprogramming of the
Grissom, N.M., George, R., Reyes, T.M., 2017. The hypothalamic transcriptional response offspring gut and brain. Endocrinology 156, 3265–3276.
to stress is severely impaired in offspring exposed to adverse nutrition during ge- Jeffcoat, M.K., Geurs, N.C., Reddy, M.S., Cliver, S.P., Goldenberg, R.L., Hauth, J.C., 2001.
station. Neuroscience 342, 200–211. Periodontal infection and preterm birth: results of a prospective study. J. Am. Dent.
Grüber, C., van Stuijvenberg, M., Mosca, F., Moro, G., Chirico, G., Braegger, C.P., et al., Assoc. 132, 875–880.
2010. Reduced occurrence of early atopic dermatitis because of immunoactive pre- Jiang, H., Ling, Z., Zhang, Y., 2015. Altered fecal microbiota composition in patients with
biotics among low-atopyrisk infants. J. Allergy Clin. Immunol. 126, 791–797. major depressive disorder. Brain Behav. Immun. 48, 186–194.
Grześkowiak, Ł, Sales Teixeira, T.F., Bigonha, S.M., Lobo, G., Salminen, S., Ferreira, C.L., Jiménez, E., Fernández, L., Marín, M.L., Martín, R., Odriozola, J.M., Nueno-Palop, C.,
2015. Gut Bifidobacterium microbiota in one-month-old Brazilian newborns. et al., 2005. Isolation of commensal bacteria from umbilical cord blood of healthy
Anaerobe 35, 54–58. neonates born by cesarean section. Curr. Microbiol. 51, 270–274.
Guarner, F., Bourdet-Sicard, R., Brandtzaeg, P., Gill, H.S., McGuirk, P., van Eden, W., Jiménez, E., Marín, M.L., Martín, R., Odriozola, J.M., Olivares, M., Xaus, J., Fernández, L.,
2006. Mechanisms of disease: the hygiene hypothesis revisited. Nat. Clin. Pract. Rodríguez, J.M., 2008. Is meconium from healthy newborns actually sterile? Res.
Gastroenterol. Hepatol. 3, 275–284. Microbiol. 159, 187–193.
Gundersen, T.D., Krebs, L., Loekkegaard, E.C.L., Rasmussen, S.C., Glavind, J., Clausen, Jimenez-Solem, E., 2014. Exposure to antidepressants during pregnancy–prevalences and
T.D., 2018. Postpartum urinary tract infection by mode of delivery: a Danish outcomes. Dan. Med. J. 61 (9), B4916.

726
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Jordan, H.V., Hammond, B.F., 1972. Filamentous bacteria isolated from human root challenge is an environmental risk factor for brain and behavior change relevant to
surface caries. Arch. Oral Biol. 17, 1333–1342. schizophrenia: evidence from MRI in a mouse model. PLoS One 4, e6354.
Juckel, G., Manitz, M.P., Brüne, M., Friebe, A., Heneka, M.T., Wolf, R.J., 2011. Microglial Lichtenstein, P., Yip, B.H., Björk, C., Pawitan, Y., Cannon, T.D., Sullivan, P.F., Hultman,
activation in a neuroinflammational animal model of schizophrenia—a pilot study. C.M., 2009. Common genetic determinants of schizophrenia and bipolar disorder in
Schizophr. Res. 131, 96–100. Swedish families: a population-based study. Lancet. 373 (9659), 234–239.
Karakuła-Juchnowicz, H., Dzikowski, M., Pelczarska, M., Dzikowska, I., Juchnowicz, D., Liu, Y., Qin, S., Song, Y., Feng, Y., Lv, N., Xue, Y., Liu, F., Wang, S., Zhu, B., Ma, J., Yang,
2016. The brain-gut axis dysfunctions and hypersensitivity to food antigens in the H., 2019. The perturbation of infant gut microbiota caused by cesarean delivery is
etiopathogenesis of schizophrenia. Psychiatr. Pol. 50, 747–760. partially restored by exclusive breastfeeding. Front. Microbiol. 10 (March), 598 ISSN:
Karlsson, F.H., Tremaroli, V., Nookaew, I., Bergström, G., Behre, C.J., Fagerberg, B., 1664-302X.
Nielsen, J., Bäckhed, F., 2013. Gut metagenome in European women with normal, Lloyd-Price, J., Abu-Ali, G., Huttenhower, C., 2016. The healthy human microbiome.
impaired and diabetic glucose control. Nature. 498 (7452), 99–103. Genome Med. 8, 51.
Kato-Kataoka, A., 2016. Fermented milk containing Lactobacillus casei strain Shirota Lopetuso, L.R., Scaldaferri, F., Franceschi, F., Gasbarrini, A., 2014. The gastrointestinal
preserves the diversity of the gut microbiota and relieves abdominal dysfunction in microbiome-functional interference between stomach and intestine. Best Pract. Res.
healthy medical students exposed to academic stress. Appl. Environ. Microbiol. 82, Cl GA 28 (6), 995–1002.
3649–3658. Lopez, N.J., Smith, P.C., Gutierrez, J., 2002. Higher risk of preterm birth and low birth
Keeney, K.M., Yurist-Doutsch, S., Arrieta, M.-C., Finlay, B.B., 2014. Effects of antibiotics weight in women with periodontal disease. J. Dent. Res. 81, 58–63.
on human microbiota and subsequent disease. Annu RevMicrobiol. 68, 217–235. Louveau, A., Smirnov, I., Keyes, T.J., Eccles, J.D., Rouhani, S.J., Peske, J.D., et al., 2015.
Kelly, C.J., Zheng, L., Campbell, E.L., Saeedi, B., Scholz, C.C., Bayless, A.J., Wilson, K.E., Structural and functional features of central nervous system lymphatic vessels.
Glover, L.E., Kominsky, D.J., Magnuson, A., Weir, T.L., Ehrentraut, S.F., Pickel, C., Nature 523, 337–341.
Kuhn, K.A., Lanis, J.M., Nguyen, V., Taylor, C.T., Colgan, S.P., 2015b. Crosstalk be- Lozupone, C.A., Stombaugh, J.I., Gordon, J.I., Jansson, J.K., Knight, R., 2012. Diversity,
tween microbiota-derived short-chain fatty acids and intestinal epithelial HIF aug- stability and resilience of the human gut microbiota. Nature 489, 220–230.
ments tissue barrier function. Cell Host Microbe 17 (5), 662–671. Lupp, C., Robertson, M.L., Wickham, M.E., Sekirov, I., Champion, O.L., Gaynor, E.C.,
Kelly, J.R., Kennedy, P.J., Cryan, J.F., Dinan, T.G., Clarke, G., Hyland, N.P., 2015a. et al., 2007. Host-mediated inflammation disrupts the intestinal microbiota and
Breaking down the barriers: the gut microbiome, intestinal permeability and stress- promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe 2, 119–129.
related psychiatric disorders. Front. Cell. Neurosci. 9, 392. Lyte, M., 2011. Probiotics function mechanistically as delivery vehicles for neuroactive
Kelly, J.R., Minuto, C., Cryan, J.F., Clarke, G., Dinan, T.G., 2017. Cross talk: the micro- compounds: microbial endocrinology in the design and use of probiotics. Bioessays.
biota and neurodevelopmental disorders. Front. Neurosci. 11, 490. 33, 574–581.
Keshavan, M.S., Hogarty, G.E., 1999. Brain maturational processes and delayed onset in Ma, J., Prince, A.L., Bader, D., Hu, M., Ganu, R., Baquero, K., et al., 2014. High-fat ma-
schizophrenia. Dev. Psychopathol. 11, 525–543. ternal diet during pregnancy persistently alters the offspring microbiome in a primate
Keshavan, M.S., 1999. Development, disease and degeneration in schizophrenia: a unitary model. Nat. Commun. 5, 3889.
pathophysiological model. J. Psychiatr. Res. 33, 513–521. MacFabe, D.F., Cain, N.E., Boon, F., Ossenkopp, K.P., Cain, D.P., 2011. Effects of the
Khachatryan, Z.A., Ktsoyan, Z.A., Manukyan, G.P., Kelly, D., Ghazaryan, K.A., Aminov, enteric bacterial metabolic product propionic acid on object-directed behavior, social
R.I., 2008. Predominant role of host genetics in controlling the composition of gut behavior, cognition, and neuroinflammation in adolescent rats: relevance to autism
microbiota. PLoS One 3 (8), e3064. spectrum disorder. Behav. Brain Res. 217, 47–54.
Khandaker, G.M., Pearson, R.M., Zammit, S., Lewis, G., Jones, P.B., 2014. Association of Macfabe, D.F., 2012. Short-chain fatty acid fermentation products of the gut microbiome:
serum interleukin 6 and c-reactive protein in childhood with depression and psy- implications in autism spectrum disorders. Microb. Ecol. Health Dis. 23.
chosis in young adult life: a population-based longitudinal study. JAMA Psychiatry Macfarlane, S., Macfarlane, G.T., 2003. Regulation of short-chain fatty acid production.
71, 1121–1128. Proc. Nutr. Soc. 62 (1), 67–72.
Kim, K.A., Gu, W., Lee, I.A., Joh, E.H., Kim, D.H., 2012. High fat diet-induced gut mi- MacIntyre, D.A., Chandiramani, M., Lee, Y.S., Kindinger, L., Smith, A., Angelopoulos, N.,
crobiota exacerbates inflammation and obesity in mice via the TLR4 signaling et al., 2015. The vaginal microbiome during pregnancy and the postpartum period in
pathway. PLoS One 7, e47713. a European population. Sci. Rep. 5, 8988.
Kim, Y.K., Shin, C., 2018. The microbiota-gut-brain axis in neuropsychiatric disorders: Mackay, D.F., Smith, G.C., Dobbie, R., Cooper, S.A., Pell, J.P., 2013. Obstetric factors and
pathophysiological mechanisms and novel treatments. Curr. Neuropharmacol. 16, different causes of special educational need: retrospective cohort study of 407,503
559–573. schoolchildren. BJOG 120, 297–307.
Kipnis, J., 2018. Immune system: the “seventh sense”. J. Exp. Med. 215, 397–398. Madan, J.C., Hoen, A.G., Lundgren, S.N., Farzan, S.F., Cottingham, K.L., Morrison, H.G.,
Klerman, G.L., Weissman, M.M., 1989. INcreasing rates of depression. JAMA 261, et al., 2016. Association of cesarean delivery and formula supplementation with the
2229–2235. intestinal microbiome of 6-weekold infants. JAMA Pediatr. 170, 212–219.
Koenig, J.E., Spor, A., Scalfone, N., Fricker, A.D., Stombaugh, J., Knight, R., et al., 2011. Madianos, P.N., Lieff, S., Murtha, A.P., et al., 2001. Maternal periodontitis and pre-
Succession of microbial consortia in the developing infant gut microbiome. Proc. maturity. Part II: maternal infection and fetal exposure. Ann. Periodontol. 6,
Natl. Acad. Sci. U. S. A. 108 (Suppl 1), 4578–4585. 175–182.
Kohman, R.A., Rhodes, J.S., 2013. Neurogenesis, inflammation and behavior. Brain Maes, M., Twisk, F.N., Kubera, M., Ringel, K., Leunis, J.C., Geffard, M., 2012. Increased
Behav. Immun. 27, 22–32. IgA responses to the LPS of commensal bacteria is associated with inflammation and
Kolde, R., Franzosa, E.A., Rahnavard, G., Hall, A.B., Vlamakis, H., Stevens, C., Daly, M.J., activation of cell-mediated immunity in chronic fatigue syndrome. J Affect Disorders.
Xavier, R.J., Huttenhower, C., 2018. Host genetic variation and its microbiome in- 136, 909–917.
teractions within the human microbiome project. Genome Med. 10 (1), 6. Magnúsdóttir, S., Heinken, A., Kutt, L., Ravcheev, D.A., Bauer, E., Noronha, A., et al.,
Koren, O., Goodrich, J.K., Cullender, T.C., Spor, A., Laitinen, K., Bäckhed, H.K., et al., 2017. Generation of genome-scale metabolic reconstructions for 773 members of the
2012. Host remodeling of the gut microbiome and metabolic changes during preg- human gut microbiota. Nat. Biotechnol. 35, 81–89.
nancy. Cell 150, 470–480. Majnik, A.V., Lane, R.H., 2015. The relationship between early-life environment, the
Kosten, T.A., Kim, J.J., Lee, H.J., 2012. Early life manipulations alter learning and epigenome and the microbiota. Epigenomics 7, 1173–1184.
memory in rats. Neurosci. Biobehav. Rev. 36 (9), 1985–2006. Malamitsi-Puchner, A., Protonotariou, E., Boutsikou, T., Makrakis, E., Sarandakou, A.,
Kostic, A.D., Gevers, D., Siljander, H., Vatanen, T., Hyotylainen, T., Hamalainen, A.M., Creatsas, G., 2005. The influence of the mode of delivery on circulating cytokine
et al., 2015. The dynamics of the human infant gut microbiome in development and concentrations in the perinatal period. Early Hum. Dev. 81, 387–392.
in progression toward type 1 diabetes. Cell Host Microbe 17, 260–273. Malkova, N.V., Yu, C.Z., Hsiao, E.Y., Moore, M.J., Patterson, P.H., 2012. Maternal im-
Kuperman, A.A., Koren, O., 2016. Antibiotic use during pregnancy: How bad is it? BMC mune activation yields offspring displaying mouse versions of the three core symp-
Med. 14, 91. toms of autism. Brain Behav. Immun. 26, 607–616.
Kurokawa, K., Itoh, T., Kuwahara, T., Oshima, K., Toh, H., Toyoda, A., et al., 2007. Marchesi, J.R., Adams, D.H., Fava, F., Hermes, G.D., Hirschfield, G.M., Hold, G., Quraishi,
Comparative metagenomics revealed commonly enriched gene sets in human gut M.N., Kinross, J., Smidt, H., Tuohy, K.M., Thomas, L.V., Zoetendal, E.G., Hart, A.,
microbiomes. DNA Res. 14, 169–181. 2016. The gut microbiota and host health: a new clinical frontier. Gut 65 (February
Ladd, C.O., Huot, R.L., Thrivikraman, K.V., Nemeroff, C.B., Meaney, M.J., Plotsky, P.M., (2)), 330–339. https://doi.org/10.1136/gutjnl-2015-309990. Epub 2015 Sep 2.
2000. Long-term behavioral and neuroendocrine adaptations to adverse early ex- Marques, T.M., Wall, R., Ross, R.P., Fitzgerald, G.F., Ryan, C.A., Stanton, C., 2010.
perience. Prog. Brain Res. 122, 81–103. Programming infant gut microbiota: influence of dietary and environmental factors.
Langdon, A., Crook, N., Dantas, G., 2016. The effects of antibiotics on the microbiome Curr. Opin. Biotechnol. 21 (2), 149–156.
throughout development and alternative approaches for therapeutic modulation. Martin, R., Makino, H., Cetinyurek Yavuz, A., Ben-Amor, K., Roelofs, M., Ishikawa, E.,
Genome Med. 8, 39. et al., 2016. Early-Life events, including mode of delivery and type of feeding, sib-
Latorre Uriza, C., Velosa-Porras, J., Roa, N.S., Quinones Lara, S.M., Silva, J., Ruiz, A.J., lings and gender, shape the developing gut microbiota. PLoS One 11, e0158498.
Escobar Arregoces, F.M., 2018. Periodontal disease, inflammatory cytokines, and Martinez, K.A., Devlin, J.C., Lacher, C.R., Yin, Y., Cai, Y., Wang, J., et al., 2017. Increased
PGE2 in pregnant patients at risk of preterm delivery: a pilot study. Infect. Dis. weight gain by C-section: functional significance of the primordial microbiome. Sci.
Obstet. Gynecol. (August), 1–7 ISSN: 1098-0997. Adv. 3, eaao1874.
Le Bastard, Q., Al-Ghalith, G.A., Grégoire, M., Chapelet, G., Javaudin, F., Dailly, E., et al., Maynard, T.M., Sikich, L., Lieberman, J.A., LaMantia, A.S., 2001. Neural development,
2018. Systematic review: human gut dysbiosis induced by non-antibiotic prescription cell-cell signaling, and the “two-hit” hypothesis of schizophrenia. Schizophr. Bull. 27,
medications. Aliment. Pharmacol. Ther. 47, 332–345. 457–476.
Lee, Y.A., Kim, Y.J., Goto, Y., 2016. Cognitive and affective alterations by prenatal and McKeen, S., Young, W., Mullaney, J., Fraser, K., McNabb, W.C., Roy, N.C., 2019. Infant
postnatal stress interaction. Physiol. Behav. 165, 146–153. complementary feeding of prebiotics for the microbiome and immunity. Nutrients 11
Ley, R.E., Peterson, D.A., Gordon, J.I., 2006. Ecological and evolutionary forces shaping (February (2)), 364 ISSN: 2072-6643.
microbial diversity in the human intestine. Cell 124, 837–848. McVey Neufeld, K.A., Kang, N., Bienenstock, J., et al., 2011. Reduced anxiety-like be-
Li, Q., Cheung, C., Wei, R., Hui, E.S., Feldon, J., Meyer, U., et al., 2009. Prenatal immune havior and central neurochemical change in germ-free mice. Neurogastroenterol.

727
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Motil. 23, 255–265. Exp. Med. 216 (1), 60–70. https://doi.org/10.1084/jem.20180199. Epub 2018
McVey Neufeld, K.A., Luczynski, P., Dinan, T.G., et al., 2016. Reframing the teenage Nov 30.
wasteland: adolescent microbiota-gut-brain axis. Can. J. Psychiatry 61, 214–221. O’Hara, A.M., Shanahan, F., 2006. The gut flora as a forgotten organ. EMBO Rep. 7,
Mennella, J.A., Beauchamp, G.K., 1998. Early flavor experiences: research update. Nutr. 688–693.
Rev. 56, 205–211. O’Mahony, S.M., Clarke, G., Borre, Y.E., Dinan, T.G., Cryan, J.F., 2015. Serotonin, tryp-
Merchant, A.T., Sutherland, M.W., Liu, J., Pitiphat, W., Dasanayake, A., 2018. Periodontal tophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 277, 32–48.
treatment among mothers with moderate periodontal disease and preterm birth: re- O’Mahony, S.M., Clarke, G., Dinan, T.G., Cryan, J.F., 2017. Early-life adversity and brain
analysis of OPT trial data accounting for selective survival. Int. J. Epidemiol. 47 (5), development: Is the microbiome a missing piece of the puzzle? Neuroscience. 342,
1670–1678. 37–54.
Messaoudi, M., Violle, N., Bisson, J.F., Desor, D., Javelot, H., Rougeot, C., 2011. Beneficial O’Mahony, S.M., Marchesi, J.R., Scully, P., Codling, C., Ceolho, A.M., Quigley, E.M., et al.,
psychological effects of a probiotic formulation (Lactobacillus helvecticus R0052 and 2009. Early life stress alters behavior, immunity, and microbiota in rats: implications
Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes 2 (4), for irritable bowel syndrome and psychiatric illnesses. Biol. Psychiatry 65, 263–267.
256–261. O’Neill, S.M., Curran, E.A., Dalman, C., et al., 2016. Birth by caesarean section and the
Messaoudi, M., 2011. Assessment of psychotropic-like properties of a probiotic for- risk of adult psychosis: a population-based cohort study. Schizophr. Bull. 42,
mulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats 633–641.
and human subjects. Br. J. Nutr. 105, 755–764. Oberbach, A., Haange, S.B., Schlichting, N., Heinrich, M., Lehmann, S., Till, H., et al.,
Methé, B.A., Nelson, K.E., Pop, M., Creasy, H.H., Giglio, M.G., et al., 2012. Human 2017. Metabolic in vivo labeling highlights differences of metabolically active mi-
Microbiome Project Consortium. A framework for human microbiome research. crobes from the mucosal gastrointestinal microbiome between high-fat and normal
Nature 486 (7402), 215–221. chow diet. J. Proteome Res. 16, 1593–1604.
Michalowicz, B.S., Novak, M.J., Hodges, J.S., DiAngelis, A., et al., 2009. Serum in- Ochoa-Repáraz, J., Mielcarz, D.W., Begum-Haque, S., Kasper, L.H., 2011. Gut, bugs, and
flammatory mediators in pregnancy: changes after periodontal treatment and asso- brain: role of commensal bacteria in the control of central nervous system disease.
ciation with pregnancy outcomes. J. Periodontol. 80, 1731–1741. Ann. Neurol. 69 (February (2)), 240–247.
Michalowicz, B.S., Hodges, J.S., DiAngelis, A.J., Lupo, V.R., Novak, M.J., Ferguson, J.E., Offenbacher, S., Jared, H.L., O’Reilly, P.G., Wells, S.R., et al., 1998. Potential pathogenic
Buchanan, W., Bofill, J., Papapanou, P.N., Mitchell, D.A., Matseoane, S., Tschida, mechanisms of periodontitis associated pregnancy complications. Ann. Periodontol.
P.A., 2006. N. Engl. J. Med. 355 (November (18)), 1885–1894. 3, 233–250.
Milani, C., Duranti, S., Bottacini, F., Casey, E., Turroni, F., Mahony, J., Belzer, C., Delgado Offenbacher, S., Katz, V., Fertik, G., et al., 1996. Periodontal infection as a possible risk
Palacio, S., Arboleya Montes, S., Mancabelli, L., Lugli, G.A., Rodriguez, J.M., Bode, L., factor for preterm low birth weight. J. Periodontol. 67, 1103–1113.
de Vos, W., Gueimonde, M., Margolles, A., van Sinderen, D., Ventura, M., 2017. The Offenbacher, S., Lieff, S., Boggess, K.A., et al., 2001. Maternal periodontitis and pre-
first microbial colonizers of the human gut: composition, activities, and health im- maturity. Part I: obstetric outcome of prematurity and growth restriction. Ann.
plications of the infant gut microbiota. Microbiol. Mol. Biol. Rev.: MMBR 81 (4). Periodontol. 6, 164–174.
Miller, A.H., Haroon, E., Raison, C.L., Felger, J.C., 2013. Cytokine targets in the brain: Offenbacher, S., Lin, D., Strauss, R., McKaig, R., et al., 2006. Effects of periodontal
impact on neurotransmitters and neurocircuits. Depress. Anxiety 30, 297–306. therapy during pregnancy on periodontal status, biologic parameters, and pregnancy
Miller, B.J., Buckley, P., Seabolt, W., Mellor, A., Kirkpatrick, B., 2011. Meta-analysis of outcomes: a pilot study. J Periodontolol. 77, 2011–2024.
cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol. Oh, J., Byrd, A.L., Park, M., Program, N.C.S., Kong, H.H., Segre, J.A., 2016. Temporal
Psychiatry 70 (7), 663–671. stability of the human skin microbiome. Cell. 165, 854–866.
Mitchell, C.M., Davy, B.M., Hulver, M.W., Neilson, A.P., Bennett, B.J., Davy, K.P., 2019. Okada, Y., Tsuzuki, Y., Sato, H., Narimatsu, K., Hokari, R., Kurihara, C., et al., 2013. Trans
Does exercise alter gut microbial composition? A systematic review. Med. Sci. Sports fatty acids exacerbate dextran sodium sulphate-induced colitis by promoting the up-
Exerc. 51 (1), 160–167. regulation of macrophage-derived proinflammatory cytokines involved in T helper 17
Mitchell-Lewis, D., Engebretson, S.P., Chen, J., Lamster, I.B., Papapanou, P.N., 2001. cell polarization. Clin. Exp. Immunol. 174, 459–471.
Periodontal infections and pre-term birth: early findings from a cohort of young Olszak, T., An, D., Zeissig, S., Vera, M.P., Richter, J., Franke, A., et al., 2012. Microbial
minority women in New York. Eur. J. Oral Sci. 109, 34–39. exposure during early life has persistent eff ;ects on natural killer T cell function.
Mittal, V.A., Ellman, L.M., Cannon, T.D., 2008. Gene-environment interaction and cov- Science 336, 489–493.
ariation in schizophrenia: the role of obstetric complications. Schizophr. Bull. 34 (6), Oskvig, D.B., Elkahloun, A.G., Johnson, K.R., Phillips, T.M., Herkenham, M., 2012.
1083–1094. Maternal immune activation by LPS selectively alters specific gene expression profiles
Monk, C., Georgieff, M.K., Osterholm, E.A., 2013. Research review: maternal prenatal of interneuron migration and oxidative stress in the fetus without triggering a fetal
distress and poor nutrition—mutually influencing risk factors affecting infant neu- immune response. Brain Behav. Immun. 26, 623–634.
rocognitive development. J. Child Psychol. Psychiatry 54, 115–130. Pantelis, C., Yucel, M., Wood, S.J., McGorry, P.D., Velakoulis, D., 2003. Early and late
Moreno-Küstner, B., Martín, C., Pastor, L., 2018. Prevalence of psychotic disorders and its neurodevelopmental disturbances in schizophrenia and their functional con-
association with methodological issues. A systematic review and meta-analyses. PLoS sequences. Aust. N. Z. J. Psychiatry 37, 399–406.
One 13 (4), e0195687. Parashar, A., Udayabanu, M., 2016. Gut microbiota regulates key modulators of social
Morgan, A.P., Crowley, J.J., Nonneman, R.J., Quackenbush, C.R., Miller, C.N., Ryan, A.K., behavior. Eur. Neuropsychopharmacol. 26 (1), 78–91.
Bogue, M.A., et al., 2014. The antipsychotic olanzapine interacts with gut micro- Pärtty, A., Kalliomäki, M., Wacklin, P., Salminen, S., Isolauri, E., 2015. A possible link
biome to cause weight gain in mouse. PLoS One 9 (12), e115225. between early probiotic intervention and the risk of neuropsychiatric disorders later
Moya-Pérez, A., Luczynski, P., Renes, I.B., Wang, S., Borre, Y., Ryan, C.A., Knol, J., in childhood: a randomized trial. Pediatr. Res. 77, 823–828.
Stanton, C., Dinan, T.G., Cryan, J.F., 2017. Intervention strategies for cesarean sec- Patel, R.V., et al., 2017. Whole genome sequencing resource identifies 18 new candidate
tion- induced alterations in the microbiota-gut-brain axis. Nutr. Rev. 75 (April (4)), genes for autism spectrum disorder. Nat. Neurosci. 20, 602–611.
225–240. Patterson, E., O’Doherty, R.M., Murphy, E.F., Wall, R., O’Sullivan, O., Nilaweera, K., et al.,
Moya-Pérez, A., Perez-Villalba, A., Benítez-Páez, A., Campillo, I., Sanz, Y., 2017b. 2014. Impact of dietary fatty acids on metabolic activity and host intestinal micro-
Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine biota composition in C57BL/6J mice. Br. J. Nutr. 111, 1905–1917.
and behavioral alterations in mice. Brain Behav. Immun. 65, 43–56. Pavlov, V.A., Tracey, K.J., 2017. Neural regulation of immunity: molecular mechanisms
Mueller, N.T., Bakacs, E., Combellick, J., Grigoryan, Z., Dominguez-Bello, M.G., 2015. and clinical translation. Nat. Neurosci. 20, 156–166.
The infant microbiome development: mom matters. Trends Mol. Med. 21, 109–117. Pedersen, A., Diedrich, M., Kaestner, F., Koelkebeck, K., Ohrmann, P., Ponath, G., Kipp,
Mueller, N.T., Mao, G., Bennet, W.L., Hourigan, S.K., Dominguez-Bello, M.G., Appel, L.J., F., Abel, S., Siegmund, A., Suslow, T., von Eiff, C., Arolt, V., Rothermundt, M., 2008.
Wang, X., 2017. Does vaginal delivery mitigate or strengthen the intergenerational Memory impairment correlates with increased S100B serum concentrations in pa-
association of overweight and obesity? Findings from the Boston Birth Cohort. Int. J. tients with chronic schizophrenia. Prog. Neuro Psychopharmacol. Biol. Psychiatr. 32
Obes. 41, 497–501. (8), 1789–1792.
Munoz-Bellido, J.L., Munoz-Criado, S., Garcìa-Rodrìguez, J.A., 2000. Antimicrobial ac- Pédron, T., Sansonetti, P., 2008. Commensals, bacterial pathogens and intestinal in-
tivity of psychotropic drugs: selective serotonin reuptake inhibitors. Int. J. flammation: an intriguing ménage à trois. Cell Host Microbe 3, 344–347.
Antimicrob. Agents 14, 177–180. Pei, Z., Heinrich, J., Fuertes, E., Flexeder, C., Hoffmann, B., Lehmann, I., Schaaf, B., von
Nagpal, R., Tsuji, H., Takahashi, T., Kawashima, K., Nagata, S., Nomoto, K., Yamashiro, Berg, A., Koletzko, S., 2014. Cesarean delivery and risk of childhood obesity. J.
Y., 2016. Sensitive quantitative analysis of the meconium bacterial microbiota in Pediatr. 164, 1068–1073 e1062.
healthy term infants born vaginally or by cesarean section. Front. Microbiol. 7. Pellizzon, M.A., Ricci, M.R., 2018. The common use of improper control diets in diet-
Nagpal, R., Tsuji, H., Takahashi, T., Nomoto, K., Kawashima, K., Nagata, S., Yamashiro, induced metabolic disease research confounds data interpretation: the fiber factor.
Y., 2017. Ontogenesis of the gut microbiota composition in healthy, full-term, vag- Nutr. Metab. (Lond) 15, 3.
inally born and breast-fed infants over the first 3 years of life: a quantitative bird’s- Penders, J., Thijs, C., Vink, C., Stelma, F.F., Snijders, B., Kummeling, I., van den Brandt,
eye view. Front. Microbiol. 8. P.A., Stobberingh, E.E., 2006. Factors influencing the composition of the intestinal
Naseribafrouei, A., Hestad, K., Avershina, E., 2014. Correlation between the human fecal microbiota in early infancy. Pediatrics 118 (2), 511–521.
microbiota and depression. Neurogastroenterol. Motil. 26, 1155–1162. Penders, J., Thijs, C., van den Brandt, P.A., Kummeling, I., Snijders, B., Stelma, F., et al.,
Natividad, J.M.M., Verdu, E.F., 2013. Modulation of intestinal barrier by intestinal mi- 2007. Gut microbiota composition and development of atopic manifestations in in-
crobiota: pathological and therapeutic implications. Pharmacol. Res. 69, 42–51. fancy: the KOALA Birth Cohort Study. Gut 56, 661–667.
Negele, K., Heinrich, J., Borte, M., Von Berg, A., Schaaf, B., Lehmann, I., Wichmann, H.E., Peng, L., Li, Z.R., Green, R.S., Holzman, I.R., Lin, J., 2009. Butyrate enhances the in-
Bolte, G., 2004. Mode of delivery and development of atopic disease during the first 2 testinal barrier by facilitating tight junction assembly via activation of AMP-activated
years of life. Pediatr. Allergy Immunol. 15, 48–54. protein kinase in Caco-2 cell monolayers. J. Nutr. 139 (9), 1619–1625.
Neish, A.S., 2009. Microbes in gastrointestinal health and disease. Gastroenterology. 136, Perez-Muñoz, M.E., Arrieta, M.C., Ramer-Tait, A.E., Walter, J., 2017. A critical assessment
65–80. of the “sterile womb” and “in utero colonization” hypotheses: implications for re-
Norris, G.T., Kipnis, J., 2019. Immune cells and CNS physiology: microglia and beyond. J. search on the pioneer infant microbiome. Microbiome 5, 48.

728
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Persaud, R., Azad, M.B., Konya, T., Guttman, D.S., Chari, R.S., Sears, M.R., et al., 2014. Saha, S., Chant, D., Welham, J., McGrath, J., 2005. A systematic review of the prevalence
Impact of perinatal antibiotic exposure on the infant gut microbiota at one year of of schizophrenia. PLoS Med. 2 (5), e141.
age. Allergy Asthma Clin. Immunol. 10 (Suppl 1), A31. Salminen, S., Gibson, G.R., McCartney, A.L., Isolauri, E., 2004. Influence of mode of
Pesonen, A.K., Eriksson, J.G., Heinonen, K., Kajantie, E., Tuovinen, S., Alastalo, H., delivery on gut microbiota composition in seven year old children. Gut 53,
Henriksson, M., Leskinen, J., Osmond, C., Barker, D.J., Räikkönen, K., 2013. 1388–1389.
Cognitive ability and decline after early life stress exposure. Neurobiol. Aging 34 (6), Sampson, T.R., Debelius, J.W., Thron, T., Janssen, S., Shastri, G.G., et al., 2016. Gut
1674–1679. microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s
Pirie, M., Linden, G., Irwin, C., 2013. Intrapregnancy nonsurgical periodontal treatment disease. Cell. 167 (6), 1469–1480 e12.
and pregnancy outcome: a randomized controlled trial. J. Periodontol. 84, Sampson, T.R., Mazmanian, S.K., 2015. Control of brain development, function, and
1391–1400. behavior by the microbiome. Cell Host Microbe 17, 565–576.
Polidano, C., Zhu, A., Bornstein, J.C., 2017. The relation between cesarean birth and child Sánchez, M.M., Ladd, C.O., Plotsky, P.M., 2001. Early adverse experience as a develop-
cognitive development. Sci. Rep. 7 (September (1)), 1–10 ISSN: 2045-2322. mental risk factor for later psychopathology: evidence from rodent and primate
Powell, C.M., Miyakawa, T., 2006. Schizophrenia-relevant behavioral testing in rodent models. Dev. Psychopathol. 13 (3), 419–449.
models: a uniquely human disorder? Biol. Psychiatry 59 (12), 1198–1207. Santacruz, A., Collado, M.C., García-Valdés, L., Segura, M.T., Martín-Lagos, J.A., Anjos,
Priyadarshini, M., Thomas, A., Reisetter, A.C., Scholtens, D.M., Wolever, T.M., Josefson, T., et al., 2010. Gut microbiota composition is associated with body weight, weight
J.L., et al., 2014. Maternal short-chain fatty acids are associated with metabolic gain, and biochemical parameters in pregnant women. Br. J. Nutr. 104, 83–92.
parameters in mothers and newborns. Transl. Res. 164, 153–157. Sarkar, A., Lehto, S.M., Harty, S., Dinan, T.G., Cryan, J.F., Burnet, P.W.J., 2016.
Pryce, C.R., Feldon, J., 2003. Long-term neurobehavioural impact of the postnatal en- Psychobiotics and the manipulation of bacteria–gut–brain signals. Trends Neurosci.
vironment in rats: manipulations, effects and mediating mechanisms. Neurosci. 39, 763–781.
Biobehav. Rev. 27 (1–2), 57–71. Saulnier, D.M., Ringel, Y., Heyman, M.B., Foster, J.A., Bercik, P., Shulman, R.J., et al.,
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K.S., et al., 2010. A human gut microbial 2013. The intestinal microbiome, probiotics and prebiotics in neurogastroenterology.
gene catalogue established by metagenomic sequencing. Nature 464 (7285), 59–65. Gut Microbes 4, 17–27.
Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., et al., 2012. A metagenome-wide association study of Scepanovic, P., Hodel, F., Mondot, S., Partula, V., Byrd, A., Hammer, C., Alanio, C.,
gut microbiota in type 2 diabetes. Nature 490 (7418), 55–60. Bergstedt, J., Patin, E., Touvier, M., Lantz, O., Albert, M.L., Duffy, D., Quintana-
Rackers, H.S., Thomas, S., Williamson, K., Posey, R., Kimmel, M.C., 2018. Emerging lit- Murci, L., Fellay, J., Milieu Intérieur Consortium, 2019. A comprehensive assessment
erature in the microbiota-brain Axis and perinatal mood and anxiety disorders. of demographic, environmental, and host genetic associations with gut microbiome
Psychoneuroendocrinology. 95, 86–96. diversity in healthy individuals. Microbiome 7 (1), 130.
Rajilić-Stojanović, M., Heilig, H.G., Molenaar, D., Kajander, K., Surakka, A., Smidt, H., de Schmidt, K., Cowen, P.J., Harmer, C.J., Tzortzis, G., Errington, S., Burnet, P.W., 2015.
Vos, W.M., 2009. Development and application of the human intestinal tract chip, a Prebiotic intake reduces the waking cortisol response and alters emotional bias in
phylogenetic microarray: analysis of universally conserved phylotypes in the abun- healthy volunteers. Psychopharmacology 232, 1793–1801.
dant microbiota of young and elderly adults. Environ. Microbiol. 11 (July (7)), Schroeder, B.O., Bäckhed, F., 2016. Signals from the gut microbiota to distant organs in
1736–1751. physiology and disease. Nat. Med. 22, 1079–1089.
Rana, S., Pugh, P.C., Jackson, N., Clinton, S.M., Kerman, I.A., 2015. Inborn stress re- Schwarz, E., Maukonen, J., Hyytiäinen, T., Kieseppä, T., Orešiè, M., Sabunciyan, S., et al.,
activity shapes adult behavioral consequences of early-life maternal separation stress. 2018. Analysis of microbiota in first episode psychosis identifies preliminary asso-
Neurosci. Lett. 584, 146–150. ciations with symptom severity and treatment response. Schizophr. Res. 192,
Rao, S., Srinivasjois, R., Patole, S., 2009. Prebiotic supplementation in full-term neonates: 398–403.
a systematic review of randomized controlled trials. Arch. Pediatr. Adolesc. Med. 163 Sela, D.A., Li, Y., Lerno, L., Wu, S., Marcobal, A.M., German, J.B., et al., 2011. An infant-
(8), 755–764. associated bacterial commensal utilizes breast milk sialyloligosaccharides. J. Biol.
Ravel, J., Gajer, P., Abdo, Z., Schneider, G.M., Koenig, S.S., McCulle, S.L., et al., 2011. Chem. 286, 11909–11918.
Vaginal microbiome of reproductive-age women. Proc. Natl. Acad. Sci. U. S. A. 108 Sender, R., Fuchs, S., 2016. Revised estimates for the number of human and bacteria cells
(Suppl 1), 4680–4687. in the body. PLoS Biol. 14, e1002533.
Rea, K., Dinan, T.G., Cryan, J.F., 2016. The microbiome: a key regulator of stress and Sender, R., Fuchs, S., Milo, R., 2016. Revised estimates for the number of human and
neuroinflammation. Neurobiol. Stress 4, 23–33. Bacteria cells in the body. PLoS Biol. 14 (8), e1002533.
Reber, S.O., Siebler, P.H., Donner, N.C., Morton, J.T., Smith, D.G., Kopelman, J.M., et al., Severance, E.G., Alaedini, A., Yang, S., Halling, M., Gressitt, K.L., Stallings, C.R., et al.,
2016. Immunization with a heat-killed preparation of the environmental bacterium 2012. Gastrointestinal inflammation and associated immune activation in schizo-
Mycobacterium vaccae promotes stress resilience in mice. Proc. Natl. Acad. Sci. U. S. phrenia. Schizophr. Res. 138, 48–53.
A. 113, E3130–E3139. Severance, E.G., Gressitt, K.L., Alaedini, A., Rohleder, C., Enning, F., Bumb, J.M., et al.,
Rhee, S.H., Pothoulakis, C., Mayer, E.A., 2009. Principles and clinical implications of the 2015. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow
brain-gut-enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 6, 306–314. dysfunction in first-episode schizophrenia. Brain Behav. Immun. 44, 148–158.
Rijkers, G.T., Bengmark, S., Enck, P., Haller, D., Herz, U., Kalliomaki, M., Kudo, S., Lenoir- Severance, E.G., Gressitt, K.L., Stallings, C.R., Katsafanas, E., Schweinfurth, L.A., Savage,
Wijnkoop, I., Mercenier, A., Myllyluoma, E., Rabot, S., Rafter, J., Szajewska, H., C.L., et al., 2016a. Candida albicans exposures, sex specificity and cognitive deficits
Watzl, B., Wells, J., Wolvers, D., Antoine, J.M., 2010. Guidance for substantiating the in schizophrenia and bipolar disorder. NPJ Schizophr. 2, 16018.
evidence for beneficial effects of probiotics: current status and recommendations for Severance, E.G., Gressitt, K.L., Stallings, C.R., Katsafanas, E., Schweinfurth, L.A., Savage,
future research. J. Nutr. 140 (3), 671–676. C.L., et al., 2017. Probiotic normalization of Candida albicans in schizophrenia: a
Romero, E., Ali, C., Molina-Holgado, E., Castellano, B., Guaza, C., Borrell, J., 2007. randomized, placebo-controlled, longitudinal pilot study. Brain Behav. Immun. 62,
Neurobehavioral and immunological consequences of prenatal immune activation in 41–45.
rats: influence of antipsychotics. Neuropsychopharmacology 32, 1791–1804. Severance, E.G., Yolken, R.H., Eaton, W.W., 2016. Autoimmune diseases, gastrointestinal
Romero, R., Hassan, S.S., Gajer, P., Tarca, A.L., Fadrosh, D.W., Nikita, L., et al., 2014. The disorders and the microbiome in schizophrenia: more than a gut feeling. Schizophr.
composition and stability of the vaginal microbiota of normal pregnant women is Res. 176, 23–35.
different from that of non-pregnant women. Microbiome 2, 4. Severance, E.G., Gressitt, K.L., Stallings, C.R., Origoni, A.E., Khushalani, S., Leweke, F.M.,
Rook, G.A., Lowry, C.A., 2008. The hygiene hypothesis and psychiatric disorders. Trends Dickerson, F.B., Yolken, R.H., 2013. Discordant patterns of bacterial translocation
Immunol. 29, 150–158. markers and implications for innate immune imbalances in schizophrenia. Schizophr.
Rook, G.A., Lowry, C.A., Raison, C.L., 2013. Microbial ‘Old Friends’, immunoregulation Res. 148 (1–3), 130–137.
and stress resilience. Evol. Med. Public Health 2013, 46–64. Shen, Y., Xu, J., Li, Z., Huang, Y., Yuan, Y., Wang, J., et al., 2018. Analysis of gut mi-
Rook, G.A., Martinelli, R., Brunet, L.R., 2003. Innate immune responses to mycobacteria crobiota diversity and auxiliary diagnosis as a biomarker in patients with schizo-
and the downregulation of atopic responses. Curr. Opin. Allergy Clin. Immunol. 3, phrenia: a cross-sectional study. Schizophr. Res. 197, 470–477.
337–342. Sherwin, E., Rea, K., Dinan, T.G., Cryan, J.F., 2016b. A gut (microbiome) feeling about
Rook, G.A., Raison, C.L., Lowry, C.A., 2014. Microbiota, immunoregulatory old friends the brain. Curr. Opin. Gastroenterol. 32 (2), 96–102.
and psychiatric disorders. Adv. Exp. Med. Biol. 817, 319–356. Sherwin, E., Sandhu, K.V., Dinan, T.G., Cryan, J.F., 2016a. May the force be with you: the
Rothschild, D., Weissbrod, O., Barkan, E., Kurilshikov, A., Korem, T., Zeevi, D., Costea, light and dark sides of the microbiota–gut–brain axis in neuropsychiatry. CNS Drugs
P.I., Godneva, A., Kalka, I.N., Bar, N., Shilo, S., Lador, D., Vila, A.V., Zmora, N., 30 (11), 1019–1041.
Pevsner-Fischer, M., Israeli, D., Kosower, N., Malka, G., Wolf, B.C., Avnit-Sagi, T., Shi, H., Kokoeva, M.V., Inouye, K., Tzameli, I., Yin, H., Flier, J.S., 2006. TLR4 links innate
Lotan-Pompan, M., Weinberger, A., Halpern, Z., Carmi, S., Fu, J., Wijmenga, C., immunity and fatty acid-induced insulin resistance. J. Clin. Invest. 116, 3015–3025.
Zhernakova, A., Elinav, E., Segal, E., 2018. Environment dominates over host genetics Silberman, D.M., Acosta, G.B., Zorrilla Zubilete, M.A., 2016. Long-term effects of early life
in shaping human gut microbiota. Nature. 555, 210–215. stress exposure: role of epigenetic mechanisms. Pharmacol. Res. 109, 64–73.
Round, J.L., Mazmanian, S.K., 2009. The gut microbiota shapes intestinal immune re- Silva, D., Colvin, L., Hagemann, E., Bower, C., 2014. Environmental risk factors by gender
sponses during health and disease. Nat. Rev. Immunol. 9, 313–323. associated with attention-deficit/hyperactivity disorder. Pediatrics. 133, e14–22.
Rutayisire, E., Huang, K., Liu, Y., Tao, F., 2016. The mode of delivery affects the diversity Slack, J., Gerencser, M., 1975. Actinomyces, Filamentous Bacteria. Biology and
and colonization pattern of the gut microbiota during the first year of infants’ life: a Pathogenicity. Burgess Publishing Co., Minneapolis, pp. 104.
systematic review. BMC Gastroenterol. 16, 86. Slavich, G.M., Way, B.M., Eisenberger, N.I., Taylor, S.E., 2010. Neural sensitivity to social
Rutayisire, E., Wu, X., Huang, K., Tao, S., Chen, Y., Tao, F., 2017. Childhood emotional rejection is associated with inflammatory responses to social stress. Proc. Natl. Acad.
and behavior problems and their associations with cesarean delivery. Revista Sci. U. S. A. 107, 14817–14822.
Brasileira De Psiquiatria (Sao Paulo, Brazil: 1999) 40 (October (2)), 145–153 ISSN: Smaill, F.M., Grivell, R.M., 2014. Antibiotic prophylaxis versus no prophylaxis for pre-
1809-452X. venting infection after cesarean section. Cochrane Database Syst. Rev. 10, CD007482.
Sachs, H.C., Drugs, C.O., 2013. The transfer of drugs and therapeutics into human breast Smilowitz, J.T., Lebrilla, C.B., Mills, D.A., German, J.B., Freeman, S.L., 2014. Breast milk
milk: an update on selected topics. Pediatrics 132, e796–e809. oligosaccharides: structure-function relationships in the neonate. Annu. Rev. Nutr.

729
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

34, 143–169. Udina, M., Castellvi, P., Moreno-Espana, J., Navines, R., Valdes, M., Forns, X., et al., 2012.
Smith, C.J., Emge, J.R., Berzins, K., Lung, L., Khamishon, R., Lung, L., et al., 2014. Interferon-induced depression in chronic hepatitis C: a systematic review and meta-
Probiotics normalize the gut-brain-microbiota axis in immunodeficient mice. Am. J. analysis. J. Clin. Psychiatry 73, 1128–1138.
Physiol. Gastrointest. Liver Physiol. 307, G793–G802.. Valkanova, V., Ebmeier, K.P., Allan, C.L., 2013. CRP, IL-6 and depression: a systematic
Socransky, S.S., Haffajee, A.D., Cugini, M.A., Smith, C., Kent Jr., R.L., 1998. Microbial review and meta-analysis of longitudinal studies. J. Affect. Disord. 150, 736–744.
complexes in subgingival plaque. J. Clin. Periodontol. 25, 134–144. Voreades, N., Kozil, A., Weir, T.L., 2014. Diet and the development of the human in-
Sohn, K., Underwood, M.A., 2017. Prenatal and postnatal administration of prebiotics and testinal microbiome. Front. Microbiol. 5, 494.
probiotics. Semin. Fetal Neonatal Med. 22 (5), 284–289. Wampach, L., Heintz-Buschart, A., Hogan, A., Muller, E.E.L., Narayanasamy, S., Laczny,
Srinivasjois, R., Rao, S., Patole, S., 2013. Prebiotic supplementation in preterm neonates: C.C., Hugerth, L.W., Bindl, L., Bottu, J., Andersson, A.F., et al., 2017. Colonization
updated systematic review and meta-analysis of randomised controlled trials. Clin. and succession within the human gut microbiome by archaea, bacteria, and micro-
Nutr. 32 (6), 958–965. eukaryotes during the first year of life. Front. Microbiol. 8.
Stecher, B., Robbiani, R., Walker, A.W., Westendorf, A.M., Barthel, M., Kremer, M., et al., Wang, J., Thingholm, L.B., Skiecevičienė, J., Rausch, P., Kummen, M., et al., 2016.
2007. Salmonella enterica serovar typhimurium exploits inflammation to compete Genome-wide association analysis identifies variation in vitamin D receptor and
with the intestinal microbiota. PLoS Biol. 5, 244. other host factors influencing the gut microbiota. Nat. Genet. 48, 1396–1406.
Steegenga, W.T., Mischke, M., Lute, C., Boekschoten, M.V., Lendvai, A., Pruis, M.G., et al., Wassenaar, T.M., Panigrahi, P., 2014. Is a foetus developing in a sterile environment?
2017. Maternal exposure to a Western-style diet causes differences in intestinal mi- Lett. Appl. Microbiol. 59 (December (6)), 572–579 ISSN: 1472-765X.
crobiota composition and gene expression of suckling mouse pups. Mol. Nutr. Food Wexler, H.M., 2007. Bacteroides: the good, the bad, and the nitty-gritty. Clin. Microbiol.
Res. 61 (January (1)). Rev. 20 (4), 593–621.
Steenbergen, L., 2015. A randomized controlled trial to test the effect of multispecies Wichman, C.L., Fothergill, A., Moore, K.M., Lang, T.R., Heise Jr, R.H., Watson, W.J.,
probiotics on cognitive reactivity to sad mood. Brain Behav. Immun. 48, 258–264. 2008. Recent trends in selective serotonin reuptake inhibitor use in pregnancy. J.
Stein, M.M., Hrusch, C.L., Gozdz, J., Igartua, C., Pivniouk, V., Murray, S.E., et al., 2016. Clin. Psychopharmacol. 28 (6), 714–716.
Innate immunity and asthma risk in amish and hutterite farm children. N. Engl. J. Williamson, L.L., McKenney, E.A., Holzknecht, Z.E., Belliveau, C., Rawls, J.F., Poulton, S.,
Med. 375, 411–421. et al., 2015. Got worms? Perinatal exposure to helminths prevents persistent immune
Stewart, C.J., Embleton, N.D., Clements, E., Luna, P.N., Smith, D.P., Fofanova, T.Y., sensitization and cognitive dysfunction induced by early-life infection. Brain Behav.
Nelson, A., Taylor, G., Orr, C.H., Petrosino, J.F., et al., 2017. Cesarean or vaginal Immun. 51, 14–28.
birth does not impact the longitudinal development of the gut microbiome in a cohort Willyard, C., 2018. Could baby’s first bacteria take root before birth? Nature 553,
of exclusively preterm infants. Front. Microbiol. 8, 1008. 264–266.
Stilling, R.M., Dinan, T.G., Cryan, J.F., 2014. Microbial genes, brain & behaviour - epi- Wong, M.L., Inserra, A., Lewis, M.D., Mastronardi, C.A., Leong, L., Choo, J., Kentish, S.,
genetic regulation of the gut-brain axis. Genes Brain Behav. 13, 69–86. Xie, P., Morrison, M., Wesselingh, S.L., Rogers, G.B., Licinio, J., 2016. Inflammasome
Stilling, R.M., Ryan, F.J., Hoban, A.E., et al., 2015. Microbes and neurodevelopment – signaling affects anxiety- and depressive-like behavior and gut microbiome compo-
absence of microbiota during early life increases activity-related transcriptional sition. Mol. Psychiatry 21 (6), 797–805.
pathways in the amygdala. Brain Behav. Immun. 50, 209–220. Woolhouse, H., Perlen, S., Gartland, D., Brown, S.J., 2012. Physical health and recovery
Stinson, L.F., Payne, M.S., Keelan, J.A., 2018. A critical review of the bacterial baptism in the first 18 months postpartum: Does cesarean section reduce long-term mor-
hypothesis and the impact of cesarean delivery on the infant microbiome. Front. Med. bidity? Birth 39, 221–229.
5 (May), 135 ISSN: 2296-858X. Wopereis, H., Oozeer, R., Knipping, K., Belzer, C., Knol, J., 2014. The first thousand days –
Stout, M.J., Conlon, B., Landeau, M., Lee, I., Bower, C., Zhao, Q., et al., 2013. intestinal microbiology of early life: establishing a symbiosis. Pediatr. Allergy
Identification of intracellular bacteria in the basal plate of the human placenta in Immunol. 25 (August (5)), 428–438 ISSN: 1399-3038.
term and preterm gestations. Am. J. Obstet. Gynecol. 208 (226), e1–e226 e7. Wright Jr, J.R., Stinson, D., Wade, A., Haldane, D., Heifetz, S.A., 1994. Necrotizing fu-
Strachan, D.P., 1989. Hay fever, hygiene, and household size. BMJ 299, 1259–1260. nisitis associated with Actinomyces meyeri infection: a case report. Pediatr. Pathol.
Sudo, N., Chida, Y., Aiba, Y., et al., 2004. Postnatal microbial colonization programs the 14, 927–934.
hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 558, Yang, C., Qu, Y., Fujita, Y., Ren, Q., Ma, M., Dong, C., et al., 2017. Possible role of the gut
263–275. microbiota-brain axis in the antidepressant effects of (R)-ketamine in a social defeat
Suhr, M.J., Hallen-Adams, H.E., 2015. The human gut mycobiome: pitfalls and po- stress model. Transl. Psychiatry 7, 1294.
tentials–a mycologist’s perspective. Mycologia 107, 1057–1073. Yassour, M., Vatanen, T., Siljander, H., Hämäläinen, A.M., Härkönen, T., Ryhänen, S.J.,
Sullivan, P.F., Kendler, K.S., Neale, M.C., 2003. Schizophrenia as a complex trait: evi- et al., 2016. Natural history of the infant gut microbiome and impact of antibiotic
dence from a meta-analysis of twin studies. Arch. Gen. Psychiatry 60 (12), treatment on bacterial strain diversity and stability. Sci. Transl. Med. 8, 343ra381.
1187–1192. Yatsunenko, T., Rey, F.E., Manary, M.J., Trehan, I., Dominguez-Bello, M.G., Contreras,
Thavagnanam, S., Fleming, J., Bromley, A., Shields, M.D., Cardwell, C.R., 2008. A meta- M., et al., 2012. Human gut microbiome viewed across age and geography. Nature
analysis of the association between Caesarean section and childhood asthma. Clin. 486, 222–227.
Exp. Allergy 38, 629–633. Yokoe, D.S., Christiansen, C.L., Johnson, R., Sands, K.E., Livingston, J., Shtatland, E.S.,
The Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium, Platt, R., 2001. Epidemiology of and surveillance for postpartum infections. Emerg
2011. Genome-wide association study identifies five new schizophrenia loci. Nat. Infect Dis. 7 (5), 837–841.
Genet. 43 (10), 969–976. Yolken, R.H., Severance, E.G., Sabunciyan, S., Gressitt, K.L., Chen, O., Stallings, C.R.,
Thum, C., Cookson, A.L., Otter, D.E., McNabb, W.C., Hodgkinson, A.J., Dyer, J., et al., et al., 2015. Metagenomic sequencing indicates that the oropharyngeal phageome of
2012. Can nutritional modulation of maternal intestinal microbiota influence the individuals with schizophrenia differs from that of controls. Schizophr. Bull. 41,
development of the infant gastrointestinal tract? J. Nutr. 142 (11), 1921–1928. 1153–1161.
Tillisch, K., Labus, J., Kilpatrick, L., Jiang, Z., Stains, J., Ebrat, B., Guyonnet, D., Legrain- Yonkers, K.A., Wisner, K.L., Stewart, D.E., Oberlander, T.F., Dell, D.L., Stotland, N.,
Raspaud, S., Trotin, B., Naliboff, B., Mayer, E.A., 2013. Consumption of fermented Ramin, S., Chaudron, L., Lockwood, C., 2009. The management of depression during
milk product with probiotic modulates brain activity. Gastroenterology 144, pregnancy: a report from the American Psychiatric Association and the American
1394–1401. College of Obstetricians and Gynecologists. Obstet. Gynecol. 114 (3), 703–713.
Tita, A.T., Landon, M.B., Spong, C.Y., Lai, Y., Leveno, K.J., Varner, M.W., Moawad, A.H., Yuan, X., Zhang, P., Wang, Y., Liu, Y., Li, X., Kumar, B.U., et al., 2018. Changes in me-
Caritis, S.N., Meis, P.J., Wapner, R.J., Sorokin, Y., Miodovnik, M., Carpenter, M., tabolism and microbiota after 24-week risperidone treatment in drug naïve, normal
Peaceman, A.M., O’Sullivan, M.J., Sibai, B.M., Langer, O., Thorp, J.M., Ramin, S.M., weight patients with first episode schizophrenia. Schizophr. Res S0920–9964(18)
Mercer, B.M., 2009. Timing of elective repeat cesarean delivery at term and neonatal 30274-3.
outcomes. N. Engl. J. Med. 360, 111–120. Zeevi, D., Korem, T., Zmora, N., Israeli, D., Rothschild, D., Weinberger, A., Ben-Yacov, O.,
Tomasik, J., Yolken, R.H., Bahn, S., Dickerson, F.B., 2015. Immunomodulatory effects of Lador, D., Avnit-Sagi, T., Lotan-Pompan, M., Suez, J., Mahdi, J.A., Matot, E., Malka,
probiotic supplementation in schizophrenia patients: a randomized, placebo-con- G., Kosower, N., Rein, M., Zilberman-Schapira, G., Dohnalová, L., Pevsner-Fischer,
trolled trial. Biomark. Insights 10, 47–54. M., Bikovsky, R., Halpern, Z., Elinav, E., Segal, E., 2015. Personalized nutrition by
Tomova, A., Husarova, V., Lakatosova, S., Bakos, J., Vlkova, B., Babinska, K., et al., 2015. prediction of glycemic responses. Cell. 163, 1079–1094.
Gastrointestinal microbiota in children with autism in Slovakia. Physiol. Behav. 138, Zheng, L., Kelly, C.J., Battista, K.D., Schaefer, R., Lanis, J.M., Alexeev, E.E., Wang, R.X.,
179–187. Onyiah, J.C., Kominsky, D.J., Colgan, S.P., 2017a. Microbial-derived butyrate pro-
Trahair, J., 2001. Digestive system. In: Harding, R., Bocking, A.D. (Eds.), Fetal Growth motes epithelial barrier function through IL-10 receptor-dependent repression of
and Development. Cambridge University Press, Cambridge, UK, pp. 137–153 ISBN Claudin-2. J. Immunol. 199 (8), 2976–2984.
0521645433. Zheng, P., Cheng, K., Zeng, L., Zhou, C.J., Xie, P., 2017b. A new pathway for the gut
Tun, H.M., Bridgman, S.L., Chari, R., Field, C.J., Guttman, D.S., Becker, A.B., et al., 2018. microbiota to modulate the brain: activation of pattern-recognition receptors by
Roles of birth mode and infant gut microbiota in intergenerational transmission of microbial products. Mol. Psychiatry 22 (2), 162–163.
overweight and obesity from mother to offspring. JAMA Pediatr. 172, 368–377. Zheng, P., Zeng, B., Liu, M., Chen, J., Pan, J., Han, Y., Liu, Y., Cheng, K., Zhou, C., Wang,
Turnbaugh, P.J., Hamady, M., Yatsunenko, T., Cantarel, B.L., Duncan, A., Ley, R.E., Sogin, H., Zhou, X., Gui, S., Perry, S.W., Wong, M.L., Licinio, J., Wei, H., Xie, P., 2019. The
M.L., Jones, W.J., Roe, B.A., Affourtit, J.P., Egholm, M., Henrissat, B., Heath, A.C., gut microbiome from patients with schizophrenia modulates the glutamate-gluta-
Knight, R., Gordon, J.I., 2009. A core gut microbiome in obese and lean twins. mine-GABA cycle and schizophrenia-relevant behaviors in mice. Sci. Adv. 5 (2),
Nature. 457 (7228), 480–484. eaau8317.
Turpin, W., Espin-Garcia, O., Xu, W., Silverberg, M.S., Kevans, D., Smith, M.I., Guttman, Zheng, P., Zeng, B., Zhou, C., Liu, M., Fang, Z., Xu, X., Zeng, L., Chen, J., Fan, S., Du, X.,
D.S., Griffiths, A., Panaccione, R., Otley, A., Xu, L., Shestopaloff, K., Moreno- Zhang, X., Yang, D., Yang, Y., Meng, H., Li, W., Melgiri, N.D., Licinio, J., Wei, H., Xie,
Hagelsieb, G., GEM Project Research Consortium, Paterson, A.D., Croitoru, K., 2016. P., 2016. Gut microbiome remodeling induces depressive-like behaviors through a
Association of host genome with intestinal microbial composition in a large healthy pathway mediated by the host’s metabolism. Mol. Psychiatry 21 (6), 786–796.
cohort. Nat. Genet. 48, 1413–1417. Zhernakova, A., Kurilshikov, A., Bonder, M.J., Tigchelaar, E.F., Schirmer, M., Vatanen, T.,

730
B. Golofast and K. Vales Neuroscience and Biobehavioral Reviews 108 (2020) 712–731

Mujagic, Z., Vila, A.V., Falony, G., Vieira-Silva, S., Wang, J., Imhann, F., Brandsma, Zhuang, L., Chen, H., Zhang, S., Zhuang, J., Li, Q., Feng, Z., 2019. Intestinal microbiota in
E., Jankipersadsing, S.A., Joossens, M., Cenit, M.C., Deelen, P., Swertz, M.A., early life and its implications on childhood health. Microbiome and Health,
Weersma, R.K., Feskens, E.J., Netea, M.G., Gevers, D., Jonkers, D., Franke, L., Genomics, Proteomics & Bioinformatics. 17 (February (1)), 13–25.
Aulchenko, Y.S., Huttenhower, C., Raes, J., Hofker, M.H., Xavier, R.J., Wijmenga, C., Zijlmans, M.A., Korpela, K., Riksen-Walraven, J.M., de Vos, W.M., de Weerth, C., 2015.
Fu, J., 2016. Populationbased metagenomics analysis reveals markers for gut mi- Maternal prenatal stress is associated with the infant intestinal microbiota.
crobiome composition and diversity. Science. 352, 565–569. Psychoneuroendocrinology 53, 233–245.

731

You might also like