Skip to main content
Frontiers in Neuroscience logoLink to Frontiers in Neuroscience
. 2017 Sep 15;11:490. doi: 10.3389/fnins.2017.00490

Cross Talk: The Microbiota and Neurodevelopmental Disorders

John R Kelly 1,2, Chiara Minuto 1,2, John F Cryan 2,3, Gerard Clarke 1,2, Timothy G Dinan 1,2,*
PMCID: PMC5605633  PMID: 28966571

Abstract

Humans evolved within a microbial ecosystem resulting in an interlinked physiology. The gut microbiota can signal to the brain via the immune system, the vagus nerve or other host-microbe interactions facilitated by gut hormones, regulation of tryptophan metabolism and microbial metabolites such as short chain fatty acids (SCFA), to influence brain development, function and behavior. Emerging evidence suggests that the gut microbiota may play a role in shaping cognitive networks encompassing emotional and social domains in neurodevelopmental disorders. Drawing upon pre-clinical and clinical evidence, we review the potential role of the gut microbiota in the origins and development of social and emotional domains related to Autism spectrum disorders (ASD) and schizophrenia. Small preliminary clinical studies have demonstrated gut microbiota alterations in both ASD and schizophrenia compared to healthy controls. However, we await the further development of mechanistic insights, together with large scale longitudinal clinical trials, that encompass a systems level dimensional approach, to investigate whether promising pre-clinical and initial clinical findings lead to clinical relevance.

Keywords: microbiota, microbiome, gut-brain axis, immune system, social cognition, autism, schizophrenia, psychobiotics

Introduction

From an evolutionary-based perspective, the host and its microbiome evolved as a cooperative unit (Rosenberg et al., 2007; Zilber-Rosenberg and Rosenberg, 2008; Martin et al., 2015; Douglas and Werren, 2016). All stages in the evolution of the human brain occurred within this microbial ecosystem (McFall-Ngai et al., 2013; Bordenstein and Theis, 2015). The predominant theory to account for the evolution of the enlargement of the human brain implicates social interaction. Brain areas such as the prefrontal cortex and the amygdala have undergone pronounced changes in the evolution of social mammals (Kolb et al., 2012; Janak and Tye, 2015). Brains of social species exhibit a set of features that need to integrate for group living to become advantageous, and the development of the complex neural circuitry underlying social and emotional cognition is of fundamental importance to neurodevelopmental disorders, such as ASD and schizophrenia (Adolphs, 2001; Lederbogen et al., 2011; Janak and Tye, 2015; Averbeck and Costa, 2017).

Neurodevelopment requires the intricate interplay of genetic expression, influenced by pre-and post-natal environmental events. Critical periods or “windows” of brain development exist, during which time neural circuits are particularly sensitive to, and require, the influence of appropriate environmental inputs, in order to develop properly. Human brain development begins in the third gestational week (Stiles and Jernigan, 2010) and at time of birth approximately 86 billion neurons (Azevedo et al., 2009) with up to 100 trillion connections are produced. These connections form simple circuits, and when reinforced through repeated use, under the influence of environmental cues, form more complex interconnected circuits, leading to complex networks (Bassett and Sporns, 2017). The developmental trajectory of social, emotional and cognitive brain domains occur in parallel, though social cognition may be linked to certain specific subnetworks (Dunbar, 2012; Sliwa and Freiwald, 2017).

The development of this neural circuitry requires precise regulation from molecular signaling pathways. Hormones, such as oxytocin (Kirsch et al., 2005), neurotransmitters, such as serotonin (Whitaker-Azmitia, 2001), and the immune system (Bilbo et al., 2012), all play pivotal roles in sculpting the neural circuitry underlying social cognition, emotion and behavior. Many of the brain regions involved and the molecular substrates subserving relevant functions are also responsive to microbiome-gut-brain axis signaling (Clarke et al., 2013; Semple et al., 2013; Montiel et al., 2014; Dinan et al., 2015; Erny et al., 2015; Buffington et al., 2016; Vuong and Hsiao, 2017) Figure 1.

Figure 1.

Figure 1

The microbiome-gut-brain axis in psychiatry. A number of factors have an influence on the assembly, composition and stability of the gut microbiota including mode of birth, lifestyle factors such as diet and exercise, and stress. These factors could thus impact signaling along the microbiome-gut-brain axis, which has been implicated in a variety of behavioral features relevant to schizophrenia and autism including anxiety and cognition. This impact may be underpinned by microbial regulation of the host immune system, CNS BDNF expression and microglial activation states.

The trajectory of early post-natal brain development overlaps with the acquisition and reorganization of the gut microbiota (Borre et al., 2014; Chu et al., 2017). The gut microbiota in the initial days of life is unstable and of low diversity (Arrieta et al., 2014). By age three, a stage by which verbal communication and Theory of Mind develops (the ability to infer and reason about the intentions, emotions and thoughts of others) (Grosse Wiesmann et al., 2017), the gut microbiota composition stabilizes into a pattern that more resembles an adult-like profile (Voreades et al., 2014). These social, cognitive and emotional domains, and their neurodevelopment, are compromised in neurodevelopmental disorders, such as ASD and schizophrenia. Deciphering the gut microbiota compositional trajectories and profiles, corresponding metabolic output and precise signaling pathways that play a pertinent role in molding the neural circuitry underlying the social-communicative domains of the brain, is one of the great challenges of modern neuroscience (Chen et al., 2013; Mayer et al., 2014a; Dinan and Cryan, 2017; Sherwin et al., 2017).

Autism spectrum disorders (ASD)

ASD is a heterogeneous neurodevelopmental disorder, affecting approximately 1 in 68 children (Christensen et al., 2016). It is characterized by deficits in social communication, social interaction and restricted/repetitive behavioral patterns. The processing of emotional stimuli, be it language or facial expressions is impaired in individuals with ASD (Dalton et al., 2005; Preissler and Carey, 2005; Monk et al., 2010; Lartseva et al., 2014; Neuhaus et al., 2016; Wang and Adolphs, 2017). Therefore, deficits in social communication, together with emotional processing, and a lack of social interest in communication can result in language delay, and a proportion of children at the severe end of the spectrum will not develop language abilities (Landry and Loveland, 1988).

The heritability of ASD is estimated at between 64 and 91% (Tick et al., 2016), and genes that encode proteins for synaptic formation, microglial function, transcriptional regulation and chromatin-remodeling pathways are implicated (De Rubeis et al., 2014; Parikshak et al., 2016). New mutations contribute to the risk, and a recent large scale study, showed that approximately one third of spontaneous, non-inherited genetic mutations found in people with ASD were also found in the general population (Kosmicki et al., 2017). A recent study suggests that those ASD children with de novo mutations show relative strengths in verbal and language abilities, including a smaller discrepancy between non-verbal and verbal IQ and a greater likelihood of having achieved fluent language, relative to those with no identified genetic abnormalities (Bishop et al., 2017). Taken together, these genetic studies in ASD highlight the neurodiversity of the disorder (Baron-Cohen, 2017; Vorstman et al., 2017; Yuen et al., 2017).

The origins of ASD are likely to occur during the prenatal timeframe, a time window during which important connections are formed (Willsey et al., 2013). A study, using a high-resolution transcriptional atlas in primates, showed that many ASD-related genes are activated in new-born neurons during prenatal development, while schizophrenia related genes are activated from infancy through adulthood (Bakken et al., 2016). Maternal infections during pregnancy are associated with the development of neurodevelopmental disorders (Atladottir et al., 2010; Jiang et al., 2016; Careaga et al., 2017). Certain subtypes of ASD are associated with increased levels of maternal peripheral chemokines and cytokines during gestation (Goines et al., 2011; Jones et al., 2017; Graham et al., in press). Moreover, subgroups of children diagnosed with ASD have elevated levels of peripheral cytokines (Ashwood et al., 2011), and microglial activation in young adults with ASD has been demonstrated using positron emission tomography (PET) imaging (Suzuki et al., 2013). Progress is being made in diagnosing infants at high risk of developing ASD, by utilizing imaging techniques such as fMRI (Hazlett et al., 2017; Shen et al., 2017). A recent structural and diffusion MRI study of 3 year old infants diagnosed with Neurodevelopmental Disorders (32 ASD and 16 other developmental disorders, including intellectual disability and language disorder) reported an over-connectivity pattern in ASD in networks primarily involving the fronto-temporal nodes, known to be crucial for social-skill development (Conti et al., 2017).

Risk factors such as advanced parental age (Durkin et al., 2008; Sandin et al., 2016), low birth weight (Schendel and Bhasin, 2008) and multiple births (Croen et al., 2002) have been identified, while others such as mode of birth have been advanced. However, epidemiological data suggests that C-section mode of delivery, currently far in excess of WHO recommendations (WHO, 2015) and known to alter microbiome signatures (see below), is associated only with a slightly increased risk of ASD and that this may be due to familial confounds (Curran et al., 2014, 2015, 2016; O'Neill et al., 2015). Identifying additional modifiable environmental factors that play a causal role in ASD, particularly during the prenatal and early post-natal period, is of vital importance.

Schizophrenia spectrum disorders (SSD)

Schizophrenia is a heterogeneous neurodevelopmental disorder, with a general population lifetime prevalence of approximately 0.87% (Perala et al., 2007), and an annual incidence of approximately 0.20/1,000/year (Messias et al., 2007). There is a slightly greater risk for males (Aleman et al., 2003) and psychotic symptoms usually manifest clinically during the adolescent period. This disorder can have a major detrimental impact on functioning, and is associated with a reduced life expectancy (Laursen et al., 2014; Schoenbaum et al., 2017; Strati et al., 2017) and a suicide rate of 5% (Hor and Taylor, 2010). schizophrenia is classically characterized by positive (delusions, hallucinations), negative (affective flattening, alogia, and avolition), and cognitive symptoms (Aleman et al., 1999; Kahn and Keefe, 2013; Schaefer et al., 2013). Social interaction and communication deficiencies, including disorganized speech, can be prominent, even early in the course of this disorder (Sullivan et al., 2003; Roche et al., 2016; Morgan et al., 2017).

Similar to ASD, the precise cause of schizophrenia is unknown. A complex and dynamic bidirectional interaction of genomic and environmental factors converge to shape the trajectory of schizophrenia (O'Tuathaigh et al., 2017). Prenatal and early post-natal environmental factors sensitize the vulnerable brain. Although psychotic symptoms usually manifest during the adolescent period, it has been established that schizophrenia is associated with poor premorbid functioning, cognitive impairment, and social deficits prior to the onset of psychotic symptoms (Schenkel and Silverstein, 2004). Indeed, previously considered distinct forms of psychopathology may in fact have characteristics in common, and exhibit age adjusted variations of common underlying dispositions (Casey et al., 2014; Hommer and Swedo, 2015).

The immune system is an important player in the pathophysiology of schizophrenia (Benros et al., 2012; Feigenson et al., 2014; Muller, 2014). At the genetic level, genes related to B-lymphocyte lineages involved in acquired immunity (CD19 and CD20 lines) and major histocompatibility complex locus have been linked to schizophrenia (Corvin and Morris, 2014; Schizophrenia Working Group of the Psychiatric Genomics Consortium., 2014). A recent translational study implicated excessive complement activity, particularly the role of C4 in mediating synapse elimination during post-natal development (Sekar et al., 2016). Neuro-immune signaling also changes during the adolescent period, regulating changes in synaptic pruning, neurite outgrowth, and neurotransmitter release via Blood Brain Barrier (BBB) dynamics and glial activity (Brenhouse and Schwarz, 2016). It is established that subgroups of patients with schizophrenia have elevated levels of peripheral cytokines, including subgroups of medication free first-episode psychosis individuals (Miller et al., 2011; Di Nicola et al., 2013; de Witte et al., 2014; Upthegrove et al., 2014). There is also some suggestion, though not well established, that schizophrenia is associated with altered intestinal (Severance et al., 2013) and blood brain barrier function (Pollak et al., 2017). This, taken together with altered microglial activation in psychosis patients (Bloomfield et al., 2016), highlights the key role of the immune system, in at least subgroups of individuals with psychosis (Al-Diwani et al., 2017).

Infections at different stages of brain development result in varying degrees of lifelong changes in behavior and cognition (Spencer and Meyer, 2017). Certain infections are known to increase the risk of schizophrenia (Meyer et al., 2009; Brown, 2012). A large epidemiological study (n = 1,015,447), showed that treatment with anti-infective agents (primarily driven by infections treated with antibiotics), were associated with an increased risk of schizophrenia by a hazard rate ratio of 1.37 (Kohler et al., 2017). However, an earlier study found an increased risk for mood and anxiety disorders for antibiotic exposure, but no change in risk for psychosis with any antibiotic group (Lurie et al., 2015). An infection with a robust link to psychosis is the protozoan Toxoplasma gondii (Torrey and Yolken, 2003; Severance et al., 2016b). A meta-analysis of 16 studies demonstrated increased T. gondii IgM levels in patients with acute psychosis (Monroe et al., 2015). The mechanism is not completely understood, but a putative role of attenuated CD8 T-cell response in T. gondii seropositive individuals has been suggested (Bhadra et al., 2013). It is known that T. gondii induces the production of a variety of cytokines by microglia, astrocytes, and neurons (Carruthers and Suzuki, 2007). Monocytes and dendritic cells are the most important candidates for the transport of T. gondii from the periphery to the immunologically privileged sites of the brain (Feustel et al., 2012).

Indeed, latent T. gondii infection is associated with an upregulation of cerebral complement factor C1q (Xiao et al., 2016). Furthermore, T. gondii infection can alter dopamine metabolism (Prandovszky et al., 2011) and latent T. gondii is associated with reduced psychomotor performance (Havlicek et al., 2001). More recently, T. gondii has been shown to lead to deficits in goal-directed behavior in healthy elderly individuals (Beste et al., 2014). Interestingly, acute T. gondii infection can affect the gut microbiota in mice (Molloy et al., 2013). Although only a minor subset of T. gondii seropositive individuals develop serious mental impairments, taken together, the example of T. gondii, suggests that microbial agents contribute to the vulnerability to the development of subgroups of schizophrenia (Yolken and Torrey, 2008). Although we have focussed on T. gondii, it is noteworthy that other infections, such as Human Herpesvirus 2, Borna Disease Virus Human Endogenous Retrovirus W, Chlamydophila pneumoniae, and Chlamydophila psittaci are also associated with the disorder (Arias et al., 2012). It remains an open question whether there is a common mechanism through which these microbes exert their influence, albeit, that one shared general feature in most examples cited is an intracellular life stage.

Interestingly, urbanicity, known to affect microbial diversity and impact the overall functionality of the gut microbiome (Mancabelli et al., 2017), is also a risk factor for the development of schizophrenia (Pedersen and Mortensen, 2001; Krabbendam and van Os, 2005; Peen et al., 2010; Vassos et al., 2012; Newbury et al., 2016). In healthy individuals, a negative correlation was found between early-life urbanicity and gray matter volume in the right dorsolateral prefrontal cortex in males and females, and in perigenual anterior cingulate cortex volumes, a key region for regulation of amygdala, in men only (Haddad et al., 2015). Using fMRI, city living was associated with increased amygdala activity (Lederbogen et al., 2011), known to be associated with schizophrenia (Aleman and Kahn, 2005; Rasetti et al., 2009). Stamper and colleagues postulate that differential exposure to microbes in the urban compared to the rural environment interact with differences in social stressors to alter social stress neural circuitry (Stamper et al., 2016).

ASD and microbiota

GI symptoms are a common comorbidity in ASD (Molloy and Manning-Courtney, 2003; Buie et al., 2010; Berding and Donovan, 2016). However, the underlying mechanism is not fully known (Mayer et al., 2014b). The vast majority of human studies show that ASD is associated with altered microbial profiles (see Table 1). A systematic review of gut microbiota alterations in ASD, verified alterations in gut microbiota, but highlighted the heterogeneity of findings, and the limited quantity and quality of studies (Cao et al., 2013). Studies investigating ASD, the gut microbiota and SCFAs, showed significantly higher levels of Desulfovibrio species and Bacteroides vulgatus and higher levels of SCFA's in the stools of autistic children compared to controls (Finegold et al., 2010; Wang et al., 2012). Clostridium Bolteae, another species that is reported to be over-represented in the gut microbiota in ASD, and its capsular polysaccharide consisting of rhamnose and mannose units, has been proposed as a viable potential vaccine to reduce C. bolteae colonization of the intestinal tract in autistic patients (Pequegnat et al., 2013).

Table 1.

Microbiota and ASD clinical studies.

Design Diagnosis, N, Age Measures Results References
Antibiotic—12-week trial of open label oral vancomycin ASD, regressive-onset autism (n = 11)
Age (43–84 months)
No control group
Childhood Autism Rating Scale
Developmental Profile II
Coded, paired videotapes scored by a clinical psychologist blinded to treatment status
Behavioral improvement Improvement at follow-up (2–8 months)—not sustained Sandler et al., 2000
FMT—18 weeks in total; 10 week open label and 8 week follow-up ASD (n = 18)
Age (7–16 years)
Controls (n = 20)
Age and Gender matched
Gastrointestinal Symptom Rating Scale
Parent Global Impressions-III (PGI-II)
Childhood Autism Rating Scale (CARS)
Aberrant Behavior Checklist (ABC)
Social Responsiveness Scale (SRS)
Vineland Adaptive Behavior Scale II (VABS-II)
ASD-related behavior improved (PGI-II) (CARS) (SRS) (ABC)80% reduction of GI symptoms (persisted for 8 weeks post-FMT) Bifidobacterium, Prevotella, and Desulfovibrio increased post-FMT (persisted for 8 weeks post-FMT) Kang et al., 2017
Cross-sectional ASD, regressive-onset autism (n = 13)
Controls (n = 8)
All ASD had GI symptoms (diarrhea and constipation)
Gastric and small-bowel specimens (7 ASD, 4 controls)
Limited dietary data: patients were on a gluten-free (GF), casein-free (CF) diet
ASD—more Clostridial species and non-spore-forming anaerobes and microaerophilic bacteria Finegold et al., 2002
Cross-sectional ASD (n = 20)
Age (6.7 ± 2.7 years)
20 neurotypical children
Age (8.3 ± 4.4 years)
Fecal samples
Autism Diagnostics Interview—Revised (ADI-Revised)
Autism Diagnostics Observation Schedule (ADOS)
Autism Treatment Evaluation Checklist (ATEC)
Pervasive Developmental Disorder Behavior
Inventory (PDD-BI)
Limited dietary data
Most ASD had GI symptoms
ASD—less diverse gut microbial compositions with lower levels of Prevotella, Coprococcus, and unclassified Veillonellaceae
Autistic symptoms, rather than the severity of GI symptoms, was associated with less diverse gut microbiota
Kang et al., 2013
Cross-sectional ASD patients (n = 58)
Age (3–16 years)
Two control groups (n = 22);
Non-autistic sibling group (n = 12)
Age (2–10 years)
Unrelated healthy group (n = 10)
Age (3–12 years of age)
91.4% of ASD had GI Symptoms Limited dietary data; Most of the children were on GF/CF diets and many were taking probiotics/prebiotics/antibiotics ASD—higher Clostridium histolyticum group compared to controls
Non-autistic sibling group had an intermediate level of the C. histolyticum group – not significantly different from ASD or controls
Parracho et al., 2005
Cross-sectional ASD (n = 23)
Age (123 ± 9 months)
Controls (n = 31)
Age (136 ± 9 months)
SCFAs
Dietary intake of macro-nutrients
ASD—fecal acetic, butyric, isobutyric, valeric, and isovaleric acid were all significantly higher compared with controls Wang et al., 2012
Cross-sectional ASD (n = 40)
Age (11.1 ± 6.8 years) Neurotypical controls (n = 40)
Age (9.2 ± 7.9 years)
Childhood Autism Rating Scale (CARS)
Autism Diagnostic Observation Schedule and Autism Behavior Checklist
Constipation defined according to Rome III criteria
All subjects of in this study were on a Mediterranean-based diet, and no antibiotics, probiotics, or prebiotics taken in the 3 months prior to the sample collection
ASD—increase in the Firmicutes/Bacteroidetes ratio due to a reduction of the Bacteroidetes relative abundance
ASD—at the genus level—decrease in Alistipes, Bilophila, Dialister, Parabacteroides, and Veillonella, while Collinsella, Corynebacterium, Dorea, and
Lactobacillus were significantly increased
Constipated ASD—high levels of bacterial taxa belonging to Escherichia/Shigella and Clostridium cluster XVIII
ASD—fungal genus Candida increased
Strati et al., 2017
Cross-sectional ASD (n = 23, without GI symptoms)
ASD (n = 28, with GI symptoms)
Age range (2–12 years)
Neurotypical siblings (n = 53)
Age range (2–12 years)
Childhood Autism Rating Scale (CARS)
Limited dietary data; Probiotics not excluded
No significant differences in microbiota Gondalia et al., 2012
Cross-sectional ASD (n = 15, with GI symptoms)
Age (4.5 ± 1.3 years)
Controls (n = 7, with GI symptoms)
Age (4.0 ± 1.1 years)
Autism Diagnostic Interview-Revised (ADI-R) Intestinal biopsies ASD with GI symptoms had a decrease in disaccharidases and hexose transporters, and decreases in Bacteroidetes, increase in Firmicutes/Bacteroidetes ratio, and increase in Betaproteobacteria compared with controls with GI symptoms Williams et al., 2011
Cross-sectional ASD (n = 15)
Control (n = 8)
Diet not recorded ASD—elevated levels of Clostridium boltea and Clostridium group I and XI Song et al., 2004
Cross-sectional ASD (n = 58, GI symptoms)
Age (6.91 ± 3.4 years)
Controls (n = 39)
Age (7.7 ± 4.4 years)
GI symptoms (assessed by the six-item GI Severity Index (6-GSI) questionnaire)
Autism Treatment Evaluation Checklist (ATEC)
Diet not recorded, ASD on probiotics
ASD—decreased fecal SCFAs, acetate, proprionate, and valerate
ASD—lower levels of Bifidobacterium and higher levels of Lactobacillus
GI symptoms were strongly correlated with the severity of autism
Adams et al., 2011
Meta-analysis of 15 cross-sectional studies 11 studies (n = 562) reported significant gut microbiota differences between ASD children and controls, particularly in the Firmicutes, Bacteroidetes and Proteobacteria phyla
Substantial heterogeneity in methodology and the often contradictory results of different studies—not possible to pool the results into a meta-analysis
Cao et al., 2013
Cross-sectional ASD children (n = 23)
Age (123 ± 9 months)
Controls (n = 31);
Typically developing siblings (n = 22)
Community controls (n = 9)
Age (136 ± 9 months)
Macronutrient intake determined from dietary records kept by caregivers, did not differ significantly between study groups ASD—elevated fecal acetic, butyric, isobutyric, valeric, isovaleric, and caproic acids, ammonia Wang et al., 2012
Cross-sectional ASD (n = 33, varying GI symptoms)
Controls (n = 15);
7 sibling controls
8 non-sibling controls
Age (all ASD and controls between 2 and 13 years)
No diet Bacteroidetes was found at high levels in the severely autistic group
Firmicutes were more predominant in the control group
Smaller, but significant, differences also in the Actinobacterium and Proteobacterium phyla
Desulfovibrio species and Bacteroides vulgatus present in significantly higher numbers in stools of severely autistic children than in controls
Finegold et al., 2010
Probiotic Intervention—“Children Dophilus” oral capsule containing 3 strains of Lactobacillus (60%), 2 strains of Bifidumbacteria (25%) and one strain of Streptococcus (15%), times a day for 4 months ASD (n = 10)
Age (2–9 years)
Siblings (n = 9)
Age (5–17 years)
Controls (n = 10)
Age (2–11 years)
Autism Diagnostic Interview (ADI)
Childhood Autism Rating Scale (CARS)
ASD—decrease of the Bacteroidetes/Firmicutes ratio and elevation of the amount of Lactobacillus
Desulfovibrio decreased postprobiotic
Desulfovibrio spp. associated with the severity of autism (ADI) restricted/repetitive behavior subscale score
Probiotic significantly decreased fecal TNFα levels in ASD
No correlation between plasma levels of oxytocin, testosterone, DHEA-S and fecal microbiota
Tomova et al., 2015
Cross-sectional Healthy children (n = 77)
Age (18–27 months)
Early Childhood Behavior Questionnaire (ECBQ)
(18 dimensions of temperament, three composite scales: Negative Affectivity, Surgency/Extraversion, Effortful Control)
Greater surgency/extraversion was associated greater phylogenetic diversity
Boys only—subscales loading on this composite scale were associated with differences in phylogenetic diversity, the Shannon Diversity index (SDI), beta diversity, and differences in abundances of Dialister, Rikenellaceae, Ruminococcaceae, and Parabacteroides
Higher effortful control was associated with a lower SDI score and differences in both beta diversity and Rikenellaceae were observed in relation to Fear
Associations between temperament and dietary patterns were observed
Christian et al., 2015
Cross-sectional ASD (n = 17)
Asperger's syndrome (n = 6)
Mean age (123 ± 9 months)
22 typically developing siblings
Age (144 ± 12 months)
Community controls (n = 9)
Age (114 ± 15 months)
Functional gastrointestinal disorder (FGID) questionnaire
Antibiotics/probiotics not excluded
Some on Gluten- and casein-free diet
ASD—Low Relative Abundances of the Mucolytic Bacterium and Akkermansia muciniphila and Bifidobacterium spp. in Feces Wang et al., 2011
Cross-sectional ASD (n = 23, 3 without siblings)
22 typically developing siblings
Age (144 ± 12 months)
Community controls (n = 9)
Age (114 ± 15 months)
No diet ASD—Sutterella spp. elevated in feces relative to controls and Ruminococcus torques higher in the children with ASD with a reported functional gastrointestinal disorder than those without such a disorder Wang et al., 2013
Cross-sectional ASD (n = 10)
Pervasive Developmental
Disorder Not Otherwise
Specified (PDD-NOS) (n = 10)
Healthy controls (HC) siblings (n = 10)
Age (all 4–10 years)
Autism Diagnostic Interview-Revised (ADI-R)
Autistic Diagnostic Observation Schedule (ADOS)
Childhood Autism Rating Scale (CARS)
Diet not recorded
No antibiotics, probiotics and prebiotics for at least 1 month before sampling
ASD—highest microbial diversity Faecalibacterium and Ruminococcus were present at the highest level in fecal samples of PDD-NOS and HC children. Caloramator, Sarcina and Clostridium genera were the highest in ASD children
Except for Eubacterium siraeum, the lowest level of Eubacteriaceae was found in fecal samples of ASD Bifidobacterium species decreased in AD—Compared to HC children Altered levels of free amino acids and volatile organic compounds of fecal samples in ASD and PDD-NOS
De Angelis et al., 2013
Cross-sectional ASD probands (n = 66)
Neurotypical (NT) siblings (n = 37)
Age (7–14 years)
Parent-completed ROME III questionnaire for pediatric Functional gastrointestinal disorders (FGIDs)
Child Behavior Check List (CBCL)
Targeted quantitative polymerase chain reaction (qPCR) assays were conducted on selected taxa implicated in ASD, including Sutterella spp., Bacteroidetes spp., and Prevotella spp.
No significant difference in macronutrient intake between ASD and NT siblings
There was no significant difference in ASD severity scores between ASD children with and without FGID
No significant difference in diversity or overall microbial composition was detected between ASD children with NT siblings
Son et al., 2015

Most studies conducted in ASD are non-interventional, and many do not adequately record detailed dietary information or medication use. Indeed, it is well established that ASD is highly associated with atypical eating patterns (Cermak et al., 2010). The interventional studies are few, and of small sample size. An open labeled trial (n = 11), with no control group, using the poorly absorbed oral antibiotic, vancomycin for 12 weeks, reportedly resulted in a short-term improvement in ASD related behavioral symptomatology in a group of children with regressive-onset autism (Sandler et al., 2000). Follow-up which occurred between 2 and 8 months, showed that the improvement was not sustained. More recently, a small (n = 18) open label study of Fecal Microbiota Transfer (FMT) in children with ASD reported an improvement in both GI symptoms and behavioral symptoms after 8 weeks (Kang et al., 2017). In this study, the abundance of Bifidobacterium, Prevotella, and Desulfovibrio increased following the 8 weeks of FMT treatment (see Table 1). Interestingly, in a cross sectional study, in healthy prepubertal children (n = 65) dietary fiber was associated with a better performance on a task measuring attentional inhibition (Khan et al., 2015). Moreover, a study investigating microbial composition at 1 year of age showed that a higher alpha diversity was associated with lower scores on the Mullen scale, the visual reception scale, and the expressive language scale at 2 years of age (Carlson et al., in press).

Schizophrenia spectrum disorder and the microbiota

As discussed below, data from pre-clinical studies indicate that certain domains related to schizophrenia, such as social cognition, are under the partial influence of the gut microbiota (Dinan et al., 2014). However, pre-clinical models have many limitations and translating promising pre-clinical findings into discernible clinical benefits for patients can be challenging, particularly for complex disorders such as schizophrenia. Moreover, it is important to highlight that there are considerable interpersonal differences in the gut microbiota profiles of healthy individuals (Backhed et al., 2012; Falony et al., 2016; Zhernakova et al., 2016). Consequently, there are multiple possible configurations for a healthy gut microbiota and it is also likely that some stable configurations are associated with disorders (Relman, 2012). It is important also to appreciate that the functional output of multiple microbiota configurations may in fact be equivalent, given that concepts of redundancy and pleiotropy can also be applied to specific microbial members of the overall consortium (Falony et al., 2016).

Despite the significant challenges, several pilot clinical studies investigating the microbiome in schizophrenia have emerged (see Table 2). A recent study investigating the gut microbiota in schizophrenia was conducted in First Episode Psychosis (FEP) patients (n = 28) compared to healthy controls (n = 16) (Schwarz et al., in press). There were five significant differences between the groups at the family level; Lactobacillaceae, Halothiobacillaceae, Brucellaceae, and Micrococcineae were increased, whereas Veillonellaceae were decreased in FEP patients. At the genus level, Lactobacillus, Tropheryma, Halothiobacillus, Saccharophagus, Ochrobactrum, Deferribacter, and Halorubrum were increased, and Anabaena, Nitrosospira, and Gallionella were decreased in FEP. Lactobacillus group bacterial numbers correlated positively with severity of psychotic symptoms measured using the Brief Psychiatric Rating Scale, and negatively with global assessment of functioning (GAF) scale. A subgroup analysis of those classified as less physically active, confirmed significant increases in Lactobacillaceae and significant decreases in Veillonellaceae in FEP. It is noteworthy that the vast majority of FEP patients were prescribed antipsychotic medication, which can impact gut microbiota composition (Davey et al., 2012, 2013; Bahra et al., 2015; Bahr et al., 2015). A small study (n = 32) of the oropharyngeal microbiome in schizophrenia also showed an increased abundance of Lactobacillus in schizophrenia patients, in addition to, Bifidobacterium and Ascomycota, compared to healthy controls (Castro-Nallar et al., 2015). Another study of the oral pharynx of 41 individuals with schizophrenia and 33 controls demonstrated that one bacteriophage genome Lactobacillus phage phiadh, was significantly more abundant in schizophrenia patients than in controls after adjustment for multiple comparisons and demographic covariates (Yolken et al., 2015).

Table 2.

Microbiota and clinical schizophrenia (SCZ) studies.

Design Diagnosis, N, Years Measures Results References
Cross-sectional Schizophrenia (n = 16)
Years (34.7 ± 4.8)
Controls (n = 16)
Years (34.3 ± 10.1)
Differences in smoking and BMI between groups
Shotgun metagenomic analysis of the oropharyngeal microbiome SCZ—higher proportions of Firmicutes, Ascomycota, Bifidobacterium and Lactobacilli (largest effect was observed in Lactobacillus gasseri)
SCZ—increase Candida and Eubacterium and reduction of Neisseria, Haemophilus and Capnocytophaga
SCZ—increased number of metabolic pathways related to metabolite transport systems including siderophores, glutamate and vitamin B12
Carbohydrate and lipid pathways and energy metabolism were abundant in controls
Castro-Nallar et al., 2015
Cross-sectional Schizophrenia (n = 41)
Years (39.2 ± 9.9)
Controls (n = 33)
Years (30.9 ± 8.8)
Differences in smoking, BMI and age
Metagenomic analysis to characterize bacteriophage genomes in oral pharynx SCZ—increased Lactobacillus phage phiadh (controlling for age, gender, race, socioeconomic status, or smoking) Yolken et al., 2015
Two case-control cohorts (n = 947) Schizophrenia (n = 261), including;
First-episode schizophrenia (n = 139, 78 antipsychotic naïve)
Years (37.71 ± 13.69)
Bipolar (n = 270)
Years (34.08 ± 13.15)
Controls (n = 277)
Years (32.02 ± 11.31)
Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) No differences in C. albicans exposures were found until diagnostic groups stratified by sex
SCZ—in males, C. albicans seropositivity conferred increased odds (OR 2.04–9.53) for a SCZ diagnosis
SCZ—in females, C. albicans seropositivity conferred increased odds (OR 1.12) for lower cognitive scores on RBANS with significant decreases on memory modules C. albicans IgG levels were not impacted by antipsychotic medications
Gastrointestinal (GI) disturbances were associated with elevated C. albicans in males with SCZ and females with bipolar
Severance et al., 2016a
14 week double-blind, placebo controlled .Lactobacillus rhamnosus strain GG and Bifidobacterium animalis subsp. lactis Bb12 (109 cfu) Schizophrenia (n = 56)
Probiotic (n = 30)
Placebo (n = 26)
Years (44.66 + 11.4)
Biweekly Positive and Negative Syndrome Scale (PANSS)
Self-reported—bowel score (scale of 1–4)
SCZ—in males—reduced C. albicans antibodies
S. cerevisiae were not altered Trends toward improvement in positive psychiatric symptoms in males
treated with probiotics who were seronegative for C. albicans
Severance et al., 2017
Lactobacillus rhamnosus strain GG and Bifidobacterium animalis subsp. lactis Bb12 (109 cfu) 14 week double-blind, placebo controlled Schizophrenia (n = 65)
33 probiotic
Years (44.4 ± 11.0)
32 placebo
Years (48.1 ± 9.4)
All on antipsychotic medication
Positive and Negative Syndrome Scale (PANSS) every 2 weeks
Self-reported—bowel score (scale of 1–4)
No significant differences in the PANSS
Probiotic group—significantly less likely to develop severe bowel difficulty
Dickerson et al., 2014
Longitudinal (12 months) First Episode Psychosis (FEP) (n = 28)
Years (25.9 ± 5.5)
Most on antipsychotics
Healthy controls (n = 16)
Years (27.8 ± 6.0)
Brief Psychiatric Rating Scale (BPRS) Global assessment of functioning (GAF) scale
Diet adapted from “Health Behavior and Health among the Finnish Adult Population” survey
FEP—at family level;
Lactobacillaceae, Halothiobacillaceae, Brucellaceae and Micrococcineae were increased whereas Veillonellaceae were decreased
FEP—at genus level; Lactobacillus, Tropheryma, Halothiobacillus, Saccharophagus, Ochrobactrum, Deferribacter and Halorubrum were increased, and Anabaena, Nitrosospira and Gallionella were decreased
Lactobacillus group bacterial numbers correlated positively with severity of psychotic symptoms measured using the BPRS and negatively with GAF scale
Schwarz et al., in press

Studies investigating the fungal composition of the human gut—the Mycobiome—are also emerging (Suhr and Hallen-Adams, 2015). A case-control cohort study that included 261 individuals with schizophrenia, 270 with bipolar disorder, and 277 non-psychiatric controls, found no differences in C. albicans exposure when analyzed at the group level. However, when stratified by sex, there was a reported increase in the odds for schizophrenia in males (Severance et al., 2016a). The same group conducted a randomized, double-blind, placebo-controlled, probiotic trial over a 14-week period, and showed that probiotic treatment significantly reduced C. albicans antibodies in males only, and a trend toward improvement in positive psychiatric symptoms in seronegative males (Severance et al., 2017). Both groups were prescribed antipsychotic medication, but antipsychotic regimes were not different between probiotic and placebo groups.

Communication pathways of brain-gut-microbiota axis

The human body contains as many bacterial cells as human cells (Sender and Fuchs, 2016), the majority of which reside in the gut, with bacterial concentrations ranging from 101 to 103 cells per gram in the upper intestines to 1011–1012 bacteria per gram in the colon (O'Hara and Shanahan, 2006; Derrien and van Hylckama Vlieg, 2015). With over 1,000 species and 7,000 strains the microbiota is an ecosystem dominated by bacteria, mainly strict anaerobes, but also includes viruses and bacteriophages, protozoa, archaea and fungi (Lankelma et al., 2015). In terms of bacterial phyla found in the gut, Firmicutes (species such as Lactobacillus, Clostridium, Enterococcus) and Bacteroidetes (species such as Bacteroides) account for the majority (Dethlefsen et al., 2007), though the other phyla such as Actinobacteria (Bifidobacteria), Proteobacteria (Escherichia coli), Fusobacteria, Verrucomicrobia, and Cyanobacteria are also present in relatively low abundance (Eckburg et al., 2005; Qin et al., 2010; Lankelma et al., 2015).

Although the functional significance of the gut microbiota has yet to be fully determined (Franzosa et al., 2014; Cani, 2017), it is clear that an intricate and interlinked symbiotic relationship exists between host and microbe (Ley et al., 2008), and there are a number of bidirectional signaling pathways by which the gut microbiota, acting via the brain-gut axis, can impact the brain. A key signaling pathway involves modulation of the immune system (Erny et al., 2015), though other pathways include the hypothalamic-pituitary-adrenal (HPA) axis (Sudo et al., 2004; Mudd et al., 2017), tryptophan metabolism (O'Mahony et al., 2015), the production of bacterial metabolites, such as SCFA (Tan et al., 2014) and via the vagus nerve (Bravo et al., 2011). Although much progress has been made, the precise signaling pathways mediating the influence of microbial products derived from gut microbiota on the brain remain largely unknown. Epigenetic factors may also play a role (Dalton et al., 2014; Stilling et al., 2014a,b; Thaiss et al., 2016). Recently, a novel signaling pathway has been advanced, that involves bacterial peptidoglycan (PGN) derived from the commensal gut microbiota (Arentsen et al., 2017). PGN was shown to translocate into the brain to activate specific pattern-recognition receptors (PRRs) of the innate immune system, and this could occur in both physiological and pathological conditions (Arentsen et al., 2017).

In addition, pre-clinical evidence from germ-free (GF) mice suggests that the microbiota can modulate the Blood Brain Barrier (BBB). Exposure of GF adult mice to the fecal microbiota from pathogen-free donors decreased BBB permeability (Braniste et al., 2014). Moreover, monocolonization of the intestine of GF adult mice with SCFA-producing bacterial strains normalized BBB permeability, whilst sodium butyrate was associated with increased expression of the tight junction protein occludin in the frontal cortex and hippocampus (Braniste et al., 2014). Together with a study that showed antibiotic-induced gut dysbiosis reduced the expression of tight junction proteins (claudin and occludin) mRNA in the hippocampus, and increased the expression of tight junction protein 1 and occludin mRNA in the amygdala (Frohlich et al., 2016), suggests that the BBB may be partially modulated by changes in the gut microbiota.

Microbiota and the immune system

A bidirectional communication system exists between the immune system and the CNS. Neuroimmune signaling during the prenatal or early post-natal developmental stages can have long lasting effects on the brain, and is an important determinant of cognitive function and emotional behavior (Dantzer et al., 2008; Bilbo et al., 2012; Filiano et al., 2017; Freytag et al., 2017). Peripheral cytokine signaling can modulate astrocytes, microglia and neurons in the CNS (Kohman and Rhodes, 2013). This occurs through leaky regions in the BBB such as circumventricular organs, active transport through transport molecules, activation of cells lining the cerebral vasculature (endothelial cells and perivascular macrophages), binding to cytokine receptors associated with the vagus nerve, stimulating the HPA axis at the anterior pituitary or hypothalamus and recruitment of activated cells such as monocytes/macrophages from the periphery to the brain (Haroon et al., 2012). In addition, functional lymphatic vessels lining the dural sinuses have been discovered, which serve as a route by which immune cells can communicate with the CNS (Louveau et al., 2015). Consequently, peripheral cytokines can modulate neurogenesis, synapse formation and plasticity (Hodes et al., 2015). It is established that cytokines can impact cognition and mood (Dowlati et al., 2010; Udina et al., 2012; Valkanova et al., 2013; Khandaker et al., 2014). Brain regions affected by administration of inflammatory stimuli include the basal ganglia and the dorsal anterior cingulate cortex (dACC), part of the limbic system, involved in cognitive and emotional processing (Harrison et al., 2009; Slavich et al., 2010; Capuron et al., 2012; Felger and Miller, 2012; Felger et al., 2013; Miller et al., 2013).

A critical function of the gut microbiota is to prime the development of the neuroimmune system (Round and Mazmanian, 2009; Olszak et al., 2012; Chistiakov et al., 2014; Francino, 2014). Alterations in the gut microbiota signature early in life can predispose to immune disorders (Penders et al., 2007; Fujimura et al., 2016) and the luminal surface of the gut is a key interface in this process (O'Hara and Shanahan, 2006). Indeed, the hygiene hypothesis first proposed in the late 1980's (Strachan, 1989; Patel and Gruchalla, 2017) and reconceptualized as the “old friends hypothesis” (Rook et al., 2003; Williamson et al., 2015) proposes that encountering less microbial biodiversity may contribute to the increase in chronic inflammatory disorders (Klerman and Weissman, 1989; Guarner et al., 2006; Rook and Lowry, 2008; Turnbaugh et al., 2009; Hidaka, 2012; Rook et al., 2013, 2014; Kostic et al., 2015; Stein et al., 2016). An intriguing strategy of “reintroducing” old friends has been suggested by a pre-clinical study using heat-killed Mycobacterium vaccae, an immunoregulatory environmental microorganism. Mice given this vaccine exhibited reduced subordinate, flight, and avoiding behavioral responses to a dominant aggressor in a murine model of chronic psychosocial stress when tested 1–2 weeks following the final immunization, compared to the control group (Reber et al., 2016). Depletion of regulatory T cells negated the protective effects of immunization with M. vaccae on anxiety-like or fear behaviors.

Mechanistic influences of microbiota on brain function and development

Toll-like receptors (TLRs)

Structural components of bacteria interact with the immune system via TLRs (McCusker and Kelley, 2013). Different TLRs recognize specific bacterial structures, for example; TLR2 recognizes structures from Gram positive bacteria whereas TLR4 mediates responses to structures such as lipopolysaccharide (LPS) primarily from Gram negative bacteria (Marteau and Shanahan, 2003). In the CNS, neurons and glial cells can express various TLRs (Bsibsi et al., 2002; Kielian, 2006; Trudler et al., 2010). Activation of TLRs trigger the induction of pro and anti-inflammatory cytokines (Takeda and Akira, 2005) and, as mentioned above, there are a number of routes by which peripheral cytokines can impact the brain (Haroon et al., 2012; Miller et al., 2013; Louveau et al., 2015). Dysregulation of this process, or excessive TLR activation, can result in chronic inflammatory and over-exuberant repair responses. Consequently, TLRs may serve as molecular communication channels between gut microbiota alterations and immune system homeostasis (Rogier et al., 2015). Indeed, TLR2 and TLR4 knockout mice showed subtle impairments in behavior and cognitive functions (Park et al., 2015; Too et al., 2016). A clinical study in subjects diagnosed with psychotic disorders showed specific alterations in TLR agonist-mediated cytokine release compared to healthy controls (McKernan et al., 2011), and more recently it has been shown that abnormal expression of TLRs can be modulated by antipsychotics (Kéri et al., 2017). Moreover, in post-mortem prefrontal cortex samples from subjects diagnosed with psychosis, alterations in TLR4 have been shown, which were dependent on antipsychotic treatment status at time of death (García-Bueno et al., 2016).

Microbiota and microglia

Microglia, central to the inflammatory process (Facci et al., 2014) are emerging as playing key roles in brain development, plasticity and cognition (Tay et al., 2017). These phagocytic innate immune cells account for approximately 10% of cells in the brain (Prinz et al., 2014), contribute to the plasticity of neural circuits by modulating synaptic architecture and function (Graeber and Streit, 2010) and can be modulated by glutamatergic and GABAergic neurotransmission (Fontainhas et al., 2011). Pre-clinical studies have shown that acute stress results in microglia activation and increased levels of proinflammatory cytokines in areas such as the hippocampus (Frank et al., 2007) and hypothalamus (Blandino et al., 2009; Sugama et al., 2011). Most studies show increases in activated microglia in response to chronic stress (Tynan et al., 2010; Hinwood et al., 2011, 2012; Bollinger et al., 2016).

Preliminary changes in the microenvironment of the microglia may result in a susceptibility to a secondary inflammatory stimulus (Perry and Holmes, 2014). This concept of microglia priming may be of relevance to neurodevelopmental disorders, such as ASD and schizophrenia, which often require multiple environmental “hits” (Feigenson et al., 2014; Fenn et al., 2014). In an environmental two-hit rodent model in which the first experimental manipulation targeted pregnant dams, and the second manipulation was given to the resulting offspring, exposure to prenatal immune challenge and peripubertal stress synergistically induced pathological effects on adult behavioral functions and neurochemistry (Giovanoli et al., 2013, 2015). Thus, early-life stress may prime microglia, leading to a potentiated response to subsequent stress (Calcia et al., 2016).

In human studies, microglial dysregulation has been demonstrated in several psychiatric disorders. In medication free depressed patients, microglial activation has been demonstrated in the prefrontal cortex, ACC, and insula, using translocator protein density measured by distribution volume in a PET study positron emission tomography (PET) study (Setiawan et al., 2015). Using a different tracer, (11)[C]PBR28, subjects at high risk of psychosis, and those with schizophrenia also showed evidence of altered microglial activation compared to healthy controls (van Berckel et al., 2008; Bloomfield et al., 2016). However, not all studies are consistent and no clear consensus exists (Holmes et al., 2016; Narendran and Frankle, 2016; Collste et al., 2017; Notter and Meyer, 2017).

The gut microbiota, emerging as an important neuroimmunomodulator (Foster, 2016; Rea et al., 2016), is also involved in the maturation and activation of microglia (Cryan and Dinan, 2015; Erny et al., 2015). Interestingly, GF mice display underdeveloped and immature microglia in the cortex, corpus callosum, hippocampus, olfactory bulb, and cerebellum (Erny et al., 2015). There was an upregulation of microglia transcription and survival factors, and downregulation of cell activation genes and genes for type 1 IFN receptor signaling compared with those isolated from conventionally colonized control mice. These defects were partially restored by recolonization with a complex microbiota, and SCFAs reversed the defective microglia in the absence of complex microbiota (Erny et al., 2015). Collectively, these studies suggest that subtle alterations in gut microbiota acquisition and development, by regulating neuro-inflammatory processes, may act as additional vulnerability factors that predispose to neurodevelopmental disorders such as ASD and schizophrenia.

Microbiota and neurochemistry

At the cellular level, brain development and function requires a complex and coordinated birth, migration and differentiation of both neurons and glia, followed by synaptic integration and neural circuit formation. Both ASD and schizophrenia are associated with dysregulation of synaptic function and structure (McGlashan and Hoffman, 2000; Faludi and Mirnics, 2011; Spooren et al., 2012; Habela et al., 2016). The gut microbiota plays a role in developmental programming of the brain, specifically, synapse maturation and synaptogenesis (Diaz Heijtz et al., 2011) Figure 2. Synaptophysin, a marker of synaptogenesis, and PSD 95, a marker of excitatory synapse maturation, were decreased in the striatum in GF animals compared to specific-pathogen-free (SPF) animals. This suggests that the gut microbiota may programme certain brain circuits when colonized by maternal microbiota. However, the authors point out that exposure to gut microbiota metabolites during embryogenesis may also be a possible mechanism. Interestingly, reduced levels of synaptophysin have been demonstrated in the cerebral cortex of post-mortem samples from schizophrenia subjects (Hu et al., 2015).

Figure 2.

Figure 2

The gut microbiome and the neurobiology of schizophrenia and autism. Autism and schizophrenia are associated with a number of alterations in the CNS including altered availability of neuroactive precursors. Studies in germ free animals indicate a substantial overlap between these neurobiological characteristics and the scope of influence of the gut microbiome in the CNS.

Brain-derived neurotrophic factor (BDNF)

A key regulator of synaptic plasticity and neurogenesis in the brain, throughout life, is the neurotrophin, BDNF (Monteggia et al., 2004). Given the role of BDNF in the regulation of synaptic strengthening and pruning, maintaining appropriate levels of BDNF and other neurotrophins, especially during critical neurodevelopmental windows is vital for both ASD and schizophrenia (Nieto et al., 2013). Meta-analysis showed reduced blood levels in both medication naïve and medicated adult individuals diagnosed with schizophrenia (Green et al., 2011). Conversely, children with ASD have increased levels of blood BDNF (Qin et al., 2016; Saghazadeh and Rezaei, 2017). In GF rodents, levels of BDNF were reduced in the cortex and hippocampus in GF mice (Sudo et al., 2004). In a study by Clarke et al. this finding was replicated, but in male mice only (Clarke et al., 2013). However, not all studies are consistent; Neufeld et al. (2011) confirmed a decreased level of anxiety like behavior in GF animals, but found an increase in BDNF mRNA in the hippocampus in female mice. Prebiotics can alter BDNF levels (Savignac et al., 2013) and increase BDNF gene expression in the hippocampus (Burokas et al., in press). Collectively, these pre-clinical studies suggest that certain neurotransmitters and neuromodulators of relevance to the pathophysiology of ASD and schizophrenia are under the influence of the gut microbiota Figure 2.

γ-aminobutyric acid (GABA) and glutamate

At the neurotransmitter level, several signaling pathways have been shown to be dysfunctional in ASD and schizophrenia. Glutamatergic and GABAergic dysfunction and the consequences on excitatory to inhibitory cortical activity is one hypothesis to account for the similarities in the social and cognitive disturbances in ASD and schizophrenia (Canitano and Pallagrosi, 2017). GABA is an important inhibitory neurotransmitter in the brain, and GABA dysfunction has been implicated in ASD and schizophrenia (Schmidt and Mirnics, 2015). Although not a central source, it is interesting to note that certain bacteria can produce neuroactive metabolites (Wikoff et al., 2009; Lyte, 2011, 2013), for example specific strains of Lactobacillus and Bifidobacteria can produce GABA by metabolizing dietary glutamate (Barrett et al., 2012). Indeed, L. rhamnosus (JB-1) was shown to reduce anxiety and depression related behavior in mice and increase GABA receptor levels in the hippocampus (Bravo et al., 2011). Interestingly, in vagotomized mice, these effects were not found, further supporting the concept that the vagus nerve is an important neural signaling pathway between the microbiota and brain. A pre-clinical magnetic resonance spectroscopy study adds further evidence to support the concept that oral L. rhamnosus can increase central GABA levels (Janik et al., 2016). In a recent study, prebiotics, fructo-oligosaccharide (FOS) and galacto-oligosaccharide (GOS), increased GABA-B1 and GABA-B2 receptor gene expression in the hippocampus (Burokas et al., in press).

The glutamate hypothesis of schizophrenia, has suggested that hypofunction of signaling through NMDA receptors (NMDARs) plays a causal role in schizophrenia (Gonzalez-Burgos and Lewis, 2012). The glutamatergic system appears to contribute to certain cognitive deficits in schizophrenia (Thomas et al., 2017). Similarly, glutamatergic dysfunction has been implicated in ASD (Rojas, 2014). In GF mice Neufeld and colleagues demonstrated a decrease in the NMDAR subunit NR2B mRNA expression in the amygdala (Neufeld et al., 2011) Figure 2. Although a review of post-mortem studies of subjects with schizophrenia found consistent evidence of morphological alterations of dendrites of glutamatergic neurons in the cerebral cortex, there were no consistent alterations of mRNA expression of glutamate receptors (Hu et al., 2015).

Serotonin

Serotonin (5-HT) has a wide range of physiological functions, and is involved in the modulation of anxiety, conditioned fear, stress responses, reward, and social behavior (Lucki, 1998; Dayan and Huys, 2008; Asan et al., 2013). A meta-analysis of post-mortem studies found an elevation in prefrontal 5-HT1A receptors and a reduction in prefrontal 5-HT2A receptors in schizophrenia (Selvaraj et al., 2014). Serotonin, and its pre-cursor tryptophan, are critical signaling molecules in the brain-gut-microbiota axis (O'Mahony et al., 2015). In GF mice decreased 5-HT1A in hippocampus has been shown (Neufeld et al., 2011). In the gastrointestinal tract (GI), 5-HT plays an important role in secretion, sensing and signaling (Mawe and Hoffman, 2013). The largest reserve of 5-HT is located in enterochromaffin cells (Berger et al., 2009). Emerging evidence also suggests that the serotonergic system may be under the influence of gut microbiota, especially, but not limited to, periods prior to the emergence of a stable adult-like gut microbiota (Desbonnet et al., 2008; El Aidy et al., 2012; Clarke et al., 2013). A metabolomics study demonstrated that the gut microbiota has a significant impact on blood metabolites and showed an almost three-fold increase in plasma serotonin levels when GF mice are colonized by gut microbiota (Wikoff et al., 2009). The gut microbiota itself is also an important regulator of 5-HT synthesis and secretion. For example, colonic tryptophan hydroxylase 1 (Tph1) mRNA and protein were increased in humanized GF and conventionally raised mice. Bacterial metabolites have also been demonstrated to influence Tph1 transcription in a human enterochromaffin cell model (Reigstad et al., 2015). Others have demonstrated that distinct microbial metabolites produced by spore forming bacteria increase colonic and blood 5-HT in chromaffin cell cultures (Yano et al., 2015).

Kynurenine

The regulation of circulating tryptophan availability, and the distribution and subsequent kynurenine pathway metabolism, in the periphery and CNS, is tightly regulated during all stages of life (Ruddick et al., 2006; Badawy, 2017). The enzyme indoleamine 2,3-dioxygenase (IDO) found in macrophages and microglia cells is the first and rate limiting step in the kynurenine pathway of tryptophan catabolism. The expression of tryptophan-2,3-dioxygenase (TDO) can be induced by circulating glucocorticoids (O'Connor et al., 2009) and has been reported to be regulated by the gut microbiota during colonization (El Aidy et al., 2014). Under normal physiological conditions, approximately 99% of tryptophan is metabolized to kynurenine in the liver by TDO. However, proinflammatory cytokines such as IFN-γ, CRP, IL-1, IL-6, and TNF-α can induce IDO resulting in the metabolism of tryptophan along the kynurenine pathway (Schwarcz et al., 2012). Kynurenine, tryptophan and 3-hydroxykynurenine (3-HK) can cross the BBB and tryptophan's conversion to kynurenine and 3-HK in the peripheral circulation can therefore contribute to CNS levels (Schwarcz et al., 2012; Myint and Kim, 2014). In the brain, kynurenine metabolism occurs in all cells, though the two kynurenine pathway branches are physically segregated into distinct cell types. Astrocytes contain kynurenine aminotransferases (KATs), not kynurenine 3-monooxygenase (KMO) and therefore cannot produce 3-hydroxykynurenine (3-HK) from Kynurenine (Guidetti et al., 2007). The end result of the metabolic pathway in astrocytes is the neuroprotective Kynurenic acid (KYNA) (Gramsbergen et al., 1997), whereas, in microglia, it is the neurotoxic metabolite quinolinic acid (Alberati-Giani et al., 1996).

As mentioned above, regulation of the kynurenine pathway is important throughout life, but especially during sensitive periods of early neurodevelopment. KYNA is an NMDA and alpha7 nicotinic (α7nACh) receptor antagonist, both important in modulating brain development (Myint and Kim, 2014). Administration of kynurenine, starting during embryogenesis, reduced the expression of α7nACh receptor and mGluR2 expression, and induced deficits in prefrontal cortex mediated cognition in adult rats (Pershing et al., 2015). Indeed, prenatal, but not adolescent, kynurenine treatment caused significant impairments in hippocampal-mediated behavioral tasks (Pocivavsek et al., 2014). Combining perinatal choline-supplementation, with embryonic kynurenine manipulation, to potentially increase activation of α7nACh receptors during development, can attenuate cognitive impairments in adult rat offspring (Notarangelo and Pocivavsek, 2017). Furthermore, prenatal kynurenine induces age-dependent changes in NMDA receptor expression (NR2A, NR1) (Pershing et al., 2016). This study also showed that juvenile rats that were given kynurenine performed better in a trace fear conditioning task, whereas the adults showed deficits. Prenatal inhibition of kynurenine pathway, using the kynurenine-3-monoxygenase inhibitor (Ro61-8048), results in altered synaptic transmission and protein expression in the brains of adult offspring (Forrest et al., 2013; Khalil et al., 2014; Pisar et al., 2014), and also changes hippocampal plasticity (Forrest et al., 2015). Using a kynurenine 3-monooxygenase knockout mouse model (Kmo−/−), which increased brain KYNA levels, showed impairments in contextual memory, social behavior, and increased anxiety-like behavior (Erhardt et al., in press). Interestingly, administering D-amphetamine to Kmo−/− mice showed potentiated horizontal activity in the open field paradigm.

In schizophrenia, increased KYNA levels in CSF, including in drug naïve patients (Nilsson et al., 2005), and in post-mortem brain samples have been shown (Erhardt et al., 2001; Plitman et al., 2017). In a clinical study, patients with schizophrenia (n = 64) were more intolerant to a psychological stress challenge than healthy controls, and while salivary KYNA levels increased significantly between baseline and 20 min following the stressor in both patients and controls, patients who were unable to tolerate the stressful tasks showed significantly higher levels of KYNA than patients who tolerated the psychological stressor or healthy controls (Chiappelli et al., 2014). A recent pre-clinical study showed that restraint stress in pregnant mice caused significant elevations of KYNA levels in the maternal plasma, placenta, and fetal brain (Notarangelo and Schwarcz, 2017). Furthermore, the kynurenine/tryptophan ratio was significantly higher in patients diagnosed with psychotic disorder (Barry et al., 2009). Collectively, these pre-clinical and clinical studies highlight the importance of the kynurenine pathway during neurodevelopment, and there is a growing appreciation that integrating these important insights with the emerging importance of microbial regulation of this pathway will be an important research objective (Kennedy et al., 2017).

Zinc signaling

The essential micronutrient Zinc plays an important role in immune function and GI development and function (Kau et al., 2011; Vela et al., 2015). Multiple independent factors affect Zinc status, including diet, prenatal and early life stress, immune system dysregulation, and impaired GI function (Vela et al., 2015). Zinc deficiency, particularly during the prenatal phase, has been proposed as an environmental risk factor for ASD. Indeed, in rats, acute Zinc deficiency can result in hyperactivity and over-responsivity, whereas prenatal deficiency can impair vocalizations and social behavior (Grabrucker et al., 2014). It has been suggested that the post-synaptic protein Shank3, which is localized at synapses in the brain and is associated with neuro-developmental disorders such as ASD and schizophrenia, is an important component of zinc-sensitive signaling system that regulates excitatory synaptic transmission, and may lead to cognitive and behavioral abnormalities in infants with ASD (Grabrucker et al., 2014; Arons and Lee, 2016). In clinical studies, Zinc deficiency has been reported in infants with ASD (Yorbik et al., 2004; Yasuda et al., 2011; Li et al., 2014). However, studies investigating Zinc levels in schizophrenia have yielded inconsistent results (Cai et al., 2015). The impact of micronutrient imbalances on the gut microbiota are beginning to emerge (Hibberd et al., 2017). In a study using chicks, Zinc deficiency induced gut microbiota alterations and decreased species richness and diversity (Reed et al., 2015). Excess dietary Zinc significantly altered the gut microbiota and in turn reduced the threshold of antibiotics needed to confer susceptibility to C. difficile infection in mice (Zackular et al., 2016).

Epigenetic influences

Dietary factors can result in epigenetic alterations that lead to disease susceptibility (Jirtle and Skinner, 2007). It has been established that prenatal malnutrition increases the risk of schizophrenia (Susser and Lin, 1992; St Clair et al., 2005; Xu et al., 2009). Furthermore, it has been suggested that the microbiota is an important mediator of gene–environment interactions (Stilling et al., 2014b). SCFAs (butyrate, acetate and propionate) are neurohormonal signaling molecules produced by certain classes of bacteria such as Bacteroides, Bifidobacterium, Propionibacterium, Eubacterium, Lactobacillus, Clostridium, Roseburia, and Prevotella (Macfarlane and Macfarlane, 2012). SCFAs are transported by monocarboxylate transporters, which notably are expressed at the BBB (Steele, 1986; Vijay and Morris, 2014). A pre-clinical imaging study demonstrated that microbiota-derived acetate can cross the BBB where it can subsequently alter hypothalamic gene expression (Frost et al., 2014). Butyrate has been shown to be associated with increased expression of the tight junction protein occludin in the frontal cortex and hippocampus (Braniste et al., 2014). Butyrate, which acts as a potent inhibitor of Histone deacetylase (HDAC), is also a ligand for a subset of G protein-coupled receptors (Bourassa et al., 2016). It is clear that supra-physiological levels do have marked behavioral consequences (MacFabe et al., 2007; Macfabe, 2012; Thomas et al., 2012). However, the ability of physiological levels of SCFAs to substantially effect behavior via central mechanism are likely to be subtle, though cumulative chronic delivery may produce long-lasting stable effects on gene expression.

Microbiota and social behavior and cognition

Neuronal activity in the amygdala is altered in GF mice (Stilling et al., 2015). In these mice, expression of immediate early response genes such as Fos, Fosb, Egr2, or Nr4a1 were increased in the amygdala, in conjunction with increased signaling of the transcription factor CREB (Stilling et al., 2015). Differential expression and recoding of several genes involved in fundamental brain processes ranging from neuronal plasticity, metabolism, neurotransmission and morphology were identified and a significant downregulation was noted for immune system-related genes (Stilling et al., 2015). In addition to an altered transcriptional profile in the amygdala, GF mice have recently been shown to exhibit reduced freezing behavior during a cued memory retention test, while colonized GF mice were behaviorally comparable to conventionally raised mice during the retention test (Hoban et al., 2017). Furthermore, adult GF mice have distinct dendritic morphological changes in the amygdala and hippocampus (Luczynski et al., 2016) and myelination of the prefrontal cortex has also been shown to be under the influence of the gut microbiota (Hoban et al., 2016b). Using GF mice, Desbonnet et al. (2014) showed that the microbiota is crucial for the development of normal social behaviors, including social motivation and preference for social novelty, while also being an important regulator of repetitive behaviors (Arentsen et al., 2015; Buffington et al., 2016). This decreased sociability has also been demonstrated in rats (Crumeyrolle-Arias et al., 2014). Interestingly, the peptidoglycan (PGN)-sensing molecule, Pglyrp2, has been shown to modulate the development of social behavior in mice and alterations in the expression of the ASD risk gene c-Met (Arentsen et al., 2017).

Oxytocin, a neuropeptide produced in the paraventricular nucleus (PVN) of the hypothalamus, is important for sociability (Teng et al., 2013). Offspring of mothers fed a high-fat diet showed reduced levels of oxytocin PVN neurons, in addition to behavioral and gut microbiota alterations (Buffington et al., 2016). L. reuteri treatment restored oxytocin levels and social behaviors. A recent study, using low dose penicillin, administered to dams in late pregnancy and early post-natal life showed that this antibiotic induced gut microbiota alterations, increased cytokine expression in frontal cortex, modified BBB integrity and decreased anxiety-like and social behaviors, in offspring (Leclercq et al., 2017). Interestingly, concurrent supplementation with L. rhamnosus (JB-1) attenuated the penicillin induced decrease in social novelty.

The maternal immune activation (MIA) model serves as a useful model for neurodevelopmental disorders such as ASD and schizophrenia, and it is well established that prenatal infection can act as “neurodevelopmental disease primer,” the consequences of which are dependent on precise timing of MIA (Meyer et al., 2006; Smith et al., 2007; Knuesel et al., 2014; Meyer, 2014; Coiro et al., 2015; Meehan et al., 2016; Pendyala et al., 2017). MIA rodents display all three of the core features of human ASD, including limited social interactions, a tendency toward repetitive behavior and reduced communication (Patterson, 2011). A recent study showed that MIA induces dysregulation of fetal brain transcriptome by downregulating genes related to ASD (Lombardo et al., 2017). MIA has been associated with altered gut microbiota. Furthermore, the commensal Bacteroides fragilis reversed the deficits in communicative, stereotypic, anxiety-like and sensorimotor behaviors (Hsiao et al., 2013).

Autistic like behavior and neurochemical alterations have also been demonstrated in a mouse model of food allergy (de Theije et al., 2014b). The same author showed an altered gut microbiota profile in an autism model, using valproic acid (VPA) (de Theije et al., 2014a). Interestingly, VPA, a medication used as a mood stabilizer in bipolar affective disorder and as an antiepileptic, functions as a HDAC inhibitor and has a similar structure to the SCFA propionic acid. It is well established that VPA acid use during pregnancy increases the risk of autism (Jacob et al., 2013), and propionic acid can also modulate mitochondrial function in autism and control cell lines (Frye et al., 2016).

As indicated above, multiple cognitive domains are impacted in ASD and schizophrenia and the gut microbiota has been implicated in a number of relevant cognitive functions. The combination of acute stress and infection can impact cognition. Citrobacter rodentium infected C57BL/6 mice that were exposed to acute stress exhibited memory dysfunction (Gareau et al., 2011). Moreover, GF Swiss-Webster mice displayed memory impairment at baseline, in the absence of acute stress (Gareau et al., 2011). In male C57BL/6 mice, higher percentages of Clostridiales and lower levels of Bacteroidales in high-energy diets were related to poorer cognitive flexibility (Magnusson et al., 2015). In BALB/c mice, treatment with B. Longum resulted in an improvement in stress related behavior and cognition (Savignac et al., 2015). Hippocampal neurogenesis, a pivotal process in learning and memory consolidation (Deng et al., 2010; Levone et al., 2015; Anacker and Hen, 2017; Hueston et al., 2017) has been shown to be regulated by the gut microbiota. GF mice exhibit increased adult hippocampal neurogenesis in the dorsal hippocampus, and post-weaning microbial colonization failed to reverse the changes in adult hippocampal neurogenesis (Ogbonnaya et al., 2015). Furthermore, exercise or probiotics were able to ameliorate deficits in neurogenesis and behavior in antibiotic-treated mice (Mohle et al., 2016). A recent study showed that L. johnsonii CJLJ103 attenuated colitis and memory impairment in mice by inhibiting gut microbiota lipopolysaccharide production and NF-κB activation (Lim et al., 2017).

Using an antibiotic (ampicillin, metronidazole, vancomycin, ciprofloxacin, imipenem) treated rat model, gut microbiota depletion during adulthood resulted in deficits in spatial memory as measured by Morris water maze (Hoban et al., 2016a). In another pre-clinical study, that used ampicillin, bacitracin, meropenem, neomycin, and vancomycin, novel object recognition, but not spatial memory, was impaired in antibiotic-treated mice and this cognitive deficit was associated with brain region-specific changes in the expression of cognition-relevant signaling molecules, notably BDNF, N-methyl-D-aspartate receptor subunit 2B, serotonin transporter and neuropeptide Y system. The authors concluded that circulating metabolites and the cerebral neuropeptide Y system play an important role in the cognitive impairment and dysregulation of cerebral signaling molecules due to antibiotic-induced gut alterations (Frohlich et al., 2016). Furthermore, in a pre-clinical rodent model of diabetes, L. acidophilus, B. lactis, and L. fermentum, improved diabetes-induced impairment of cognitive function in the Morris water maze and synaptic activity in rats (Davari et al., 2013). The N-methyl-D-aspartate (NMDA) receptor antagonist, phencyclidine causes hyperlocomotion, social withdrawal, and cognitive impairment in rodents, and serves as a useful pharmacological rodent model of schizophrenia (Jones et al., 2011). A study investigating the effect of subchronic phencyclidine (subPCP) treatment on cognition and gut microbiota, found that the microbiota altered immediately after subPCP washout. Administration of ampicillin abolished the subPCP-induced memory deficit (Pyndt Jorgensen et al., 2015).

Microbiota and stress

The brain interprets perceived stressors to determine physiological and behavioral responses. This process can promote adaptation (allostasis), but when responses are exaggerated or overused (allostatic overload), pathology can ensue (McEwen, 2017). The immune system and HPA axis are pivotal to the stress response and act as mediators to alter neural circuitry and function, particularly in the hippocampus, amygdala, and prefrontal cortex (McEwen et al., 2016). Stressful life events can precipitate psychotic symptoms (Day et al., 1987), and increased sensitivity to minor stressful events are associated with more intense psychotic experiences in first episode psychosis (FEP) (Reininghaus et al., 2016b). In addition early life event stressors, such as childhood trauma (Varese et al., 2012) and social adversity/defeat stressors, such as migration/ethnic minority status can increase the risk of psychosis (Elizabeth Cantor-Graae and Jean-Paul Selten, 2005; Selten and Cantor-Graae, 2005; Fusar-Poli et al., 2017). As mentioned above, schizophrenia is a highly heterogenous disorder, and commonly co-morbid with anxiety and depressive disorders (Buckley et al., 2009; Achim et al., 2011). Similarly, approximately 40% of young people with ASD have at least one comorbid DSM-IV anxiety disorder (van Steensel et al., 2011) and there are higher levels of depression (Ghaziuddin et al., 2002; Magnuson and Constantino, 2011; Strang et al., 2012).

Stress can reshape gut microbiota composition (Wang and Wu, 2005; O'Mahony et al., 2009; Galley et al., 2014a,b; Golubeva et al., 2015; Frohlich et al., 2016). For example, early life maternal separation resulted in a significant decrease in fecal Lactobacillus numbers on day 3 post-separation which was correlated with stress related behaviors in rhesus monkeys (Bailey and Coe, 1999). In a mouse model of social disruption, stress altered the gut microbial profile and increased the levels of the pro-inflammatory cytokine IL-6 (Bailey et al., 2011). Interestingly, it was possible to transfer an anxious behavioral phenotype between two strains of mice via fecal microbiota transfer (Bercik et al., 2011). More recently, it has been shown that mice that received an obesity associated microbiota exhibit more anxiety-like behaviors associated with increased evidence of neuroinflammation compared to controls (Bruce-Keller et al., 2015).

As previously mentioned, the developmental trajectory of the gut microbiota is compatible with concepts of the early-life period as a vulnerable phase for the subsequent emergence of neurodevelopmental disorders (O'Mahony et al., 2009, 2017; Borre et al., 2014). Pre-clinical evidence suggests that the gut microbiota signature acquired and maintained during these pivotal stages may also affect stress reactivity. GF rodents demonstrate abnormal behavioral and neuroendocrine responses to stress (Sudo et al., 2004; Nishino et al., 2013; Crumeyrolle-Arias et al., 2014; Moloney et al., 2014) and the normal development of the HPA axis is contingent on microbiota colonization at specific neurodevelopmental time points (Sudo et al., 2004).

Furthermore, the expression of anxiety like behavior in a mouse model of early life stress is partially dependent on the gut microbiota (De Palma et al., 2015). Evidence suggests that prenatal stress also impacts the gut microbiota with implications for physiological outcomes in the offspring (Golubeva et al., 2015). In a mouse model of prenatal stress, maternal stress decreased the abundance of vaginal Lactobacillus, resulting in decreased transmission of this bacterium to offspring, which corresponded with changes in metabolite profiles involved in energy balance, and with disruptions of amino acid profiles in the developing brain (Jasarevic et al., 2015). Human infants of mothers with high self-reported stress and high salivary cortisol concentrations during pregnancy had significantly higher relative abundances of Proteobacterial groups known to contain pathogens and lower relative abundances of lactic acid bacteria (Lactobacillus) and Bifidobacteria (Zijlmans et al., 2015). In addition, those infants with altered microbiota composition exhibited a higher level of maternally reported infant GI symptoms and allergic reactions, highlighting the functional consequences of aberrant colonization patterns.

The stress related disorder, depression, commonly co-morbid with ASD and schizophrenia, has been associated with alterations in gut microbiota profiles (Naseribafrouei et al., 2014; Jiang et al., 2015) and altered metabolomic output (Yu et al., 2017). Fecal Microbiota Transfer (FMT) from depressed patients to GF mice (Zheng et al., 2016) and antibiotic treated rats (Kelly, 2016; Kelly et al., 2016a) resulted in the transfer of certain depressive and anxiety like behaviors in the recipient rodents. The first study investigating the gut microbiota in bipolar affective disorder patients (n = 115), showed levels of Faecalibacterium were decreased, after adjusting for age, sex, and BMI, compared to healthy control subjects (n = 64). Moreover, Faecalibacterium was associated with better self-reported health outcomes based on the Short Form Health Survey, the Patient Health Questionnaire, the Pittsburg Sleep Quality Index, the Generalized Anxiety Disorder scale, and the Altman Mania Rating Scale (Evans et al., 2017). Interestingly, reduced levels of Faecalibacterium were reported in the study by Jiang and colleagues, which negatively correlated with severity of depressive symptoms (Jiang et al., 2015).

Fecal microbiota signatures in patients with diarrhea-predominant Irritable Bowel Syndrome (IBS), a stress related GI disorder, were shown to be similar to those patients with depression (Liu et al., 2016). Moreover, FMT from IBS patients to rats, induced anxiety related behaviors in the rats (De Palma and Lynch, 2017). In a double blind RCT of IBS patients, 6 weeks of B. longum NCC3001 reduced depression scores as measured by the Hospital Anxiety and Depression scale, and responses to negative emotional stimuli in amygdala and fronto-limbic regions, using fMRI, compared to placebo (Pinto-Sanchez et al., 2017). A recent study, using structural MRI, showed that gut microbial composition correlated with sensory and salience-related brain regions (Labus et al., 2017).

Translational approaches

In pre-clinical studies, both prebiotic (Burokas et al., in press) and probiotic treatment can reduce stress related behaviors (Abildgaard et al., 2017; Moya-Pérez et al., 2017). In a recent study, L. reuteri was reported to reduce despair like behavior in mice by inhibition of intestinal Indoleamine 2,3-dioxygenase (IDO1) and decrease peripheral levels of kynurenine (Marin et al., 2017). The profusion of pre-clinical data indicating a role for the brain-gut-microbiota axis in brain development, function and behavior, prompted the growing need to translate these findings into human populations (Kelly et al., 2015). “Psychobiotics,” originally defined as live bacteria that when ingested in adequate amounts could produce a positive mental health benefit, in terms of anxiety, mood and cognition (Dinan et al., 2013), has more recently been expanded to encompass “any substance that exerts a microbiome-mediated psychological effect” (Sarkar et al., 2016; Allen et al., 2017).

The process of translating psychobiotics from bench to bedside is not without significant challenges (Arrieta et al., 2016; Kelly, 2016; Kelly et al., 2016b; Cani, 2017), but a growing number of small studies with healthy individuals suggest that prolonged pre- and probiotic consumption can positively affect aspects of mood and anxiety in healthy controls (Messaoudi et al., 2011; Mohammadi et al., 2015; Steenbergen et al., 2015; Allen et al., 2016) and modulate HPA axis function (Messaoudi et al., 2011; Schmidt et al., 2015; Allen et al., 2016). Importantly, a fermented milk containing B. animalis, Streptococcus thermophiles, L. bulgaricus, and L. lactis, administered for 4 weeks to healthy women, reduced the task-related response of a distributed functional network containing affective, viscerosensory and somatosensory cortices, independent of self-reposted GI symptoms (Tillisch et al., 2013).

In humans, studies investigating the potential cognitive enhancing effects of microbial based therapies are starting to emerge (Allen et al., 2016). In this study, 4 weeks of treatment with the probiotic B. longum 1714 modestly improved performance in a hippocampal dependent memory task in healthy volunteers. However, this effect is likely strain specific since this subtle cognitive enhancing effect was not evident following administration of L. rhamnosus (JB-1) (Kelly et al., 2017a). In a randomized, double-blind, placebo-controlled trial involving healthy human participants (n = 76), the tetracycline antibiotic doxycycline (200 mg), a matrix metalloproteinase inhibitor, resulted in reduced fear memory retention, measured with fear-potentiated startle, 7 days post-acquisition compared to participants that received placebo (Bach et al., 2017). Doxycycline can alter the composition of the gut microbiota and its metabolomic output (Angelakis et al., 2014; Behr et al., 2017). Considering the recent pre-clinical data suggesting a role for the gut microbiota in the behavioral response during amygdala-dependent memory retention (Hoban et al., 2017), it would be a compelling prospect to ascertain if alterations in gut microbiota played a physiological role in this antibiotic human study. A cross sectional MRI study comparing 20 obese individuals to 19 age and sex matched non-obese controls, reported that the relative abundance of Actinobacteria phylum was associated with magnetic diffusion tensor imaging variables in the thalamus, hypothalamus, and amygdala and also with to cognitive test scores related to speed, attention, and cognitive flexibility (Fernandez-Real et al., 2015). Although preliminary, these studies, and others (Pinto-Sanchez et al., 2017), are beginning to merge microbiome research with neuroimaging to further delineate the role of the gut microbiota on cognition and neural circuitry.

To date, there are two interventional studies investigating potential psychobiotics in clinical depression, with conflicting results. In the first study, 8 weeks of a multispecies probiotic containing L. acidophilus, L. casei, and B. bifidum, added to an SSRI, reportedly reduced depressive symptoms in moderately depressed patients compared to placebo (Akkasheh et al., 2016). The other study, conducted in antidepressant free depressed subjects, failed to show superiority of L. helveticus and B. longum over placebo, in an 8-week double blind randomized controlled trial (Romijn et al., 2017). A Mediterranean diet, suggested as protective for depression, has been associated with beneficial microbiome-related metabolomic profiles (De Filippis et al., 2015) and there is increasing awareness of the role of a healthy diet in reducing the risk of depression (Jacka et al., 2010, 2017; Opie et al., 2017). Collectively, these studies suggest that the gut microbiota may play a pathophysiological role in stress-related disorders. However, given the small sample sizes and lack of a standardized approach in these studies, a robust and consistent gut microbiota signature in stress-related disorders, remains elusive. Moreover, a systematic review found very limited evidence for the efficacy of psychobiotics in psychological outcomes (Romijn and Rucklidge, 2015). Similarly, even in GI disorders, gut microbiota analysis as a diagnostic or prognostic tool has not transitioned into routine clinical practice (Quigley, 2017).

There has been one clinical interventional study investigating probiotics in patients diagnosed with schizophrenia. This randomized, double-blind, placebo-controlled trial (n = 65), used Lactobacillus rhamnosus strain GG and Bifidobacterium animalis subsp. lactis strain Bb12, improved GI symptoms, but failed to impact positive or negative symptoms (Dickerson et al., 2014). A number of small studies have shown that the antibiotic minocycline, notwithstanding a complex mechanism of action, is known to modulate the brain-gut-microbiota axis, (Wong et al., 2016), and may improve negative and cognitive symptoms in schizophrenia (Miyaoka et al., 2008; Levkovitz et al., 2010; Jhamnani et al., 2013; Khodaie-Ardakani et al., 2014). This raises the question of whether microbiome based therapies could play a role in the amelioration of cognitive or negative symptoms in subgroups of psychosis spectrum disorder.

Schizophrenia spectrum disorder and stratified psychiatry

The full neuropsychiatric implications of specific aberrations in the gut microbiota at early developmental stages or during adolescence have not been fully explored. It is an intriguing prospect that these aberrations may serve as additional risk factors or mediators for the development of psychotic disorders. It remains an unanswered question whether the gut microbiota is a state or trait marker and whether it plays a role, in conjunction with for example, stress, as a trigger factor for a psychotic relapse. The role of psychobiotics in schizophrenia remains under investigated (see Table 2). It would be interesting to explore whether a microbial based therapy could be a useful preventative strategy, or as an adjunctive agent in subgroups or whether it could reduce conversion to psychosis in subgroups at risk of developing the disorder. Well powered, longitudinal studies, encompassing neuroimaging markers would be required to definitively answer these questions.

In recent years, the categorical diagnostic system in clinical psychiatry has been challenged. Even the term schizophrenia has been disputed (van Os, 2016), with evidence showing that renaming the disorder can reduce stigma and benefit communication between clinicians, patients and families (George and Klijn, 2013; Lasalvia et al., 2015). There is growing momentum toward a more precise, dimensional approach, designed to uncover the biological mechanisms of these complex disorders. Functional dimensional constructs grouped into domains such as negative valance (acute threat (fear), potential threat (anxiety), sustained threat, loss, frustrative non-reward), positive valence (approach motivation, initial responsiveness to reward attainment, sustained/longer term responsiveness to reward attainment, reward learning, habit), cognitive (attention, perception, declarative memory, language, cognitive control, working memory), social processing (affiliation and attachment, social communication, perception, and understanding of self/others), and arousal/regulatory systems (arousal, circadian rhythms, sleep-wakefulness) examined across units of analysis from genes, molecules, cells, circuits, physiology, neuroimaging, behavior and self-report have been proposed (Insel et al., 2010). This dimensional approach is more difficult in disorders such as psychosis, compared to mood disorders, but this exciting process has begun (Reininghaus et al., 2016a; Cohen et al., 2017; Joyce et al., 2017). By deconstructing heterogenous systems-disorders (Öngür, 2017; Silbersweig and Loscalzo, 2017), such as schizophrenia into transdiagnostic constructs, and stratifying subgroups of patients based on similar pathophysiology, such as microbiome alterations and related signaling pathways, this opens up the possibility to advance personalized and precision treatments options (Kaiser and Feng, 2015).

Additionally, by removing the constraints of classical psychiatric disease diagnosis it has the potential to better align pre-clinical and clinical studies to build a common framework of comparable neurobiological abnormalities. Clearly, it is impossible to fully mimic a complex neuropsychiatric disorder such as schizophrenia or ASD in non-human animals. Hallucinations, delusions, thought disorder, and language impairments cannot be modeled. Thus, rather than modeling an entire disorder, the focus should be aimed at more precise constructs such those mentioned above, with the addition of the gut microbiota. Most currently used behavioral models do not include the gut microbiota as a factor. Clearly demonstrating causality in microbiome research is challenging (Hanage, 2014; Cani, 2017). The humanized FMT model is an integral component to demonstrate cause and effect in gut microbiota studies involving neurodevelopmental disorders such as schizophrenia, and provided a reliable and reproducible model can be developed, the precise temporal dynamics of the emergence and possible persistence of the behavioral alterations post FMT could be further delineated.

Furthermore, whether different human donor symptom profiles can be transferred via FMT may further disentangle the contribution of the gut microbiota to the pathophysiology of aspects of psychosis, by attempting to transfer sub-categories of psychotic subjects, including medication free subjects, with different constructs such as negative valance, positive valence, cognitive, social processing and arousal/regulatory systems. While it must be acknowledged that significant neuroscientific advances have frequently been lost in translation and not had appreciable benefits for psychiatric patients as yet, an evolving dimensional framework, consolidating multiple disciplines, and encompassing the gut microbiota as an additional environmental construct linked to other constructs, offers potential to identify sub-groups of patients that may be more likely to respond to a microbiome-based therapeutic approach at specific neurodevelopmental time points (Kelly, 2016; Severance et al., 2016c; Kelly et al., 2017b).

Conclusions and perspectives

Highly complex neurodevelopmental disorders such as ASD and schizophrenia require a systems level approach. The human brain develops and functions within the context of a complex network of lifelong microbial signaling pathways from gut to brain. Pre-clinical studies are beginning to provide mechanistic insights into these signaling pathways as they relate to the social, emotional and cognitive domains of the brain. Furthermore, they suggest that psychobiotics can ameliorate certain defects. However, translating these promising pre-clinical benefits to human neurodevelopment disorders is challenging. The majority of clinical studies investigating the gut microbiota in ASD are cross sectional and underpowered, and there is insufficient evidence of solid clinical relevance. In schizophrenia, there is emerging preliminary evidence of an altered gut microbiota. An intriguing prospect would be to focus on different neurodevelopmental time points, for example during adolescence, in subgroups at risk of developing neuropsychiatric symptoms, and to encompass a dimensional construct approach. Larger prospective interventional clinical studies, with central markers of brain function, utilizing therapeutic modulation of the gut microbiota or its metabolites are required. Furthermore, exploration of the interaction of the gut microbiota and nutritional modification, at different neurodevelopmental stages, including pre-conception, warrants exploration as a preventative strategy for neurodevelopmental disorders in addition to stress-related disorders (Jacka, 2017). Although it is premature to draw firm conclusions about the clinical utility of microbiome based treatment strategies in neurodevelopmental disorders at this point, it is an exciting frontier in psychiatry research.

Author contributions

JK wrote the manuscript. CM created the figures. GC, JC, and TD critiqued and edited the manuscript drafts.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The APC Microbiome Institute is funded by Science Foundation Ireland (SFI). This publication has emanated from research conducted with the financial support of Science Foundation Ireland (SFI) under Grant Number SFI/12/RC/2273. The authors and their work were also supported by the Health Research Board (HRB) through Health Research Awards (grants no HRA_POR/2011/23; TD, JC, and GC, HRA_POR/2012/32; JC, TD, and HRA-POR-2-14-647: GC, TD) and through EUGRANT613979(MYNEWGUTFP7-KBBE-2013-7). The Centre has conducted studies in collaboration with several companies including GSK, Pfizer, Wyeth and Mead Johnson. GC is supported by a NARSAD Young Investigator Grant from the Brain and Behavior Research Foundation (Grant Number 20771). We would like to acknowledge work conducted as part of a PhD thesis; JK (2016). The gut microbiota in depression (PhD Thesis), University College Cork.

References

  1. Abildgaard A., Elfving B., Hokland M., Wegener G., Lund S. (2017). Probiotic treatment reduces depressive-like behaviour in rats independently of diet. Psychoneuroendocrinology 79, 40–48. 10.1016/j.psyneuen.2017.02.014 [DOI] [PubMed] [Google Scholar]
  2. Achim A. M., Maziade M., Raymond É., Olivier D., Mérette C., Roy M.-A. (2011). How prevalent are anxiety disorders in schizophrenia? A meta-analysis and critical review on a significant association. Schizophr. Bull. 37, 811–821. 10.1093/schbul/sbp148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Adams J. B., Johansen L. J., Powell L. D., Quig D., Rubin R. A. (2011). Gastrointestinal flora and gastrointestinal status in children with autism–comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 11:22. 10.1186/1471-230X-11-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Adolphs R. (2001). The neurobiology of social cognition. Curr. Opin. Neurobiol. 11, 231–239. 10.1016/S0959-4388(00)00202-6 [DOI] [PubMed] [Google Scholar]
  5. Akkasheh G., Kashani-Poor Z., Tajabadi-Ebrahimi M., Jafari P., Akbari H., Taghizadeh M., et al. (2016). Clinical and metabolic response to probiotic administration in patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. Nutrition 32, 315–320. 10.1016/j.nut.2015.09.003 [DOI] [PubMed] [Google Scholar]
  6. Alberati-Giani D., Ricciardi-Castagnoli P., Kohler C., Cesura A. M. (1996). Regulation of the kynurenine metabolic pathway by interferon-gamma in murine cloned macrophages and microglial cells. J. Neurochem. 66, 996–1004. 10.1046/j.1471-4159.1996.66030996.x [DOI] [PubMed] [Google Scholar]
  7. Al-Diwani A. A. J., Pollak T. A., Irani S. R., Lennox B. R. (2017). Psychosis: an autoimmune disease? Immunology. [Epub ahead of print]. 10.1111/imm.12795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Aleman A., Hijman R., de Haan E. H., Kahn R. S. (1999). Memory impairment in schizophrenia: a meta-analysis. Am. J. Psychiatry 156, 1358–1366. [DOI] [PubMed] [Google Scholar]
  9. Aleman A., Kahn R. S. (2005). Strange feelings: do amygdala abnormalities dysregulate the emotional brain in schizophrenia? Prog. Neurobiol. 77, 283–298. 10.1016/j.pneurobio.2005.11.005 [DOI] [PubMed] [Google Scholar]
  10. Aleman A., Kahn R. S., Selten J. (2003). Sex differences in the risk of schizophrenia: evidence from meta-analysis. Arch. Gen. Psychiatry 60, 565–571. 10.1001/archpsyc.60.6.565 [DOI] [PubMed] [Google Scholar]
  11. Allen A. P., Dinan T. G., Clarke G., Cryan J. F. (2017). A psychology of the human brain-gut-microbiome axis. Soc. Pers. Psychol. Compass 11, e12309. 10.1111/spc3.12309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Allen A. P., Hutch W., Borre Y. E., Kennedy P. J., Temko A., Boylan G., et al. (2016). Bifidobacterium longum 1714 as a translational psychobiotic: modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl. Psychiatry 6:e939. 10.1038/tp.2016.191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Anacker C., Hen R. (2017). Adult hippocampal neurogenesis and cognitive flexibility [mdash] linking memory and mood. Nat. Rev. Neurosci. 18, 335–346. 10.1038/nrn.2017.45 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Angelakis E., Million M., Kankoe S., Lagier J. C., Armougom F., Giorgi R., et al. (2014). Abnormal weight gain and gut microbiota modifications are side effects of long-term doxycycline and hydroxychloroquine treatment. Antimicrob. Agents Chemother. 58, 3342–3347. 10.1128/AAC.02437-14 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  15. Arentsen T., Qian Y., Gkotzis S., Femenia T., Wang T., Udekwu K., et al. (2017). The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior. Mol. Psychiatry 22, 257–266. 10.1038/mp.2016.182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Arentsen T., Raith H., Qian Y., Forssberg H., Heijtz R. D. (2015). Host microbiota modulates development of social preference in mice. Microb. Ecol. Health Dis. 26:29719. 10.3402/mehd.v26.29719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Arias I., Sorlozano A., Villegas E., Luna J. D. D., McKenney K., Cervilla J., et al. (2012). Infectious agents associated with schizophrenia: a meta-analysis. Schizophr. Res. 136, 128–136. 10.1016/j.schres.2011.10.026 [DOI] [PubMed] [Google Scholar]
  18. Arons M. H., Lee K. (2016). Shank3 is part of a zinc-sensitive signaling system that regulates excitatory synaptic strength. J. Neurosci. 36, 9124–9134. 10.1523/JNEUROSCI.0116-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Arrieta M.-C., Stiemsma L. T., Amenyogbe N., Brown E. M., Finlay B. (2014). The intestinal microbiome in early life: health and disease. Front. Immunol. 5:427. 10.3389/fimmu.2014.00427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Arrieta M. C., Walter J., Finlay B. B. (2016). Human microbiota-associated mice: a model with challenges. Cell Host Microbe 19, 575–578. 10.1016/j.chom.2016.04.014 [DOI] [PubMed] [Google Scholar]
  21. Asan E., Steinke M., Lesch K. P. (2013). Serotonergic innervation of the amygdala: targets, receptors, and implications for stress and anxiety. Histochem. Cell Biol. 139, 785–813. 10.1007/s00418-013-1081-1 [DOI] [PubMed] [Google Scholar]
  22. Ashwood P., Krakowiak P., Hertz-Picciotto I., Hansen R., Pessah I., Van de Water J. (2011). Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav. Immun. 25, 40–45. 10.1016/j.bbi.2010.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Atladottir H. O., Thorsen P., Ostergaard L., Schendel D. E., Lemcke S., Abdallah M., et al. (2010). Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J. Autism Dev. Disord. 40, 1423–1430. 10.1007/s10803-010-1006-y [DOI] [PubMed] [Google Scholar]
  24. Averbeck B. B., Costa V. D. (2017). Motivational neural circuits underlying reinforcement learning. Nat. Neurosci. 20, 505–512. 10.1038/nn.4506 [DOI] [PubMed] [Google Scholar]
  25. Azevedo F. A., Carvalho L. R., Grinberg L. T., Farfel J. M., Ferretti R. E., Leite R. E., et al. (2009). Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J. Comp. Neurol. 513, 532–541. 10.1002/cne.21974 [DOI] [PubMed] [Google Scholar]
  26. Bach D. R., Tzovara A., Vunder J. (2017). Blocking human fear memory with the matrix metalloproteinase inhibitor doxycycline. Mol. Psychiatry. [Epub ahead of print]. 10.1038/mp.2017.65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Backhed F., Fraser C. M., Ringel Y., Sanders M. E., Sartor R. B., Sherman P. M., et al. (2012). Defining a healthy human gut microbiome: current concepts, future directions, and clinical applications. Cell Host Microbe 12, 611–622. 10.1016/j.chom.2012.10.012 [DOI] [PubMed] [Google Scholar]
  28. Badawy A. A. B. (2017). Tryptophan availability for kynurenine pathway metabolism across the life span: control mechanisms and focus on aging, exercise, diet and nutritional supplements. Neuropharmacology 112(Pt B), 248–263. 10.1016/j.neuropharm.2015.11.015 [DOI] [PubMed] [Google Scholar]
  29. Bahr S. M., Tyler B. C., Wooldridge N., Butcher B. D., Burns T. L., Teesch L. M., et al. (2015). Use of the second-generation antipsychotic, risperidone, and secondary weight gain are associated with an altered gut microbiota in children. Transl. Psychiatry 5:e652. 10.1038/tp.2015.135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Bahra S. M., Weidemann B. J., Castro A. N., Walsh J. W., deLeon O., Burnett C. M., et al. (2015). Risperidone-induced weight gain is mediated through shifts in the gut microbiome and suppression of energy expenditure. EBioMedicine 2, 1725–1734. 10.1016/j.ebiom.2015.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Bailey M. T., Coe C. L. (1999). Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev. Psychobiol. 35, 146–155. [PubMed] [Google Scholar]
  32. Bailey M. T., Dowd S. E., Galley J. D., Hufnagle A. R., Allen R. G., Lyte M. (2011). Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain Behav. Immun. 25, 397–407. 10.1016/j.bbi.2010.10.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Bakken T. E., Miller J. A., Ding S. L., Sunkin S. M., Smith K. A., Ng L., et al. (2016). A comprehensive transcriptional map of primate brain development. Nature 535, 367–375. 10.1038/nature18637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Baron-Cohen S. (2017). Editorial perspective: neurodiversity - a revolutionary concept for autism and psychiatry. J. Child Psychol. Psychiatry 58, 744–747. 10.1111/jcpp.12703 [DOI] [PubMed] [Google Scholar]
  35. Barrett E., Ross R. P., O'Toole P. W., Fitzgerald G. F., Stanton C. (2012). gamma-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 113, 411–417. 10.1111/j.1365-2672.2012.05344.x [DOI] [PubMed] [Google Scholar]
  36. Barry S., Clarke G., Scully P., Dinan T. (2009). Kynurenine pathway in psychosis: evidence of increased tryptophan degradation. J. Psychopharmacol. 23, 287–294. 10.1177/0269881108089583 [DOI] [PubMed] [Google Scholar]
  37. Bassett D. S., Sporns O. (2017). Network neuroscience. Nat. Neurosci. 20, 353–364. 10.1038/nn.4502 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Behr C., Kamp H., Fabian E., Krennrich G., Mellert W., Peter E., et al. (2017). Gut microbiome-related metabolic changes in plasma of antibiotic-treated rats. Arch. Toxicol. [Epub ahead of print]. 10.1007/s00204-017-1949-2 [DOI] [PubMed] [Google Scholar]
  39. Benros M. E., Mortensen P. B., Eaton W. W. (2012). Autoimmune diseases and infections as risk factors for schizophrenia. Ann. N.Y. Acad. Sci. 1262, 56–66. 10.1111/j.1749-6632.2012.06638.x [DOI] [PubMed] [Google Scholar]
  40. Bercik P., Denou E., Collins J., Jackson W., Lu J., Jury J., et al. (2011). The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 141, 599–609, 609.e1–3. 10.1053/j.gastro.2011.04.052 [DOI] [PubMed] [Google Scholar]
  41. Berding K., Donovan S. M. (2016). Microbiome and nutrition in autism spectrum disorder: current knowledge and research needs. Nutr. Rev. 74, 723–736. 10.1093/nutrit/nuw048 [DOI] [PubMed] [Google Scholar]
  42. Berger M., Gray J. A., Roth B. L. (2009). The expanded biology of serotonin. Annu. Rev. Med. 60, 355–366. 10.1146/annurev.med.60.042307.110802 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Beste C., Getzmann S., Gajewski P. D., Golka K., Falkenstein M. (2014). Latent Toxoplasma gondii infection leads to deficits in goal-directed behavior in healthy elderly. Neurobiol. Aging 35, 1037–1044. 10.1016/j.neurobiolaging.2013.11.012 [DOI] [PubMed] [Google Scholar]
  44. Bhadra R., Cobb D. A., Weiss L. M., Khan I. A. (2013). Psychiatric disorders in toxoplasma seropositive patients–the CD8 connection. Schizophr. Bull. 39, 485–489. 10.1093/schbul/sbt006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Bilbo S. D., Smith S. H., Schwarz J. M. (2012). A lifespan approach to neuroinflammatory and cognitive disorders: a critical role for glia. J. Neuroimmun. Pharmacol. 7, 24–41. 10.1007/s11481-011-9299-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Bishop S. L., Farmer C., Bal V., Robinson E. B., Willsey A. J., Werling D. M., et al. (2017). Identification of developmental and behavioral markers associated with genetic abnormalities in autism spectrum disorder. Am. J. Psychiatry 174, 576–585. 10.1176/appi.ajp.2017.16101115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Blandino P., Jr., Barnum C. J., Solomon L. G., Larish Y., Lankow B. S., Deak T. (2009). Gene expression changes in the hypothalamus provide evidence for regionally-selective changes in IL-1 and microglial markers after acute stress. Brain Behav. Immun. 23, 958–968. 10.1016/j.bbi.2009.04.013 [DOI] [PubMed] [Google Scholar]
  48. Bloomfield P. S., Selvaraj S., Veronese M., Rizzo G., Bertoldo A., Owen D. R., et al. (2016). Microglial activity in people at ultra high risk of psychosis and in schizophrenia: an [(11)C]PBR28 PET brain imaging study. Am. J. Psychiatry 173, 44–52. 10.1176/appi.ajp.2015.14101358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Bollinger J. L., Bergeon Burns C. M., Wellman C. L. (2016). Differential effects of stress on microglial cell activation in male and female medial prefrontal cortex. Brain Behav. Immun. 52, 88–97. 10.1016/j.bbi.2015.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Bordenstein S. R., Theis K. R. (2015). Host biology in light of the microbiome: ten principles of holobionts and hologenomes. PLoS Biol. 13:e1002226. 10.1371/journal.pbio.1002226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Borre Y. E., O'Keeffe G. W., Clarke G., Stanton C., Dinan T. G., Cryan J. F. (2014). Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol. Med. 20, 509–518. 10.1016/j.molmed.2014.05.002 [DOI] [PubMed] [Google Scholar]
  52. Bourassa M. W., Alim I., Bultman S. J., Ratan R. R. (2016). Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? Neurosci. Lett. 625, 56–63. 10.1016/j.neulet.2016.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Braniste V., Al-Asmakh M., Kowal C., Anuar F., Abbaspour A., Toth M., et al. (2014). The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 6:263ra158. 10.1126/scitranslmed.3009759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Bravo J. A., Forsythe P., Chew M. V., Escaravage E., Savignac H. M., Dinan T. G., et al. (2011). Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. U.S.A. 108, 16050–16055. 10.1073/pnas.1102999108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Brenhouse H. C., Schwarz J. M. (2016). Immunoadolescence: neuroimmune development and adolescent behavior. Neurosci. Biobehav. Rev. 70, 288–299. 10.1016/j.neubiorev.2016.05.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Brown A. S. (2012). Epidemiologic studies of exposure to prenatal infection and risk of schizophrenia and autism. Dev. Neurobiol. 72, 1272–1276. 10.1002/dneu.22024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Bruce-Keller A. J., Salbaum J. M., Luo M., Blanchard E. T., Taylor C. M., Welsh D. A., et al. (2015). Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol. Psychiatry 77, 607–615. 10.1016/j.biopsych.2014.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Bsibsi M., Ravid R., Gveric D., van Noort J. M. (2002). Broad expression of Toll-like receptors in the human central nervous system. J. Neuropathol. Exp. Neurol. 61, 1013–1021. 10.1093/jnen/61.11.1013 [DOI] [PubMed] [Google Scholar]
  59. Buckley P. F., Miller B. J., Lehrer D. S., Castle D. J. (2009). Psychiatric Comorbidities and Schizophrenia. Schizophr. Bull. 35, 383–402. 10.1093/schbul/sbn135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Buffington S. A., Di Prisco G. V., Auchtung T. A., Ajami N. J., Petrosino J. F., Costa-Mattioli M. (2016). Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell 165, 1762–1775. 10.1016/j.cell.2016.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Buie T., Fuchs G. J., III., Furuta G. T., Kooros K., Levy J., Lewis J. D., et al. (2010). Recommendations for evaluation and treatment of common gastrointestinal problems in children with ASDs. Pediatrics 125(Suppl. 1), S19–29. 10.1542/peds.2009-1878D [DOI] [PubMed] [Google Scholar]
  62. Burokas A., Arboleya S., Moloney R. D., Peterson V. L., Murphy K., Clarke G., et al. (in press). Targeting the microbiota-gut-brain axis: prebiotics have anxiolytic antidepressant-like effects reverse the impact of chronic stress in mice. Biol. Psychiatry. 10.1016/j.biopsych.2016.12.031 [DOI] [PubMed] [Google Scholar]
  63. Cai L., Chen T., Yang J., Zhou K., Yan X., Chen W., et al. (2015). Serum trace element differences between Schizophrenia patients and controls in the Han Chinese population. Sci. Rep. 5:15013. 10.1038/srep15013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Calcia M. A., Bonsall D. R., Bloomfield P. S., Selvaraj S., Barichello T., Howes O. D. (2016). Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology 233, 1637–1650. 10.1007/s00213-016-4218-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Cani P. D. (2017). Gut microbiota—at the intersection of everything? Nat. Rev. Gastroenterol. Hepatol. 14, 321–322. 10.1038/nrgastro.2017.54 [DOI] [PubMed] [Google Scholar]
  66. Canitano R., Pallagrosi M. (2017). Autism spectrum disorders and schizophrenia spectrum disorders: excitation/inhibition imbalance and developmental trajectories. Front. Psychiatry 8:69. 10.3389/fpsyt.2017.00069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Cao X., Lin P., Jiang P., Li C. (2013). Characteristics of the gastrointestinal microbiome in children with autism spectrum disorder: a systematic review. Shanghai Arch. Psychiatry 25, 342–353. 10.3969/j.issn.1002-0829.2013.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Capuron L., Pagnoni G., Drake D. F., Woolwine B. J., Spivey J. R., Crowe R. J., et al. (2012). Dopaminergic mechanisms of reduced basal ganglia responses to hedonic reward during interferon alfa administration. Arch. Gen. Psychiatry 69, 1044–1053. 10.1001/archgenpsychiatry.2011.2094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Careaga M., Murai T., Bauman M. D. (2017). Maternal immune activation and autism spectrum disorder: from rodents to nonhuman and human primates. Biol. Psychiatry 81, 391–401. 10.1016/j.biopsych.2016.10.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Carlson A. L., Xia K., Azcarate-Peril M. A., Goldman B. D., Ahn M., Styner M. A., et al. (in press). Infant gut microbiome associated with cognitive development. Biol. Psychiatry. 10.1016/j.biopsych.2017.06.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Carruthers V. B., Suzuki Y. (2007). Effects of Toxoplasma gondii infection on the brain. Schizophr. Bull. 33, 745–751. 10.1093/schbul/sbm008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Casey B. J., Oliveri M. E., Insel T. (2014). A neurodevelopmental perspective on the research domain criteria (RDoC) framework. Biol. Psychiatry 76, 350–353. 10.1016/j.biopsych.2014.01.006 [DOI] [PubMed] [Google Scholar]
  73. Castro-Nallar E., Bendall M. L., Perez-Losada M., Sabuncyan S., Severance E. G., Dickerson F. B., et al. (2015). Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ 3:e1140. 10.7717/peerj.1140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Cermak S. A., Curtin C., Bandini L. G. (2010). Food selectivity and sensory sensitivity in children with autism spectrum disorders. J. Am. Diet. Assoc. 110, 238–246. 10.1016/j.jada.2009.10.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Chen X., D'Souza R., Hong S. T. (2013). The role of gut microbiota in the gut-brain axis: current challenges and perspectives. Protein Cell 4, 403–414. 10.1007/s13238-013-3017-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Chiappelli J., Pocivavsek A., Nugent K. L., Notarangelo F. M., Kochunov P., Rowland L. M., et al. (2014). Stress-induced increase in kynurenic acid as a potential biomarker for patients with schizophrenia and distress intolerance. JAMA Psychiatry 71, 761–768. 10.1001/jamapsychiatry.2014.243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Chistiakov D. A., Bobryshev Y. V., Kozarov E., Sobenin I. A., Orekhov A. N. (2014). Intestinal mucosal tolerance and impact of gut microbiota to mucosal tolerance. Front. Microbiol. 5:781. 10.3389/fmicb.2014.00781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Christensen D. L., Baio J., Van Naarden Braun K., Bilder D., Charles J., Constantino J. N., et al. (2016). Prevalence and characteristics of autism spectrum disorder among children aged 8 years–autism and developmental disabilities monitoring network, 11 sites, United States, 2012. MMWR. Surveill. Summ. 65, 1–23. 10.15585/mmwr.ss6503a1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Christian L. M., Galley J. D., Hade E. M., Schoppe-Sullivan S., Kamp Dush C., Bailey M. T. (2015). Gut microbiome composition is associated with temperament during early childhood. Brain Behav. Immun. 45, 118–127. 10.1016/j.bbi.2014.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Chu D. M., Ma J., Prince A. L., Antony K. M., Seferovic M. D., Aagaard K. M. (2017). Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 23, 314–326. 10.1038/nm.4272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Clarke G., Grenham S., Scully P., Fitzgerald P., Moloney R. D., Shanahan F., et al. (2013). The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 18, 666–673. 10.1038/mp.2012.77 [DOI] [PubMed] [Google Scholar]
  82. Cohen A. S., Le T. P., Fedechko T. L., Elvevag B. (2017). Can RDoC help find order in thought disorder? Schizophr. Bull. 43, 503–508. 10.1093/schbul/sbx030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Coiro P., Padmashri R., Suresh A., Spartz E., Pendyala G., Chou S., et al. (2015). Impaired synaptic development in a maternal immune activation mouse model of neurodevelopmental disorders. Brain Behav. Immun. 50, 249–258. 10.1016/j.bbi.2015.07.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Collste K., Plaven-Sigray P., Fatouros-Bergman H., Victorsson P., Schain M., Forsberg A., et al. (2017). Lower levels of the glial cell marker TSPO in drug-naive first-episode psychosis patients as measured using PET and [lsqb]11C[rsqb]PBR28. Mol. Psychiatry 22, 850–856. 10.1038/mp.2016.247 [DOI] [PubMed] [Google Scholar]
  85. Conti E., Mitra J., Calderoni S., Pannek K., Shen K. K., Pagnozzi A., et al. (2017). Network over-connectivity differentiates autism spectrum disorder from other developmental disorders in toddlers: a diffusion MRI study. Hum. Brain Mapp. 38, 2333–2344. 10.1002/hbm.23520 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Corvin A., Morris D. W. (2014). Genome-wide association studies: findings at the major histocompatibility complex locus in psychosis. Biol. Psychiatry 75, 276–283. 10.1016/j.biopsych.2013.09.018 [DOI] [PubMed] [Google Scholar]
  87. Croen L. A., Grether J. K., Selvin S. (2002). Descriptive epidemiology of autism in a California population: who is at risk? J. Autism Dev. Disord. 32, 217–224. 10.1023/A:1015405914950 [DOI] [PubMed] [Google Scholar]
  88. Crumeyrolle-Arias M., Jaglin M., Bruneau A., Vancassel S., Cardona A., Dauge V., et al. (2014). Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 42, 207–217. 10.1016/j.psyneuen.2014.01.014 [DOI] [PubMed] [Google Scholar]
  89. Cryan J. F., Dinan T. G. (2015). Gut microbiota: microbiota and neuroimmune signalling-Metchnikoff to microglia. Nat. Rev. Gastroenterol. Hepatol. 12, 494–496. 10.1038/nrgastro.2015.127 [DOI] [PubMed] [Google Scholar]
  90. Curran E. A., Cryan J. F., Kenny L. C., Dinan T. G., Kearney P. M., Khashan A. S. (2016). Obstetrical mode of delivery and childhood behavior and psychological development in a British Cohort. J. Autism Dev. Disord. 46, 603–614. 10.1007/s10803-015-2616-1 [DOI] [PubMed] [Google Scholar]
  91. Curran E. A., Dalman C., Kearney P. M., Kenny L. C., Cryan J. F., Dinan T. G., et al. (2015). Association Between obstetric mode of delivery and autism spectrum disorder: a population-based sibling design study. JAMA Psychiatry 72, 935–942. 10.1001/jamapsychiatry.2015.0846 [DOI] [PubMed] [Google Scholar]
  92. Curran E. A., O'Neill S. M., Cryan J. F., Kenny L. C., Dinan T. G., Khashan A. S., et al. (2014). Research Review: birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry. 56, 500–508. 10.1111/jcpp.12351 [DOI] [PubMed] [Google Scholar]
  93. Dalton K. M., Nacewicz B. M., Johnstone T., Schaefer H. S., Gernsbacher M. A., Goldsmith H. H., et al. (2005). Gaze fixation and the neural circuitry of face processing in autism. Nat. Neurosci. 8, 519–526. 10.1038/nn1421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Dalton V. S., Kolshus E., McLoughlin D. M. (2014). Epigenetics and depression: return of the repressed. J. Affect. Disord. 155, 1–12. 10.1016/j.jad.2013.10.028 [DOI] [PubMed] [Google Scholar]
  95. Dantzer R., O'Connor J. C., Freund G. G., Johnson R. W., Kelley K. W. (2008). From inflammation to sickness and depression: when the immune system subjugates the brain. Nat. Rev. Neurosci. 9, 46–56. 10.1038/nrn2297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Davari S., Talaei S. A., Alaei H., Salami M. (2013). Probiotics treatment improves diabetes-induced impairment of synaptic activity and cognitive function: behavioral and electrophysiological proofs for microbiome-gut-brain axis. Neuroscience 240, 287–296. 10.1016/j.neuroscience.2013.02.055 [DOI] [PubMed] [Google Scholar]
  97. Davey K. J., Cotter P. D., O'Sullivan O., Crispie F., Dinan T. G., Cryan J. F., et al. (2013). Antipsychotics and the gut microbiome: olanzapine-induced metabolic dysfunction is attenuated by antibiotic administration in the rat. Transl. Psychiatry 3:e309. 10.1038/tp.2013.83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Davey K. J., O'Mahony S. M., Schellekens H., O'Sullivan O., Bienenstock J., Cotter P. D., et al. (2012). Gender-dependent consequences of chronic olanzapine in the rat: effects on body weight, inflammatory, metabolic and microbiota parameters. Psychopharmacology 221, 155–169. 10.1007/s00213-011-2555-2 [DOI] [PubMed] [Google Scholar]
  99. Day R., Nielsen J. A., Korten A., Ernberg G., Dube K. C., Gebhart J., et al. (1987). Stressful life events preceding the acute onset of schizophrenia: a cross-national study from the World Health Organization. Cult. Med. Psychiatry 11, 123–205. 10.1007/BF00122563 [DOI] [PubMed] [Google Scholar]
  100. Dayan P., Huys Q. J. (2008). Serotonin, inhibition, and negative mood. PLoS Comput. Biol. 4:e4. 10.1371/journal.pcbi.0040004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. De Angelis M., Piccolo M., Vannini L., Siragusa S., De Giacomo A., Serrazzanetti D. I., et al. (2013). Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. PLoS ONE 8:e76993 10.1371/journal.pone.0076993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. De Filippis F., Pellegrini N., Vannini L., Jeffery I. B., La Storia A., Laghi L., et al. (2015). High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 65, 1812–1821. 10.1136/gutjnl-2015-309957 [DOI] [PubMed] [Google Scholar]
  103. Deng W., Aimone J. B., Gage F. H. (2010). New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat. Rev. Neurosci. 11, 339–350. 10.1038/nrn2822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. De Palma G., Blennerhassett P., Lu J., Deng Y., Park A. J., Green W., et al. (2015). Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nat. Commun. 6:7735. 10.1038/ncomms8735 [DOI] [PubMed] [Google Scholar]
  105. De Palma G., Lynch M. D. (2017). Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice. Sci. Transl. Med. 9:eaaf6397 9. 10.1126/scitranslmed.aaf6397 [DOI] [PubMed] [Google Scholar]
  106. Derrien M., van Hylckama Vlieg J. E. (2015). Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol. 23, 354–366. 10.1016/j.tim.2015.03.002 [DOI] [PubMed] [Google Scholar]
  107. De Rubeis S., He X., Goldberg A. P., Poultney C. S., Samocha K., Cicek A. E., et al. (2014). Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515, 209–215. 10.1038/nature13772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Desbonnet L., Clarke G., Shanahan F., Dinan T. G., Cryan J. F. (2014). Microbiota is essential for social development in the mouse. Mol. Psychiatry 19, 146–148. 10.1038/mp.2013.65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Desbonnet L., Garrett L., Clarke G., Bienenstock J., Dinan T. G. (2008). The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J. Psychiatr. Res. 43, 164–174. 10.1016/j.jpsychires.2008.03.009 [DOI] [PubMed] [Google Scholar]
  110. de Theije C. G., Wopereis H., Ramadan M., van Eijndthoven T., Lambert J., Knol J., et al. (2014a). Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain Behav. Immun. 37, 197–206. 10.1016/j.bbi.2013.12.005 [DOI] [PubMed] [Google Scholar]
  111. de Theije C. G., Wu J., Koelink P. J., Korte-Bouws G. A., Borre Y., Kas M. J., et al. (2014b). Autistic-like behavioural and neurochemical changes in a mouse model of food allergy. Behav. Brain Res. 261, 265–274. 10.1016/j.bbr.2013.12.008 [DOI] [PubMed] [Google Scholar]
  112. Dethlefsen L., McFall-Ngai M., Relman D. A. (2007). An ecological and evolutionary perspective on human-microbe mutualism and disease. Nature 449, 811–818. 10.1038/nature06245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. de Witte L., Tomasik J., Schwarz E., Guest P. C., Rahmoune H., Kahn R. S., et al. (2014). Cytokine alterations in first-episode schizophrenia patients before and after antipsychotic treatment. Schizophr. Res. 154, 23–29. 10.1016/j.schres.2014.02.005 [DOI] [PubMed] [Google Scholar]
  114. Diaz Heijtz R., Wang S., Anuar F., Qian Y., Bjorkholm B., Samuelsson A., et al. (2011). Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. U.S.A. 108, 3047–3052. 10.1073/pnas.1010529108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Dickerson F. B., Stallings C., Origoni A., Katsafanas E., Savage C. L., Schweinfurth L. A., et al. (2014). Effect of probiotic supplementation on schizophrenia symptoms and association with gastrointestinal functioning: a randomized, placebo-controlled trial. Prim. Care Companion CNS Disord. 16:13m01579. 10.4088/PCC.13m01579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Dinan T. G., Borre Y. E., Cryan J. F. (2014). Genomics of schizophrenia: time to consider the gut microbiome? Mol. Psychiatry 19, 1252–1257. 10.1038/mp.2014.93 [DOI] [PubMed] [Google Scholar]
  117. Dinan T. G., Cryan J. F. (2017). Gut instincts: microbiota as a key regulator of brain development, ageing and neurodegeneration. J. Physiol. 595, 489–503. 10.1113/JP273106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Dinan T. G., Stanton C., Cryan J. F. (2013). Psychobiotics: a novel class of psychotropic. Biol. Psychiatry 74, 720–726. 10.1016/j.biopsych.2013.05.001 [DOI] [PubMed] [Google Scholar]
  119. Dinan T. G., Stilling R. M., Stanton C., Cryan J. F. (2015). Collective unconscious: how gut microbes shape human behavior. J. Psychiatr. Res. 63, 1–9. 10.1016/j.jpsychires.2015.02.021 [DOI] [PubMed] [Google Scholar]
  120. Di Nicola M., Cattaneo A., Hepgul N., Di Forti M., Aitchison K. J., Janiri L., et al. (2013). Serum and gene expression profile of cytokines in first-episode psychosis. Brain Behav. Immun. 31, 90–95. 10.1016/j.bbi.2012.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Douglas A. E., Werren J. H. (2016). Holes in the hologenome: why host-microbe symbioses are not holobionts. MBio 7:e02099 10.1128/mBio.02099-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Dowlati Y., Herrmann N., Swardfager W., Liu H., Sham L., Reim E. K., et al. (2010). A meta-analysis of cytokines in major depression. Biol. Psychiatry 67, 446–457. 10.1016/j.biopsych.2009.09.033 [DOI] [PubMed] [Google Scholar]
  123. Dunbar R. I. (2012). The social brain meets neuroimaging. Trends Cogn. Sci. 16, 101–102. 10.1016/j.tics.2011.11.013 [DOI] [PubMed] [Google Scholar]
  124. Durkin M. S., Maenner M. J., Newschaffer C. J., Lee L. C., Cunniff C. M., Daniels J. L., et al. (2008). Advanced parental age and the risk of autism spectrum disorder. Am. J. Epidemiol. 168, 1268–1276. 10.1093/aje/kwn250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Eckburg P. B., Bik E. M., Bernstein C. N., Purdom E., Dethlefsen L., Sargent M., et al. (2005). Diversity of the human intestinal microbial flora. Science 308, 1635–1638. 10.1126/science.1110591 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. El Aidy S., Derrien M., Aardema R., Hooiveld G., Richards S. E., Dane A., et al. (2014). Transient inflammatory-like state and microbial dysbiosis are pivotal in establishment of mucosal homeostasis during colonisation of germ-free mice. Benef. Microbes 5, 67–77. 10.3920/BM2013.0018 [DOI] [PubMed] [Google Scholar]
  127. El Aidy S., Kunze W., Bienenstock J., Kleerebezem M. (2012). The microbiota and the gut-brain axis: insights from the temporal and spatial mucosal alterations during colonisation of the germfree mouse intestine. Benef. Microbes 3, 251–259. 10.3920/BM2012.0042 [DOI] [PubMed] [Google Scholar]
  128. Cantor-Graae Elizabet, Selten Jean-Paul. (2005). Schizophrenia and Migration: a meta-analysis and review. Am. J. Psychiatry 162, 12–24. 10.1176/appi.ajp.162.1.12 [DOI] [PubMed] [Google Scholar]
  129. Erhardt S., Blennow K., Nordin C., Skogh E., Lindstrom L. H., Engberg G. (2001). Kynurenic acid levels are elevated in the cerebrospinal fluid of patients with schizophrenia. Neurosci. Lett. 313, 96–98. 10.1016/S0304-3940(01)02242-X [DOI] [PubMed] [Google Scholar]
  130. Erhardt S., Pocivavsek A., Repici M., Liu X.-C., Imbeault S., Maddison D. C., et al. (in press). Adaptive behavioral changes in kynurenine 3-monooxygenase knockout mice: relevance to psychotic disorders. Biol. Psychiatry. 10.1016/j.biopsych.2016.12.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Erny D., Hrabe de Angelis A. L., Jaitin D., Wieghofer P., Staszewski O., David E., et al. (2015). Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 18, 965–977. 10.1038/nn.4030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Evans S. J., Bassis C. M., Hein R., Assari S., Flowers S. A., Kelly M. B., et al. (2017). The gut microbiome composition associates with bipolar disorder and illness severity. J. Psychiatr. Res. 87, 23–29. 10.1016/j.jpsychires.2016.12.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Facci L., Barbierato M., Marinelli C., Argentini C., Skaper S. D., Giusti P. (2014). Toll-like receptors 2, -3 and -4 prime microglia but not astrocytes across central nervous system regions for ATP-dependent interleukin-1beta release. Sci. Rep. 4:6824 10.1038/srep06824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Falony G., Joossens M., Vieira-Silva S., Wang J., Darzi Y., Faust K., et al. (2016). Population-level analysis of gut microbiome variation. Science 352, 560–564. 10.1126/science.aad3503 [DOI] [PubMed] [Google Scholar]
  135. Faludi G., Mirnics K. (2011). Synaptic changes in the brain of subjects with schizophrenia. Int. J. Dev. Neurosci. 29, 305–309. 10.1016/j.ijdevneu.2011.02.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Feigenson K. A., Kusnecov A. W., Silverstein S. M. (2014). Inflammation and the two-hit hypothesis of Schizophrenia. Neurosci. Biobehav. Rev. 38, 72–93. 10.1016/j.neubiorev.2013.11.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Felger J. C., Miller A. H. (2012). Cytokine effects on the basal ganglia and dopamine function: the subcortical source of inflammatory malaise. Front. Neuroendocrinol. 33, 315–327. 10.1016/j.yfrne.2012.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Felger J. C., Mun J., Kimmel H. L., Nye J. A., Drake D. F., Hernandez C. R., et al. (2013). Chronic interferon-alpha decreases dopamine 2 receptor binding and striatal dopamine release in association with anhedonia-like behavior in nonhuman primates. Neuropsychopharmacology 38, 2179–2187. 10.1038/npp.2013.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Fenn A. M., Gensel J. C., Huang Y., Popovich P. G., Lifshitz J., Godbout J. P. (2014). Immune activation promotes depression 1 month after diffuse brain injury: a role for primed microglia. Biol. Psychiatry 76, 575–584. 10.1016/j.biopsych.2013.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Fernandez-Real J. M., Serino M., Blasco G., Puig J., Daunis-i-Estadella J., Ricart W., et al. (2015). Gut microbiota interacts with brain microstructure and function. j. Clin. Endocrinol. Metab. 100, 4505–4513. 10.1210/jc.2015-3076 [DOI] [PubMed] [Google Scholar]
  141. Feustel S. M., Meissner M., Liesenfeld O. (2012). Toxoplasma gondii and the blood-brain barrier. Virulence 3, 182–192. 10.4161/viru.19004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Filiano A. J., Gadani S. P., Kipnis J. (2017). How and why do T cells and their derived cytokines affect the injured and healthy brain? Nat. Rev. Neurosci. 18, 375–384. 10.1038/nrn.2017.39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Finegold S. M., Dowd S. E., Gontcharova V., Liu C., Henley K. E., Wolcott R. D. (2010). Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 16, 444–453. 10.1016/j.anaerobe.2010.06.008 [DOI] [PubMed] [Google Scholar]
  144. Finegold S. M., Molitoris D., Song Y., Liu C., Vaisanen M. L., Bolte E., et al. (2002). Gastrointestinal microflora studies in late-onset autism. Clin. Infect. Dis. 35, S6–S16. 10.1086/341914 [DOI] [PubMed] [Google Scholar]
  145. Fontainhas A. M., Wang M., Liang K. J., Chen S., Mettu P., Damani M., et al. (2011). Microglial morphology and dynamic behavior is regulated by ionotropic glutamatergic and GABAergic neurotransmission. PLoS ONE 6:e15973. 10.1371/journal.pone.0015973 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Forrest C. M., Khalil O. S., Pisar M., McNair K., Kornisiuk E., Snitcofsky M., et al. (2013). Changes in synaptic transmission and protein expression in the brains of adult offspring after prenatal inhibition of the kynurenine pathway. Neuroscience 254, 241–259. 10.1016/j.neuroscience.2013.09.034 [DOI] [PubMed] [Google Scholar]
  147. Forrest C. M., McNair K., Pisar M., Khalil O. S., Darlington L. G., Stone T. W. (2015). Altered hippocampal plasticity by prenatal kynurenine administration, kynurenine-3-monoxygenase (KMO) deletion or galantamine. Neuroscience 310, 91–105. 10.1016/j.neuroscience.2015.09.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Foster J. A. (2016). Gut Microbiome and behavior: focus on neuroimmune interactions. Int. Rev. Neurobiol. 131, 49–65. 10.1016/bs.irn.2016.07.005 [DOI] [PubMed] [Google Scholar]
  149. Francino M. P. (2014). Early development of the gut microbiota and immune health. Pathogens 3, 769–790. 10.3390/pathogens3030769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Frank M. G., Baratta M. V., Sprunger D. B., Watkins L. R., Maier S. F. (2007). Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav. Immun. 21, 47–59. 10.1016/j.bbi.2006.03.005 [DOI] [PubMed] [Google Scholar]
  151. Franzosa E. A., Morgan X. C., Segata N., Waldron L., Reyes J., Earl A. M., et al. (2014). Relating the metatranscriptome and metagenome of the human gut. Proc. Natl. Acad. Sci. U.S.A. 111, E2329–E2338. 10.1073/pnas.1319284111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Freytag V., Carrillo-Roa T., Milnik A. (2017). A peripheral epigenetic signature of immune system genes is linked to neocortical thickness and Memory 8:15193. 10.1038/ncomms15193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Frohlich E. E., Farzi A., Mayerhofer R., Reichmann F., Jacan A., Wagner B., et al. (2016). Cognitive impairment by antibiotic-induced gut dysbiosis: analysis of gut microbiota-brain communication. Brain Behav. Immun. 56, 140–155. 10.1016/j.bbi.2016.02.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Frost G., Sleeth M. L., Sahuri-Arisoylu M., Lizarbe B., Cerdan S., Brody L., et al. (2014). The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 5:3611. 10.1038/ncomms4611 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Frye R. E., Rose S., Chacko J., Wynne R., Bennuri S. C., Slattery J. C., et al. (2016). Modulation of mitochondrial function by the microbiome metabolite propionic acid in autism and control cell lines. Transl. Psychiatry 6:e927. 10.1038/tp.2016.189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Fujimura K. E., Sitarik A. R., Havstad S., Lin D. L., Levan S., Fadrosh D., et al. (2016). Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 22, 1187–1191. 10.1038/nm.4176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Fusar-Poli P., Tantardini M., De Simone S., Ramella-Cravaro V., Oliver D., Kingdon J., et al. (2017). Deconstructing vulnerability for psychosis: meta-analysis of environmental risk factors for psychosis in subjects at ultra high-risk. Eur. Psychiatry 40, 65–75. 10.1016/j.eurpsy.2016.09.003 [DOI] [PubMed] [Google Scholar]
  158. Galley J. D., Nelson M. C., Yu Z., Dowd S. E., Walter J., Kumar P. S., et al. (2014a). Exposure to a social stressor disrupts the community structure of the colonic mucosa-associated microbiota. BMC Microbiol. 14:189. 10.1186/1471-2180-14-189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Galley J. D., Yu Z., Kumar P., Dowd S. E., Lyte M., Bailey M. T. (2014b). The structures of the colonic mucosa-associated and luminal microbial communities are distinct and differentially affected by a prolonged murine stressor. Gut Microbes 5, 748–760. 10.4161/19490976.2014.972241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. García-Bueno B., Gass,ó P., MacDowell K. S., Callado L. F., Mas S., Bernardo M., et al. (2016). Evidence of activation of the Toll-like receptor-4 proinflammatory pathway in patients with schizophrenia. J. Psychiatry Neurosci. 41, E46–E55. 10.1503/jpn.150195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Gareau M. G., Wine E., Rodrigues D. M., Cho J. H., Whary M. T., Philpott D. J., et al. (2011). Bacterial infection causes stress-induced memory dysfunction in mice. Gut 60, 307–317. 10.1136/gut.2009.202515 [DOI] [PubMed] [Google Scholar]
  162. George B., Klijn A. (2013). A modern name for schizophrenia (PSS) would diminish self-stigma. Psychol. Med. 43, 1555–1557. 10.1017/S0033291713000895 [DOI] [PubMed] [Google Scholar]
  163. Ghaziuddin M., Ghaziuddin N., Greden J. (2002). Depression in persons with autism: implications for research and clinical care. J. Autism Dev. Disord. 32, 299–306. 10.1023/A:1016330802348 [DOI] [PubMed] [Google Scholar]
  164. Giovanoli S., Engler H., Engler A., Richetto J., Voget M., Willi R., et al. (2013). Stress in puberty unmasks latent neuropathological consequences of prenatal immune activation in mice. Science 339, 1095–1099. 10.1126/science.1228261 [DOI] [PubMed] [Google Scholar]
  165. Giovanoli S., Notter T., Richetto J., Labouesse M. A., Vuillermot S., Riva M. A., et al. (2015). Late prenatal immune activation causes hippocampal deficits in the absence of persistent inflammation across aging. J. Neuroinflammation 12:221. 10.1186/s12974-015-0437-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Goines P. E., Croen L. A., Braunschweig D., Yoshida C. K., Grether J., Hansen R., et al. (2011). Increased midgestational IFN-γ, IL-4 and IL-5 in women bearing a child with autism: a case-control study. Mol. Autism 2:13. 10.1186/2040-2392-2-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Golubeva A. V., Crampton S., Desbonnet L., Edge D., O'Sullivan O., Lomasney K. W., et al. (2015). Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood. Psychoneuroendocrinology 60, 58–74. 10.1016/j.psyneuen.2015.06.002 [DOI] [PubMed] [Google Scholar]
  168. Gondalia S. V., Palombo E. A., Knowles S. R., Cox S. B., Meyer D., Austin D. W. (2012). Molecular characterisation of gastrointestinal microbiota of children with autism (with and without gastrointestinal dysfunction) and their neurotypical siblings. Autism Res. 5, 419–427. 10.1002/aur.1253 [DOI] [PubMed] [Google Scholar]
  169. Gonzalez-Burgos G., Lewis D. A. (2012). NMDA receptor hypofunction, parvalbumin-positive neurons, and cortical gamma oscillations in schizophrenia. Schizophr. Bull. 38, 950–957. 10.1093/schbul/sbs010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Grabrucker S., Jannetti L., Eckert M., Gaub S., Chhabra R., Pfaender S., et al. (2014). Zinc deficiency dysregulates the synaptic ProSAP/Shank scaffold and might contribute to autism spectrum disorders. Brain 137, 137–152. 10.1093/brain/awt303 [DOI] [PubMed] [Google Scholar]
  171. Graeber M. B., Streit W. J. (2010). Microglia: biology and pathology. Acta Neuropathol. 119, 89–105. 10.1007/s00401-009-0622-0 [DOI] [PubMed] [Google Scholar]
  172. Graham A. M., Rasmussen J. M., Rudolph M. D., Heim C. M., Gilmore J. H., Styner M., et al. (in press). Maternal Systemic interleukin-6 during pregnancy is associated with newborn amygdala phenotypes subsequent behavior at 2 years of age. Biol. Psychiatry. 10.1016/j.biopsych.2017.05.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Gramsbergen J. B., Hodgkins P. S., Rassoulpour A., Turski W. A., Guidetti P., Schwarcz R. (1997). Brain-specific modulation of kynurenic acid synthesis in the rat. J. Neurochem. 69, 290–298. 10.1046/j.1471-4159.1997.69010290.x [DOI] [PubMed] [Google Scholar]
  174. Green M. J., Matheson S. L., Shepherd A., Weickert C. S., Carr V. J. (2011). Brain-derived neurotrophic factor levels in schizophrenia: a systematic review with meta-analysis. Mol. Psychiatry 16, 960–972. 10.1038/mp.2010.88 [DOI] [PubMed] [Google Scholar]
  175. Grosse Wiesmann C., Schreiber J., Singer T., Steinbeis N., Friederici A. D. (2017). White matter maturation is associated with the emergence of Theory of Mind in early childhood. Nat. Commun. 8:14692. 10.1038/ncomms14692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Guarner F., Bourdet-Sicard R., Brandtzaeg P., Gill H. S., McGuirk P., van Eden W. (2006). Mechanisms of disease: the hygiene hypothesis revisited. Nat. Clin. Pract. Gastroenterol. Hepatol. 3, 275–284. 10.1038/ncpgasthep0471 [DOI] [PubMed] [Google Scholar]
  177. Guidetti P., Hoffman G. E., Melendez-Ferro M., Albuquerque E. X., Schwarcz R. (2007). Astrocytic localization of kynurenine aminotransferase II in the rat brain visualized by immunocytochemistry. Glia 55, 78–92. 10.1002/glia.20432 [DOI] [PubMed] [Google Scholar]
  178. Habela C. W., Song H., Ming G. L. (2016). Modeling synaptogenesis in schizophrenia and autism using human iPSC derived neurons. Mol. Cell. Neurosci. 73, 52–62. 10.1016/j.mcn.2015.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Haddad L., Schäfer A., Streit F., Lederbogen F., Grimm O., Wüst S., et al. (2015). Brain structure correlates of urban upbringing, an environmental risk factor for Schizophrenia. Schizophr. Bull. 41, 115–122. 10.1093/schbul/sbu072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Hanage W. P. (2014). Microbiology: microbiome science needs a healthy dose of scepticism. Nature 512, 247–248. 10.1038/512247a [DOI] [PubMed] [Google Scholar]
  181. Haroon E., Raison C. L., Miller A. H. (2012). Psychoneuroimmunology meets neuropsychopharmacology: translational implications of the impact of inflammation on behavior. Neuropsychopharmacology 37, 137–162. 10.1038/npp.2011.205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Harrison N. A., Brydon L., Walker C., Gray M. A., Steptoe A., Critchley H. D. (2009). Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol. Psychiatry 66, 407–414. 10.1016/j.biopsych.2009.03.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Havlicek J., Gasova Z. G., Smith A. P., Zvara K., Flegr J. (2001). Decrease of psychomotor performance in subjects with latent ‘asymptomatic’ toxoplasmosis. Parasitology 122, 515–520. 10.1017/S0031182001007624 [DOI] [PubMed] [Google Scholar]
  184. Hazlett H. C., Gu H., Munsell B. C., Kim S. H., Styner M., Wolff J. J., et al. (2017). Early brain development in infants at high risk for autism spectrum disorder. Nature 542, 348–351. 10.1038/nature21369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Hibberd M. C., Wu M., Rodionov D. A., Li X., Cheng J., Griffin N. W., et al. (2017). The effects of micronutrient deficiencies on bacterial species from the human gut microbiota. Sci. Transl. Med. 9:eaal4069. 10.1126/scitranslmed.aal4069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Hidaka B. H. (2012). Depression as a disease of modernity: explanations for increasing prevalence. J. Affect. Disord. 140, 205–214. 10.1016/j.jad.2011.12.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Hinwood M., Morandini J., Day T. A., Walker F. R. (2011). Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb. Cortex 22, 1442–1454. 10.1093/cercor/bhr229 [DOI] [PubMed] [Google Scholar]
  188. Hinwood M., Tynan R. J., Charnley J. L., Beynon S. B., Day T. A., Walker F. R. (2012). Chronic stress induced remodeling of the prefrontal cortex: structural re-organization of microglia and the inhibitory effect of minocycline. Cereb. Cortex 23, 1784–1797. 10.1093/cercor/bhs151 [DOI] [PubMed] [Google Scholar]
  189. Hoban A. E., Moloney R. D., Golubeva A. V., McVey Neufeld K. A., O'Sullivan O., Patterson E., et al. (2016a). Behavioural and neurochemical consequences of chronic gut microbiota depletion during adulthood in the rat. Neuroscience 339, 463–477. 10.1016/j.neuroscience.2016.10.003 [DOI] [PubMed] [Google Scholar]
  190. Hoban A. E., Stilling R. M., Moloney G., Shanahan F., Dinan T. G., Clarke G., et al. (2017). The microbiome regulates amygdala-dependent fear recall. Mol. Psychiatry. [Epub ahead of print]. 10.1038/mp.2017.100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Hoban A. E., Stilling R. M., Ryan F. J., Shanahan F., Dinan T. G., Claesson M. J., et al. (2016b). Regulation of prefrontal cortex myelination by the microbiota. Transl. Psychiatry 6:e774. 10.1038/tp.2016.42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Hodes G. E., Kana V., Menard C. (2015). Neuroimmune mechanisms of Depression 18, 1386–1393. 10.1038/nn.4113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Holmes S. E., Hinz R., Drake R. J., Gregory C. J., Conen S., Matthews J. C., et al. (2016). In vivo imaging of brain microglial activity in antipsychotic-free and medicated schizophrenia: a [11C](R)-PK11195 positron emission tomography study. Mol. Psychiatry 21, 1672–1679. 10.1038/mp.2016.180 [DOI] [PubMed] [Google Scholar]
  194. Hommer R. E., Swedo S. E. (2015). Schizophrenia and autism—related disorders. Schizophr. Bull. 41, 313–314. 10.1093/schbul/sbu188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Hor K., Taylor M. (2010). Suicide and schizophrenia: a systematic review of rates and risk factors. J. Psychopharmacol. 24, 81–90. 10.1177/1359786810385490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Hsiao E. Y., McBride S. W., Hsien S., Sharon G., Hyde E. R., McCue T., et al. (2013). Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopment disorders. Cell 155, 1451–1463. 10.1016/j.cell.2013.11.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Hu W., MacDonald M. L., Elswick D. E., Sweet R. A. (2015). The glutamate hypothesis of schizophrenia: evidence from human brain tissue studies. Ann. N.Y. Acad. Sci. 1338, 38–57. 10.1111/nyas.12547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Hueston C. M., Cryan J. F., Nolan Y. M. (2017). Stress and adolescent hippocampal neurogenesis: diet and exercise as cognitive modulators. Transl. Psychiatry 7:e1081. 10.1038/tp.2017.48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Insel T., Cuthbert B., Garvey M., Heinssen R., Pine D. S., Quinn K., et al. (2010). Research domain criteria (RDoC): toward a new classification framework for research on mental disorders. Am. J. Psychiatry 167, 748–751. 10.1176/appi.ajp.2010.09091379 [DOI] [PubMed] [Google Scholar]
  200. Jacka F. N. (2017). Nutritional psychiatry: where to next? EBioMedicine 17, 24–29. 10.1016/j.ebiom.2017.02.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Jacka F. N., O'Neil A., Opie R., Itsiopoulos C., Cotton S., Mohebbi M., et al. (2017). A randomised controlled trial of dietary improvement for adults with major depression (the ‘SMILES’ trial). BMC Med. 15:23. 10.1186/s12916-017-0791-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Jacka F. N., Pasco J. A., Mykletun A., Williams L. J., Hodge A. M., O'Reilly S. L., et al. (2010). Association of Western and traditional diets with depression and anxiety in women. Am. J. Psychiatry 167, 305–311. 10.1176/appi.ajp.2009.09060881 [DOI] [PubMed] [Google Scholar]
  203. Jacob J., Vanessa R., Steve M., Sean C., David W., James B. (2013). A chemical-genetics approach to study the molecular pathology of central serotonin abnormalities in fetal valproate syndrome. J. Neurol. Neurosurg. Psychiatr. 84:e2 10.1136/jnnp-2013-306573.3425346970 [DOI] [Google Scholar]
  204. Janak P. H., Tye K. M. (2015). From circuits to behaviour in the amygdala. Nature 517, 284–292. 10.1038/nature14188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Janik R., Thomason L. A. M., Stanisz A. M., Forsythe P., Bienenstock J., Stanisz G. J. (2016). Magnetic resonance spectroscopy reveals oral Lactobacillus promotion of increases in brain GABA, N-acetyl aspartate and glutamate. Neuroimage 125, 988–995. 10.1016/j.neuroimage.2015.11.018 [DOI] [PubMed] [Google Scholar]
  206. Jasarevic E., Howerton C. L., Howard C. D., Bale T. L. (2015). Alterations in the Vaginal Microbiome by maternal stress are associated with metabolic reprogramming of the offspring gut and brain. Endocrinology 156, 3265–3276. 10.1210/en.2015-1177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Jhamnani K., Shivakumar V., Kalmady S., Rao N. P., Venkatasubramanian G. (2013). Successful use of add-on minocycline for treatment of persistent negative symptoms in schizophrenia. J. Neuropsychiatry Clin. Neurosci. 25, E06–E07. 10.1176/appi.neuropsych.11120376 [DOI] [PubMed] [Google Scholar]
  208. Jiang H., Ling Z., Zhang Y., Mao H., Ma Z., Yin Y., et al. (2015). Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 48, 186–194. 10.1016/j.bbi.2015.03.016 [DOI] [PubMed] [Google Scholar]
  209. Jiang H. Y., Xu L. L., Shao L., Xia R. M., Yu Z. H., Ling Z. X., et al. (2016). Maternal infection during pregnancy and risk of autism spectrum disorders: a systematic review and meta-analysis. Brain Behav. Immun. 58, 165–172. 10.1016/j.bbi.2016.06.005 [DOI] [PubMed] [Google Scholar]
  210. Jirtle R. L., Skinner M. K. (2007). Environmental epigenomics and disease susceptibility. Nat. Rev. Genet. 8, 253–262. 10.1038/nrg2045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Jones C. A., Watson D. J. G., Fone K. C. F. (2011). Animal models of schizophrenia. Br. J. Pharmacol. 164, 1162–1194. 10.1111/j.1476-5381.2011.01386.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Jones K. L., Croen L. A., Yoshida C. K., Heuer L., Hansen R., Zerbo O., et al. (2017). Autism with intellectual disability is associated with increased levels of maternal cytokines and chemokines during gestation. Mol. Psychiatry 22, 273–279. 10.1038/mp.2016.77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Joyce D. W., Kehagia A. A., Tracy D. K., Proctor J., Shergill S. S. (2017). Realising stratified psychiatry using multidimensional signatures and trajectories. J. Transl. Med. 15:15. 10.1186/s12967-016-1116-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Kahn R. S., Keefe R. S. (2013). Schizophrenia is a cognitive illness: time for a change in focus. JAMA Psychiatry 70, 1107–1112. 10.1001/jamapsychiatry.2013.155 [DOI] [PubMed] [Google Scholar]
  215. Kaiser T., Feng G. (2015). Modeling psychiatric disorders for developing effective treatments. Nat. Med. 21, 979–988. 10.1038/nm.3935 [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Kang D.-W., Adams J. B., Gregory A. C., Borody T., Chittick L., Fasano A., et al. (2017). Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 5:10. 10.1186/s40168-016-0225-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Kang D. W., Park J. G., Ilhan Z. E., Wallstrom G., Labaer J., Adams J. B., et al. (2013). Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PLoS ONE 8:e68322. 10.1371/journal.pone.0068322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Kau A. L., Ahern P. P., Griffin N. W., Goodman A. L., Gordon J. I. (2011). Human nutrition, the gut microbiome and the immune system. Nature 474, 327–336. 10.1038/nature10213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Kelly J. R. (2016). The Gut Microbiota in Depression. Ph.D., thesis, University College Cork. [Google Scholar]
  220. Kelly J. R., Allen A. P., Temko A., Hutch W., Kennedy P. J., Farid N., et al. (2017a). Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain Behav. Immun. 61, 50–59. 10.1016/j.bbi.2016.11.018 [DOI] [PubMed] [Google Scholar]
  221. Kelly J. R., Borre Y., C O. B., Patterson E., El Aidy S., Deane J., et al. (2016a). Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 82, 109–118. 10.1016/j.jpsychires.2016.07.019 [DOI] [PubMed] [Google Scholar]
  222. Kelly J. R., Clarke G., Cryan J. F., Dinan T. G. (2016b). Brain-gut-microbiota axis: challenges for translation in psychiatry. Ann. Epidemiol. 26, 366–372. 10.1016/j.annepidem.2016.02.008 [DOI] [PubMed] [Google Scholar]
  223. Kelly J. R., Clarke G., Cryan J. F., Dinan T. G. (2017b). Dimensional thinking in psychiatry in the era of the Research Domain Criteria (RDoC). Irish J. Psychol. Med. [Epub ahead of print]. 10.1017/ipm.2017.7 [DOI] [PubMed] [Google Scholar]
  224. Kelly J. R., Kennedy P. J., Cryan J. F., Dinan T. G., Clarke G., Hyland N. P. (2015). Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 9:392. 10.3389/fncel.2015.00392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Kennedy P. J., Cryan J. F., Dinan T. G., Clarke G. (2017). Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology 112, 399–412. 10.1016/j.neuropharm.2016.07.002 [DOI] [PubMed] [Google Scholar]
  226. Kéri S., Szab,ó C., Kelemen O. (2017). Antipsychotics influence Toll-like receptor (TLR) expression and its relationship with cognitive functions in schizophrenia. Brain Behav. Immun. 62, 256–264. 10.1016/j.bbi.2016.12.011 [DOI] [PubMed] [Google Scholar]
  227. Khalil O. S., Pisar M., Forrest C. M., Vincenten M. C., Darlington L. G., Stone T. W. (2014). Prenatal inhibition of the kynurenine pathway leads to structural changes in the hippocampus of adult rat offspring. Eur. J. Neurosci. 39, 1558–1571. 10.1111/ejn.12535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Khan N. A., Raine L. B., Drollette E. S., Scudder M. R., Kramer A. F., Hillman C. H. (2015). Dietary fiber is positively associated with cognitive control among prepubertal children. J. Nutr. 145, 143–149. 10.3945/jn.114.198457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Khandaker G. M., Pearson R. M., Zammit S., Lewis G., Jones P. B. (2014). Association of serum interleukin 6 and c-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry 71, 1121–1128. 10.1001/jamapsychiatry.2014.1332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Khodaie-Ardakani M. R., Mirshafiee O., Farokhnia M., Tajdini M., Hosseini S. M., Modabbernia A., et al. (2014). Minocycline add-on to risperidone for treatment of negative symptoms in patients with stable schizophrenia: randomized double-blind placebo-controlled study. Psychiatry Res. 215, 540–546. 10.1016/j.psychres.2013.12.051 [DOI] [PubMed] [Google Scholar]
  231. Kielian T. (2006). Toll-like receptors in central nervous system glial inflammation and homeostasis. J. Neurosci. Res. 83, 711–730. 10.1002/jnr.20767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Kirsch P., Esslinger C., Chen Q., Mier D., Lis S., Siddhanti S., et al. (2005). Oxytocin modulates neural circuitry for social cognition and fear in humans. J. Neurosci. 25, 11489–11493. 10.1523/JNEUROSCI.3984-05.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Klerman G. L., Weissman M. M. (1989). INcreasing rates of depression. JAMA 261, 2229–2235. 10.1001/jama.1989.03420150079041 [DOI] [PubMed] [Google Scholar]
  234. Knuesel I., Chicha L., Britschgi M., Schobel S. A., Bodmer M., Hellings J. A., et al. (2014). Maternal immune activation and abnormal brain development across CNS disorders. Nat. Rev. Neurol. 10, 643–660. 10.1038/nrneurol.2014.187 [DOI] [PubMed] [Google Scholar]
  235. Kohler O., Petersen L., Mors O., Mortensen P. B., Yolken R. H., Gasse C., et al. (2017). Infections and exposure to anti-infective agents and the risk of severe mental disorders: a nationwide study. Acta Psychiatr. Scand. 135, 97–105. 10.1111/acps.12671 [DOI] [PubMed] [Google Scholar]
  236. Kohman R. A., Rhodes J. S. (2013). Neurogenesis, inflammation and behavior. Brain Behav. Immun. 27, 22–32. 10.1016/j.bbi.2012.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Kolb B., Mychasiuk R., Muhammad A., Li Y., Frost D. O., Gibb R. (2012). Experience and the developing prefrontal cortex. Proc. Natl. Acad. Sci. U.S.A. 109, 17186–17193. 10.1073/pnas.1121251109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Kosmicki J. A., Samocha K. E., Howrigan D. P., Sanders S. J. (2017). Refining the role of de novo protein-truncating variants in neurodevelopmental disorders by using population reference samples. Nat. Genet. 49, 504–510. 10.1038/ng.3789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Kostic A. D., Gevers D., Siljander H., Vatanen T., Hyotylainen T., Hamalainen A. M., et al. (2015). The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe 17, 260–273. 10.1016/j.chom.2015.01.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Krabbendam L., van Os J. (2005). Schizophrenia and urbanicity: a major environmental influence–conditional on genetic risk. Schizophr. Bull. 31, 795–799. 10.1093/schbul/sbi060 [DOI] [PubMed] [Google Scholar]
  241. Labus J. S., Hollister E. B., Jacobs J., Kirbach K., Oezguen N., Gupta A., et al. (2017). Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome. Microbiome 5:49. 10.1186/s40168-017-0260-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Landry S. H., Loveland K. A. (1988). Communication behaviors in autism and developmental language delay. J. Child Psychol. Psychiatry 29, 621–634. 10.1111/j.1469-7610.1988.tb01884.x [DOI] [PubMed] [Google Scholar]
  243. Lankelma J. M., Nieuwdorp M., de Vos W. M., Wiersinga W. J. (2015). The gut microbiota in internal medicine: implications for health and disease. Neth. J. Med. 73, 61–68. [PubMed] [Google Scholar]
  244. Lartseva A., Dijkstra T., Buitelaar J. K. (2014). Emotional language processing in autism spectrum disorders: a systematic review. Front. Hum. Neurosci. 8:991. 10.3389/fnhum.2014.00991 [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Lasalvia A., Penta E., Sartorius N., Henderson S. (2015). Should the label “schizophrenia” be abandoned? Schizophr. Res. 162, 276–284. 10.1016/j.schres.2015.01.031 [DOI] [PubMed] [Google Scholar]
  246. Laursen T. M., Nordentoft M., Mortensen P. B. (2014). Excess early mortality in schizophrenia. Annu. Rev. Clin. Psychol. 10, 425–448. 10.1146/annurev-clinpsy-032813-153657 [DOI] [PubMed] [Google Scholar]
  247. Leclercq S., Mian F. M., Stanisz A. M., Bindels L. B., Cambier E., Ben-Amram H., et al. (2017). Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat. Commun. 8:15062. 10.1038/ncomms15062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Lederbogen F., Kirsch P., Haddad L., Streit F., Tost H., Schuch P., et al. (2011). City living and urban upbringing affect neural social stress processing in humans. Nature 474, 498–501. 10.1038/nature10190 [DOI] [PubMed] [Google Scholar]
  249. Levkovitz Y., Mendlovich S., Riwkes S., Braw Y., Levkovitch-Verbin H., Gal G., et al. (2010). A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. J. Clin. Psychiatry 71, 138–149. 10.4088/JCP.08m04666yel [DOI] [PubMed] [Google Scholar]
  250. Levone B. R., Cryan J. F., O'Leary O. F. (2015). Role of adult hippocampal neurogenesis in stress resilience. Neurobiol. Stress 1, 147–155. 10.1016/j.ynstr.2014.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Ley R. E., Lozupone C. A., Hamady M., Knight R., Gordon J. I. (2008). Worlds within worlds: evolution of the vertebrate gut microbiota. Nat. Rev. Microbiol. 6, 776–788. 10.1038/nrmicro1978 [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Li S.-O., Wang J.-L., Bjørklund G., Zhao W.-N., Yin C.-H. (2014). Serum copper and zinc levels in individuals with autism spectrum disorders. Neuroreport 25, 1216–1220. 10.1097/WNR.0000000000000251 [DOI] [PubMed] [Google Scholar]
  253. Lim S.-M., Jang H.-M., Jeong J.-J., Han M. J., Kim D.-H. (2017). Lactobacillus johnsonii CJLJ103 attenuates colitis and memory impairment in mice by inhibiting gut microbiota lipopolysaccharide production and NF-κB activation. J. Funct. Foods 34, 359–368. 10.1016/j.jff.2017.05.016 [DOI] [Google Scholar]
  254. Liu Y., Zhang L., Wang X., Wang Z., Zhang J., Jiang R., et al. (2016). Similar fecal microbiota signatures in patients with diarrhea-predominant irritable bowel syndrome and patients with depression. Clin. Gastroenterol. Hepatol. 14, 1602–1611.e5. 10.1016/j.cgh.2016.05.033 [DOI] [PubMed] [Google Scholar]
  255. Lombardo M. V., Moon H. M., Su J., Palmer T. D., Courchesne E., Pramparo T. (2017). Maternal immune activation dysregulation of the fetal brain transcriptome and relevance to the pathophysiology of autism spectrum disorder. Mol. Psychiatry. [Epub ahead of print]. 10.1038/mp.2017.15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Louveau A., Smirnov I., Keyes T. J., Eccles J. D., Rouhani S. J., Peske J. D., et al. (2015). Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341. 10.1038/nature14432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Lucki I. (1998). The spectrum of behaviors influenced by serotonin. Biol. Psychiatry 44, 151–162. 10.1016/S0006-3223(98)00139-5 [DOI] [PubMed] [Google Scholar]
  258. Luczynski P., Whelan S. O., O'Sullivan C., Clarke G., Shanahan F., Dinan T. G., et al. (2016). Adult microbiota-deficient mice have distinct dendritic morphological changes: differential effects in the amygdala and hippocampus. Eur. J. Neurosci. 44, 2654–2666. 10.1111/ejn.13291 [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Lurie I., Yang Y. X., Haynes K., Mamtani R., Boursi B. (2015). Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. J. Clin. Psychiatry 76, 1522–1528. 10.4088/JCP.15m09961 [DOI] [PubMed] [Google Scholar]
  260. Lyte M. (2011). Probiotics function mechanistically as delivery vehicles for neuroactive compounds: microbial endocrinology in the design and use of probiotics. Bioessays 33, 574–581. 10.1002/bies.201100024 [DOI] [PubMed] [Google Scholar]
  261. Lyte M. (2013). Microbial endocrinology in the microbiome-gut-brain axis: how bacterial production and utilization of neurochemicals influence behavior. PLoS Pathog. 9:e1003726. 10.1371/journal.ppat.1003726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Macfabe D. F. (2012). Short-chain fatty acid fermentation products of the gut microbiome: implications in autism spectrum disorders. Microb. Ecol. Health Dis. 23:19260. 10.3402/mehd.v23i0.19260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. MacFabe D. F., Cain D. P., Rodriguez-Capote K., Franklin A. E., Hoffman J. E., Boon F., et al. (2007). Neurobiological effects of intraventricular propionic acid in rats: possible role of short chain fatty acids on the pathogenesis and characteristics of autism spectrum disorders. Behav. Brain Res. 176, 149–169. 10.1016/j.bbr.2006.07.025 [DOI] [PubMed] [Google Scholar]
  264. Macfarlane G. T., Macfarlane S. (2012). Bacteria, colonic fermentation, and gastrointestinal health. J. AOAC Int. 95, 50–60. 10.5740/jaoacint.SGE_Macfarlane [DOI] [PubMed] [Google Scholar]
  265. Magnuson K. M., Constantino J. N. (2011). Characterization of depression in children with autism spectrum disorders. J. Dev. Behav. Pediatrics 32, 332–340. 10.1097/DBP.0b013e318213f56c [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Magnusson K. R., Hauck L., Jeffrey B. M., Elias V., Humphrey A., Nath R., et al. (2015). Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience 300, 128–140. 10.1016/j.neuroscience.2015.05.016 [DOI] [PubMed] [Google Scholar]
  267. Mancabelli L., Milani C., Lugli G. A., Turroni F., Ferrario C., van Sinderen D., et al. (2017). Meta-analysis of the human gut microbiome from urbanized and pre-agricultural populations. Environ. Microbiol. 19, 1379–1390. 10.1111/1462-2920.13692 [DOI] [PubMed] [Google Scholar]
  268. Marin I. A., Goertz J. E., Ren T., Rich S. S., Onengut-Gumuscu S., Farber E., et al. (2017). Microbiota alteration is associated with the development of stress-induced despair behavior. Sci. Rep. 7:43859. 10.1038/srep43859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Marteau P., Shanahan F. (2003). Basic aspects and pharmacology of probiotics: an overview of pharmacokinetics, mechanisms of action and side-effects. Best Pract. Res. Clin. Gastroenterol. 17, 725–740. 10.1016/S1521-6918(03)00055-6 [DOI] [PubMed] [Google Scholar]
  270. Martin W. F., Garg S., Zimorski V. (2015). Endosymbiotic theories for eukaryote origin. Philos. Trans. R. Soc. Biol. Sci. 370:20140330. 10.1098/rstb.2014.0330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Mawe G. M., Hoffman J. M. (2013). Serotonin signalling in the gut–functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 10, 473–486. 10.1038/nrgastro.2013.105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Mayer E. A., Knight R., Mazmanian S. K., Cryan J. F. (2014a). Gut microbes and the brain: paradigm shift in Neuroscience. J. Neurosci. 34, 15490–15496. 10.1523/JNEUROSCI.3299-14.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Mayer E. A., Padua D., Tillisch K. (2014b). Altered brain-gut axis in autism: comorbidity or causative mechanisms? Bioessays 36, 933–939. 10.1002/bies.201400075 [DOI] [PubMed] [Google Scholar]
  274. McCusker R. H., Kelley K. W. (2013). Immune-neural connections: how the immune system's response to infectious agents influences behavior. J. Exp. Biol. 216, 84–98. 10.1242/jeb.073411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. McEwen B. S. (2017). Allostasis and the epigenetics of brain and body health over the life course: the brain on stress. JAMA Psychiatry 74, 551–552. 10.1001/jamapsychiatry.2017.0270 [DOI] [PubMed] [Google Scholar]
  276. McEwen B. S., Nasca C., Gray J. D. (2016). Stress effects on neuronal structure: hippocampus, amygdala, and prefrontal cortex. Neuropsychopharmacology 41, 3–23. 10.1038/npp.2015.171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. McFall-Ngai M., Hadfield M. G., Bosch T. C., Carey H. V., Domazet-Loso T., Douglas A. E., et al. (2013). Animals in a bacterial world, a new imperative for the life sciences. Proc. Natl. Acad. Sci. U.S.A. 110, 3229–3236. 10.1073/pnas.1218525110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. McGlashan T. H., Hoffman R. E. (2000). Schizophrenia as a disorder of developmentally reduced synaptic connectivity. Arch. Gen. Psychiatry 57, 637–648. 10.1001/archpsyc.57.7.637 [DOI] [PubMed] [Google Scholar]
  279. McKernan D. P., Dennison U., Gaszner G., Cryan J. F., Dinan T. G. (2011). Enhanced peripheral toll-like receptor responses in psychosis: further evidence of a pro-inflammatory phenotype. Transl. Psychiatry 1:e36. 10.1038/tp.2011.37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Meehan C., Harms L., Frost J. D., Barreto R., Todd J., Schall U., et al. (2016). Effects of immune activation during early or late gestation on schizophrenia-related behaviour in adult rat offspring. Brain Behav. Immun. 63, 8–20. 10.1016/j.bbi.2016.07.144 [DOI] [PubMed] [Google Scholar]
  281. Messaoudi M., Lalonde R., Violle N., Javelot H., Desor D., Nejdi A., et al. (2011). Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 105, 755–764. 10.1017/S0007114510004319 [DOI] [PubMed] [Google Scholar]
  282. Messias E., Chen C.-Y., Eaton W. W. (2007). Epidemiology of Schizophrenia: review of Findings and Myths. Psychiatr. Clin. North Am. 30, 323–338. 10.1016/j.psc.2007.04.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Meyer U. (2014). Prenatal poly(i:C) exposure and other developmental immune activation models in rodent systems. Biol. Psychiatry 75, 307–315. 10.1016/j.biopsych.2013.07.011 [DOI] [PubMed] [Google Scholar]
  284. Meyer U., Feldon J., Yee B. K. (2009). A review of the fetal brain cytokine imbalance hypothesis of schizophrenia. Schizophr. Bull. 35, 959–972. 10.1093/schbul/sbn022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Meyer U., Nyffeler M., Engler A., Urwyler A., Schedlowski M., Knuesel I., et al. (2006). The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J. Neurosci. 26, 4752–4762. 10.1523/JNEUROSCI.0099-06.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Miller A. H., Haroon E., Raison C. L., Felger J. C. (2013). Cytokine targets in the brain: impact on neurotransmitters and neurocircuits. Depress. Anxiety 30, 297–306. 10.1002/da.22084 [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Miller B. J., Buckley P., Seabolt W., Mellor A., Kirkpatrick B. (2011). Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol. Psychiatry 70, 663–671. 10.1016/j.biopsych.2011.04.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Miyaoka T., Yasukawa R., Yasuda H., Hayashida M., Inagaki T., Horiguchi J. (2008). Minocycline as adjunctive therapy for schizophrenia: an open-label study. Clin. Neuropharmacol. 31, 287–292. 10.1097/WNF.0b013e3181593d45 [DOI] [PubMed] [Google Scholar]
  289. Mohammadi A. A., Jazayeri S., Khosravi-Darani K., Solati Z., Mohammadpour N., Asemi Z., et al. (2015). Effects of probiotics on biomarkers of oxidative stress and inflammatory factors in petrochemical workers: a randomized, double-blind, placebo-controlled trial. Int. J. Prev. Med. 6:82. 10.4103/2008-7802.164146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Mohle L., Mattei D., Heimesaat M. M., Bereswill S., Fischer A., Alutis M., et al. (2016). Ly6C(hi) monocytes provide a link between antibiotic-induced changes in gut microbiota and adult hippocampal neurogenesis. Cell Rep. 15, 1945–1956. 10.1016/j.celrep.2016.04.074 [DOI] [PubMed] [Google Scholar]
  291. Molloy C. A., Manning-Courtney P. (2003). Prevalence of chronic gastrointestinal symptoms in children with autism and autistic spectrum disorders. Autism 7, 165–171. 10.1177/1362361303007002004 [DOI] [PubMed] [Google Scholar]
  292. Molloy M. J., Grainger J. R., Bouladoux N., Hand T. W., Koo L. Y., Naik S., et al. (2013). Intraluminal containment of commensal outgrowth in the gut during infection-induced dysbiosis. Cell Host Microbe 14, 318–328. 10.1016/j.chom.2013.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Moloney R. D., Desbonnet L., Clarke G., Dinan T. G., Cryan J. F. (2014). The microbiome: stress, health and disease. Mamm. Genome 25, 49–74. 10.1007/s00335-013-9488-5 [DOI] [PubMed] [Google Scholar]
  294. Monk C. S., Weng S.-J., Wiggins J. L., Kurapati N., Louro H. M. C., Carrasco M., et al. (2010). Neural circuitry of emotional face processing in autism spectrum disorders. J. Psychiatry Neurosci. 35, 105–114. 10.1503/jpn.090085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Monroe J. M., Buckley P. F., Miller B. J. (2015). Meta-analysis of anti-Toxoplasma gondii IgM antibodies in acute psychosis. Schizophr. Bull. 41, 989–998. 10.1093/schbul/sbu159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Monteggia L. M., Barrot M., Powell C. M., Berton O., Galanis V., Gemelli T., et al. (2004). Essential role of brain-derived neurotrophic factor in adult hippocampal function. Proc. Natl. Acad. Sci. U.S.A. 101, 10827–10832. 10.1073/pnas.0402141101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Montiel C., Augusto J., Baez Y., Mario G., Pacheco-Lopez G. (2014). Social neuroeconomics: the influence of microbiota in partner-choice and sociality. Curr. Pharm. Des. 20, 4774–4783. 10.2174/1381612820666140130210631 [DOI] [PubMed] [Google Scholar]
  298. Morgan C. J., Coleman M. J., Ulgen A., Boling L., Cole J. O., Johnson F. V., et al. (2017). Thought disorder in schizophrenia and bipolar disorder probands, their relatives, and nonpsychiatric controls. Schizophr. Bull. 43, 523–535. 10.1093/schbul/sbx016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Moya-Pérez A., Perez-Villalba A., Benítez-Páez A., Campillo I., Sanz Y. (2017). Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice. Brain Behav. Immun. 65, 43–56. 10.1016/j.bbi.2017.05.011 [DOI] [PubMed] [Google Scholar]
  300. Mudd A. T., Berding K., Wang M., Donovan S. M., Dilger R. N. (2017). Serum cortisol mediates the relationship between fecal Ruminococcus and brain N-acetylaspartate in the young pig. Gut. Microbes. [Epub ahead of print]. 10.1080/19490976.2017.1353849 [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Muller N. (2014). Immunology of schizophrenia. Neuroimmunomodulation 21, 109–116. 10.1159/000356538 [DOI] [PubMed] [Google Scholar]
  302. Myint A. M., Kim Y. K. (2014). Network beyond IDO in psychiatric disorders: revisiting neurodegeneration hypothesis. Prog. Neuropsychopharmacol. Biol. Psychiatry 48, 304–313. 10.1016/j.pnpbp.2013.08.008 [DOI] [PubMed] [Google Scholar]
  303. Narendran R., Frankle W. G. (2016). Comment on Analyses and conclusions of “microglial activity in people at ultra high risk of psychosis and in schizophrenia: an [(11)C]PBR28 PET brain imaging study.” Am. J. Psychiatry 173, 536–537. 10.1176/appi.ajp.2016.15111417 [DOI] [PubMed] [Google Scholar]
  304. Naseribafrouei A., Hestad K., Avershina E., Sekelja M., Linlokken A., Wilson R., et al. (2014). Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 26, 1155–1162. 10.1111/nmo.12378 [DOI] [PubMed] [Google Scholar]
  305. Neufeld K. M., Kang N., Bienenstock J., Foster J. A. (2011). Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol. Motil. 23:255-e119. 10.1111/j.1365-2982.2010.01620.x [DOI] [PubMed] [Google Scholar]
  306. Neuhaus E., Jones E. J., Barnes K., Sterling L., Estes A., Munson J., et al. (2016). The relationship between early neural responses to emotional faces at age 3 and later autism and anxiety symptoms in adolescents with autism. J. Autism Dev. Disord. 46, 2450–2463. 10.1007/s10803-016-2780-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Newbury J., Arseneault L., Caspi A., Moffitt T. E., Odgers C. L., Fisher H. L. (2016). Why are children in urban neighborhoods at increased risk for psychotic symptoms? Findings from a UK longitudinal cohort study. Schizophr. Bull. 42, 1372–1383. 10.1093/schbul/sbw052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Nieto R., Kukuljan M., Silva H. (2013). BDNF and schizophrenia: from neurodevelopment to neuronal plasticity, learning, and memory. Front. Psychiatry 4:45. 10.3389/fpsyt.2013.00045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Nilsson L. K., Linderholm K. R., Engberg G., Paulson L., Blennow K., Lindstrom L. H., et al. (2005). Elevated levels of kynurenic acid in the cerebrospinal fluid of male patients with schizophrenia. Schizophr. Res. 80, 315–322. 10.1016/j.schres.2005.07.013 [DOI] [PubMed] [Google Scholar]
  310. Nishino R., Mikami K., Takahashi H., Tomonaga S., Furuse M., Hiramoto T., et al. (2013). Commensal microbiota modulate murine behaviors in a strictly contamination-free environment confirmed by culture-based methods. Neurogastroenterol. Motil. 25, 521–528. 10.1111/nmo.12110 [DOI] [PubMed] [Google Scholar]
  311. Notarangelo F. M., Pocivavsek A. (2017). Elevated kynurenine pathway metabolism during neurodevelopment: implications for brain and behavior. Neuropharmacology 112, 275–285. 10.1016/j.neuropharm.2016.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Notarangelo F. M., Schwarcz R. (2017). Restraint stress during pregnancy rapidly raises kynurenic acid levels in mouse placenta and fetal brain. Dev. Neurosci. 38, 458–468. 10.1159/000455228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  313. Notter T., Meyer U. (2017). Microglia and schizophrenia: where next? Mol. Psychiatry 22, 788–789. 10.1038/mp.2017.67 [DOI] [PubMed] [Google Scholar]
  314. O'Tuathaigh C. M. P., Fumagalli F., Desbonnet L., Perez-Branguli F., Moloney G., Loftus S., et al. (2017). Epistatic and independent effects on schizophrenia-related phenotypes following co-disruption of the risk factors neuregulin-1 × DISC1. Schizophr. Bull. 43, 214–225. 10.1093/schbul/sbw120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. O'Connor J. C., Lawson M. A., Andre C., Moreau M., Lestage J., Castanon N., et al. (2009). Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Mol. Psychiatry 14, 511–522. 10.1038/sj.mp.4002148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Ogbonnaya E. S., Clarke G., Shanahan F., Dinan T. G., Cryan J. F., O'Leary O. F. (2015). Adult hippocampal neurogenesis is regulated by the microbiome. Biol. Psychiatry 78, e7–e9. 10.1016/j.biopsych.2014.12.023 [DOI] [PubMed] [Google Scholar]
  317. O'Hara A. M., Shanahan F. (2006). The gut flora as a forgotten organ. EMBO Rep. 7, 688–693. 10.1038/sj.embor.7400731 [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Olszak T., An D., Zeissig S., Vera M. P., Richter J., Franke A., et al. (2012). Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489–493. 10.1126/science.1219328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. O'Mahony S. M., Clarke G., Borre Y. E., Dinan T. G., Cryan J. F. (2015). Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 277, 32–48. 10.1016/j.bbr.2014.07.027 [DOI] [PubMed] [Google Scholar]
  320. O'Mahony S. M., Clarke G., Dinan T. G., Cryan J. F. (2017). Early-life adversity and brain development: is the microbiome a missing piece of the puzzle? Neuroscience 342, 37–54. 10.1016/j.neuroscience.2015.09.068 [DOI] [PubMed] [Google Scholar]
  321. O'Mahony S. M., Marchesi J. R., Scully P., Codling C., Ceolho A. M., Quigley E. M., et al. (2009). Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol. Psychiatry 65, 263–267. 10.1016/j.biopsych.2008.06.026 [DOI] [PubMed] [Google Scholar]
  322. O'Neill S. M., Curran E. A., Dalman C., Kenny L. C., Kearney P. M., Clarke G., et al. (2015). Birth by caesarean section and the risk of adult psychosis: a population-based cohort study. Schizophr Bull. 42, 633–641. 10.1093/schbul/sbv152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Öngür D. (2017). Systems research in psychiatric neuroscience. JAMA Psychiatry 74, 553–554. 10.1001/jamapsychiatry.2017.0513 [DOI] [PubMed] [Google Scholar]
  324. Opie R. S., Itsiopoulos C., Parletta N., Sanchez-Villegas A., Akbaraly T. N., Ruusunen A., et al. (2017). Dietary recommendations for the prevention of depression. Nutr. Neurosci. 20, 161–171. 10.1179/1476830515Y.0000000043 [DOI] [PubMed] [Google Scholar]
  325. Parikshak N. N., Swarup V., Belgard T. G., Irimia M., Ramaswami G., Gandal M. J., et al. (2016). Genome-wide changes in lncRNA, splicing, and regional gene expression patterns in autism. Nature 540, 423–427. 10.1038/nature20612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Park S. J., Lee J. Y., Kim S. J., Choi S. Y., Yune T. Y., Ryu J. H. (2015). Toll-like receptor-2 deficiency induces schizophrenia-like behaviors in mice. Sci. Rep. 5:8502 10.1038/srep08502 [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Parracho H. M., Bingham M. O., Gibson G. R., McCartney A. L. (2005). Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J. Med. Microbiol. 54, 987–991. 10.1099/jmm.0.46101-0 [DOI] [PubMed] [Google Scholar]
  328. Patel S., Gruchalla R. (2017). Can we be too clean for our own good? The hygiene hypothesis reviewed. Tex. Med. 113, 54–59. [PubMed] [Google Scholar]
  329. Patterson P. H. (2011). Modeling autistic features in animals. Pediatr. Res. 69, 34R–40R. 10.1203/PDR.0b013e318212b80f [DOI] [PMC free article] [PubMed] [Google Scholar]
  330. Pedersen C. B., Mortensen P. B. (2001). Evidence of a dose-response relationship between urbanicity during upbringing and schizophrenia risk. Arch. Gen. Psychiatry 58, 1039–1046. 10.1001/archpsyc.58.11.1039 [DOI] [PubMed] [Google Scholar]
  331. Peen J., Schoevers R. A., Beekman A. T., Dekker J. (2010). The current status of urban-rural differences in psychiatric disorders. Acta Psychiatr. Scand. 121, 84–93. 10.1111/j.1600-0447.2009.01438.x [DOI] [PubMed] [Google Scholar]
  332. Penders J., Thijs C., van den Brandt P. A., Kummeling I., Snijders B., Stelma F., et al. (2007). Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut 56, 661–667. 10.1136/gut.2006.100164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Pendyala G., Chou S., Jung Y., Coiro P., Spartz E., Padmashri R., et al. (2017). Maternal immune activation causes behavioral impairments and altered cerebellar cytokine and synaptic protein expression. Neuropsychopharmacology 42, 1435–1446. 10.1038/npp.2017.7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Pequegnat B., Sagermann M., Valliani M., Toh M., Chow H., Allen-Vercoe E., et al. (2013). A vaccine and diagnostic target for Clostridium bolteae, an autism-associated bacterium. Vaccine 31, 2787–2790. 10.1016/j.vaccine.2013.04.018 [DOI] [PubMed] [Google Scholar]
  335. Perala J., Suvisaari J., Saarni S. I., Kuoppasalmi K., Isometsa E., Pirkola S., et al. (2007). Lifetime prevalence of psychotic and bipolar I disorders in a general population. Arch. Gen. Psychiatry 64, 19–28. 10.1001/archpsyc.64.1.19 [DOI] [PubMed] [Google Scholar]
  336. Perry V. H., Holmes C. (2014). Microglial priming in neurodegenerative disease. Nat. Rev. Neurol. 10, 217–224. 10.1038/nrneurol.2014.38 [DOI] [PubMed] [Google Scholar]
  337. Pershing M. L., Bortz D. M., Pocivavsek A., Fredericks P. J., Jorgensen C. V., Vunck S. A., et al. (2015). Elevated levels of kynurenic acid during gestation produce neurochemical, morphological, and cognitive deficits in adulthood: implications for schizophrenia. Neuropharmacology 90, 33–41. 10.1016/j.neuropharm.2014.10.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Pershing M. L., Phenis D., Valentini V., Pocivavsek A., Lindquist D. H., Schwarcz R., et al. (2016). Prenatal kynurenine exposure in rats: age-dependent changes in NMDA receptor expression and conditioned fear responding. Psychopharmacology 233, 3725–3735. 10.1007/s00213-016-4404-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Pinto-Sanchez M. I., Hall G. B., Ghajar K., Nardelli A., Bolino C., Lau J. T., et al. (2017). Probiotic Bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: a pilot study in patients with irritable bowel syndrome. Gastroenterology 153, 448–459.e8. 10.1053/j.gastro.2017.05.003 [DOI] [PubMed] [Google Scholar]
  340. Pisar M., Forrest C. M., Khalil O. S., McNair K., Vincenten M. C., Qasem S., et al. (2014). Modified neocortical and cerebellar protein expression and morphology in adult rats following prenatal inhibition of the kynurenine pathway. Brain Res. 1576, 1–17. 10.1016/j.brainres.2014.06.016 [DOI] [PubMed] [Google Scholar]
  341. Plitman E., Iwata Y., Caravaggio F., Nakajima S., Chung J. K., Gerretsen P., et al. (2017). Kynurenic acid in schizophrenia: a systematic review and meta-analysis. Schizophr. Bull. 43, 764–777. 10.1093/schbul/sbw221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Pocivavsek A., Thomas M. A., Elmer G. I., Bruno J. P., Schwarcz R. (2014). Continuous kynurenine administration during the prenatal period, but not during adolescence, causes learning and memory deficits in adult rats. Psychopharmacology 231, 2799–2809. 10.1007/s00213-014-3452-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Pollak T. A., Drndarski S., Stone J. M., David A. S., McGuire P., Abbott N. J. (2017). The blood-brain barrier in psychosis. Lancet Psychiatry. [Epub ahead of print]. 10.1016/S2215-0366(17)30293-6 [DOI] [PubMed] [Google Scholar]
  344. Prandovszky E., Gaskell E., Martin H., Dubey J. P., Webster J. P., McConkey G. A. (2011). The neurotropic parasite Toxoplasma gondii increases dopamine metabolism. PLoS ONE 6:e23866. 10.1371/journal.pone.0023866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Preissler M. A., Carey S. (2005). The role of inferences about referential intent in word learning: evidence from autism. Cognition 97, B13–B23. 10.1016/j.cognition.2005.01.008 [DOI] [PubMed] [Google Scholar]
  346. Prinz M., Tay T. L., Wolf Y., Jung S. (2014). Microglia: unique and common features with other tissue macrophages. Acta Neuropathol. 128, 319–331. 10.1007/s00401-014-1267-1 [DOI] [PubMed] [Google Scholar]
  347. Pyndt Jorgensen B., Krych L., Pedersen T. B., Plath N., Redrobe J. P., Hansen A. K., et al. (2015). Investigating the long-term effect of subchronic phencyclidine-treatment on novel object recognition and the association between the gut microbiota and behavior in the animal model of schizophrenia. Physiol. Behav. 141, 32–39. 10.1016/j.physbeh.2014.12.042 [DOI] [PubMed] [Google Scholar]
  348. Qin J., Li R., Raes J., Arumugam M., Burgdorf K. S., Manichanh C. (2010). A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 59–65. 10.1038/nature08821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Qin X., Feng J., Cao C., Wu H., Loh Y., Cheng Y. (2016). Association of peripheral blood levels of brain-derived neurotrophic factor with autism spectrum disorder in children: a systematic review and meta-analysis. JAMA Pediatr. 170, 1079–1086. 10.1001/jamapediatrics.2016.1626 [DOI] [PubMed] [Google Scholar]
  350. Quigley E. M. M. (2017). Gut microbiome as a clinical tool in gastrointestinal disease management: are we there yet? Nat. Rev. Gastroenterol. Hepatol. 14, 315–320. 10.1038/nrgastro.2017.29 [DOI] [PubMed] [Google Scholar]
  351. Rasetti R., Mattay V. S., Wiedholz L. M., Kolachana B. S., Hariri A. R., Callicott J. H., et al. (2009). Evidence that altered amygdala activity in schizophrenia is related to clinical state and not genetic risk. Am. J. Psychiatry 166, 216–225. 10.1176/appi.ajp.2008.08020261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  352. Rea K., Dinan T. G., Cryan J. F. (2016). The microbiome: a key regulator of stress and neuroinflammation. Neurobiol Stress 4, 23–33. 10.1016/j.ynstr.2016.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Reber S. O., Siebler P. H., Donner N. C., Morton J. T., Smith D. G., Kopelman J. M., et al. (2016). Immunization with a heat-killed preparation of the environmental bacterium Mycobacterium vaccae promotes stress resilience in mice. Proc. Natl. Acad.f Sci. U.S.A. 113, E3130–E3139. 10.1073/pnas.1600324113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Reed S., Neuman H., Moscovich S., Glahn R. P., Koren O., Tako E. (2015). Chronic zinc deficiency alters chick gut microbiota composition and function. Nutrients 7, 9768–9784. 10.3390/nu7125497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  355. Reigstad C. S., Salmonson C. E., Rainey J. F., III., Szurszewski J. H., Linden D. R., Sonnenburg J. L., et al. (2015). Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 29, 1395–1403. 10.1096/fj.14-259598 [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Reininghaus U., Bohnke J. R., Hosang G., Farmer A., Burns T., McGuffin P., et al. (2016a). Evaluation of the validity and utility of a transdiagnostic psychosis dimension encompassing schizophrenia and bipolar disorder. Br. J. Psychiatry 209, 107–113. 10.1192/bjp.bp.115.167882 [DOI] [PubMed] [Google Scholar]
  357. Reininghaus U., Kempton M. J., Valmaggia L., Craig T. K., Garety P., Onyejiaka A., et al. (2016b). Stress sensitivity, aberrant salience, and threat anticipation in early psychosis: an experience sampling study. Schizophr. Bull. 42, 712–722. 10.1093/schbul/sbv190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  358. Relman D. A. (2012). The human microbiome: ecosystem resilience and health. Nutr. Rev. 70(Suppl. 1), S2–S9. 10.1111/j.1753-4887.2012.00489.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Roche E., Segurado R., Renwick L., McClenaghan A., Sexton S., Frawley T., et al. (2016). Language disturbance and functioning in first episode psychosis. Psychiatry Res. 235, 29–37. 10.1016/j.psychres.2015.12.008 [DOI] [PubMed] [Google Scholar]
  360. Rogier R., Koenders M. I., Abdollahi-Roodsaz S. (2015). Toll-like receptor mediated modulation of T cell response by commensal intestinal microbiota as a trigger for autoimmune Arthritis 2015:527696. 10.1155/2015/527696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Rojas D. C. (2014). The role of glutamate and its receptors in autism and the use of glutamate receptor antagonists in treatment. J. Neural Transm. 121, 891–905. 10.1007/s00702-014-1216-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  362. Romijn A. R., Rucklidge J. J. (2015). Systematic review of evidence to support the theory of psychobiotics. Nutr. Rev. 73, 675–693. 10.1093/nutrit/nuv025 [DOI] [PubMed] [Google Scholar]
  363. Romijn A. R., Rucklidge J. J., Kuijer R. G., Frampton C. (2017). A double-blind, randomized, placebo-controlled trial of Lactobacillus helveticus and Bifidobacterium longum for the symptoms of depression. Aust. N. Z. J. Psychiatry 51, 810–821. 10.1177/0004867416686694 [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Rook G. A., Lowry C. A. (2008). The hygiene hypothesis and psychiatric disorders. Trends Immunol. 29, 150–158. 10.1016/j.it.2008.01.002 [DOI] [PubMed] [Google Scholar]
  365. Rook G. A., Lowry C. A., Raison C. L. (2013). Microbial ‘Old Friends’, immunoregulation and stress resilience. Evol. Med. Public Health 2013, 46–64. 10.1093/emph/eot004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Rook G. A., Martinelli R., Brunet L. R. (2003). Innate immune responses to mycobacteria and the downregulation of atopic responses. Curr. Opin. Allergy Clin. Immunol. 3, 337–342. 10.1097/00130832-200310000-00003 [DOI] [PubMed] [Google Scholar]
  367. Rook G. A., Raison C. L., Lowry C. A. (2014). Microbiota, immunoregulatory old friends and psychiatric disorders. Adv. Exp. Med. Biol. 817, 319–356. 10.1007/978-1-4939-0897-4_15 [DOI] [PubMed] [Google Scholar]
  368. Rosenberg E., Koren O., Reshef L., Efrony R., Zilber-Rosenberg I. (2007). The role of microorganisms in coral health, disease and evolution. Nat. Rev. Microbiol. 5, 355–362. 10.1038/nrmicro1635 [DOI] [PubMed] [Google Scholar]
  369. Round J. L., Mazmanian S. K. (2009). The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 9, 313–323. 10.1038/nri2515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  370. Ruddick J. P., Evans A. K., Nutt D. J., Lightman S. L., Rook G. A., Lowry C. A. (2006). Tryptophan metabolism in the central nervous system: medical implications. Expert Rev. Mol. Med. 8, 1–27. 10.1017/S1462399406000068 [DOI] [PubMed] [Google Scholar]
  371. Saghazadeh A., Rezaei N. (2017). Brain-derived neurotrophic factor levels in autism: a systematic review and meta-analysis. J. Autism Dev. Disord. 47, 1018–1029. 10.1007/s10803-016-3024-x [DOI] [PubMed] [Google Scholar]
  372. Sandin S., Schendel D., Magnusson P., Hultman C., Suren P., Susser E., et al. (2016). Autism risk associated with parental age and with increasing difference in age between the parents. Mol. Psychiatry 21, 693–700. 10.1038/mp.2015.70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Sandler R. H., Finegold S. M., Bolte E. R., Buchanan C. P., Maxwell A. P., Vaisanen M. L., et al. (2000). Short-term benefit from oral vancomycin treatment of regressive-onset autism. J. Child Neurol. 15, 429–435. 10.1177/088307380001500701 [DOI] [PubMed] [Google Scholar]
  374. Sarkar A., Lehto S. M., Harty S., Dinan T. G., Cryan J. F., Burnet P. W. J. (2016). Psychobiotics and the manipulation of bacteria–gut–brain signals. Trends Neurosci. 39, 763–781. 10.1016/j.tins.2016.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Savignac H. M., Corona G., Mills H., Chen L., Spencer J. P., Tzortzis G., et al. (2013). Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-D-aspartate receptor subunits and D-serine. Neurochem. Int. 63, 756–764. 10.1016/j.neuint.2013.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Savignac H. M., Tramullas M., Kiely B., Dinan T. G., Cryan J. F. (2015). Bifidobacteria modulate cognitive processes in an anxious mouse strain. Behav. Brain Res. 287, 59–72. 10.1016/j.bbr.2015.02.044 [DOI] [PubMed] [Google Scholar]
  377. Schaefer J., Giangrande E., Weinberger D. R., Dickinson D. (2013). The global cognitive impairment in schizophrenia: consistent over decades and around the world. Schizophr. Res. 150, 42–50. 10.1016/j.schres.2013.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Schendel D., Bhasin T. K. (2008). Birth weight and gestational age characteristics of children with autism, including a comparison with other developmental disabilities. Pediatrics 121, 1155–1164. 10.1542/peds.2007-1049 [DOI] [PubMed] [Google Scholar]
  379. Schenkel L. S., Silverstein S. M. (2004). Dimensions of premorbid functioning in schizophrenia: a review of neuromotor, cognitive, social, and behavioral domains. Genet. Soc. Gen. Psychol. Monogr. 130, 241–270. 10.3200/MONO.130.3.241-272 [DOI] [PubMed] [Google Scholar]
  380. Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014). Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427. 10.1038/nature13595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Schmidt K., Cowen P. J., Harmer C. J., Tzortzis G., Errington S., Burnet P. W. (2015). Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology 232, 1793–1801. 10.1007/s00213-014-3810-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Schmidt M. J., Mirnics K. (2015). Neurodevelopment, GABA system dysfunction, and schizophrenia. Neuropsychopharmacology 40, 190–206. 10.1038/npp.2014.95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  383. Schoenbaum M., Sutherland J. M., Chappel A., Azrin S., Goldstein A. B., Rupp A., et al. (2017). Twelve-month health care use and mortality in commercially insured young people with incident psychosis in the United States. Schizophr. Bull. [Epub ahead of print]. 10.1093/schbul/sbx009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Schwarcz R., Bruno J. P., Muchowski P. J., Wu H. Q. (2012). Kynurenines in the mammalian brain: when physiology meets pathology. Nat. Rev. Neurosci. 13, 465–477. 10.1038/nrn3257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Schwarz E., Maukonen J., Hyytiainen T., Kieseppa T., Orešič M., Sabunciyan S., et al. (in press). Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity treatment response. Schizophr. Res. 10.1016/j.schres.2017.04.017 [DOI] [PubMed] [Google Scholar]
  386. Sekar A., Bialas A. R., de Rivera H., Davis A., Hammond T. R., Kamitaki N., et al. (2016). Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183. 10.1038/nature16549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  387. Selten J. P., Cantor-Graae E. (2005). Social defeat: risk factor for schizophrenia? Br. J. Psychiatry 187, 101–102. 10.1192/bjp.187.2.101 [DOI] [PubMed] [Google Scholar]
  388. Selvaraj S., Arnone D., Cappai A., Howes O. (2014). Alterations in the serotonin system in schizophrenia: a systematic review and meta-analysis of postmortem and molecular imaging studies. Neurosci. Biobehav. Rev. 45, 233–245. 10.1016/j.neubiorev.2014.06.005 [DOI] [PubMed] [Google Scholar]
  389. Semple B. D., Blomgren K., Gimlin K., Ferriero D. M., Noble-Haeusslein L. J. (2013). Brain development in rodents and humans: identifying benchmarks of maturation and vulnerability to injury across species. Prog. Neurobiol. 106–107, 1–16. 10.1016/j.pneurobio.2013.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. Sender R., Fuchs S. (2016). Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 14:e1002533. 10.1371/journal.pbio.1002533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Setiawan E., Wilson A. A., Mizrahi R., Rusjan P. M., Miler L., Rajkowska G., et al. (2015). Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry 72, 268–275. 10.1001/jamapsychiatry.2014.2427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  392. Severance E. G., Gressitt K. L., Stallings C. R., Katsafanas E., Schweinfurth L. A., Savage C. L., et al. (2016a). Candida albicans exposures, sex specificity and cognitive deficits in schizophrenia and bipolar disorder. NPJ Schizophr. 2:16018. 10.1038/npjschz.2016.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  393. Severance E. G., Gressitt K. L., Stallings C. R., Katsafanas E., Schweinfurth L. A., Savage C. L., et al. (2017). Probiotic normalization of Candida albicans in schizophrenia: a randomized, placebo-controlled, longitudinal pilot study. Brain Behav. Immun. 62, 41–45. 10.1016/j.bbi.2016.11.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  394. Severance E. G., Gressitt K. L., Stallings C. R., Origoni A. E., Khushalani S., Leweke F. M., et al. (2013). Discordant patterns of bacterial translocation markers and implications for innate immune imbalances in schizophrenia. Schizophr. Res. 148, 130–137. 10.1016/j.schres.2013.05.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Severance E. G., Xiao J., Jones-Brando L., Sabunciyan S., Li Y., Pletnikov M., et al. (2016b). Chapter seven - Toxoplasma gondii—a gastrointestinal pathogen associated with human brain diseases, in International Review of Neurobiology, eds Cryan J. F., Clarke G. (Academic Press; ), 143–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Severance E. G., Yolken R. H., Eaton W. W. (2016c). Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: more than a gut feeling. Schizophr. Res. 176, 23–35. 10.1016/j.schres.2014.06.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  397. Shen M. D., Kim S. H., McKinstry R. C., Gu H., Hazlett H. C., Nordahl C. W., et al. (2017). Increased extra-axial cerebrospinal fluid in high-risk infants who later develop autism. Biol. Psychiatry 82, 186–193. 10.1016/j.biopsych.2017.02.1095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  398. Sherwin E., Dinan T. G., Cryan J. F. (2017). Recent developments in understanding the role of the gut microbiota in brain health and disease. Ann. N. Y. Acad. Sci. [Epub ahead of print]. 10.1111/nyas.13416 [DOI] [PubMed] [Google Scholar]
  399. Silbersweig D., Loscalzo J. (2017). Precision psychiatry meets network medicine: network psychiatry. JAMA Psychiatry 74, 665–666. 10.1001/jamapsychiatry.2017.0580 [DOI] [PMC free article] [PubMed] [Google Scholar]
  400. Slavich G. M., Way B. M., Eisenberger N. I., Taylor S. E. (2010). Neural sensitivity to social rejection is associated with inflammatory responses to social stress. Proc. Natl. Acad. Sci. U.S.A. 107, 14817–14822. 10.1073/pnas.1009164107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  401. Sliwa J., Freiwald W. A. (2017). A dedicated network for social interaction processing in the primate brain. Science 356, 745–749. 10.1126/science.aam6383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Smith S. E. P., Li J., Garbett K., Mirnics K., Patterson P. H. (2007). Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 27, 10695–10702. 10.1523/JNEUROSCI.2178-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Son J. S., Zheng L. J., Rowehl L. M., Tian X., Zhang Y., Zhu W., et al. (2015). Comparison of fecal microbiota in children with autism spectrum disorders and neurotypical siblings in the simons simplex collection. PLoS ONE 10:e0137725. 10.1371/journal.pone.0137725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Song Y., Liu C., Finegold S. M. (2004). Real-time PCR quantitation of clostridia in feces of autistic children. Appl. Environ. Microbiol. 70, 6459–6465. 10.1128/AEM.70.11.6459-6465.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  405. Spencer S. J., Meyer U. (2017). Perinatal programming by inflammation. Brain Behav. Immun. 63, 1–7. 10.1016/j.bbi.2017.02.007 [DOI] [PubMed] [Google Scholar]
  406. Spooren W., Lindemann L., Ghosh A., Santarelli L. (2012). Synapse dysfunction in autism: a molecular medicine approach to drug discovery in neurodevelopmental disorders. Trends Pharmacol. Sci. 33, 669–684. 10.1016/j.tips.2012.09.004 [DOI] [PubMed] [Google Scholar]
  407. Stamper C. E., Hoisington A. J., Gomez O. M., Halweg-Edwards A. L., Smith D. G., Bates K. L., et al. (2016). Chapter fourteen - the microbiome of the built environment and human behavior: implications for emotional health and well-being in postmodern western societies, in International Review of Neurobiology, eds Cryan J. F., Clarke G. (Academic Press; ), 289–323. [DOI] [PubMed] [Google Scholar]
  408. St Clair D., Xu M., Wang P., Yu Y., Fang Y., Zhang F., et al. (2005). Rates of adult schizophrenia following prenatal exposure to the Chinese famine of 1959-1961. JAMA 294, 557–562. 10.1001/jama.294.5.557 [DOI] [PubMed] [Google Scholar]
  409. Steele R. D. (1986). Blood-brain barrier transport of the alpha-keto acid analogs of amino acids. Fed. Proc. 45, 2060–2064. [PubMed] [Google Scholar]
  410. Steenbergen L., Sellaro R., van Hemert S., Bosch J. A., Colzato L. S. (2015). A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav. Immun. 48, 258–264. 10.1016/j.bbi.2015.04.003 [DOI] [PubMed] [Google Scholar]
  411. Stein M. M., Hrusch C. L., Gozdz J., Igartua C., Pivniouk V., Murray S. E., et al. (2016). Innate immunity and asthma risk in amish and hutterite farm children. N. Engl. J. Med. 375, 411–421. 10.1056/NEJMoa1508749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Stiles J., Jernigan T. L. (2010). The basics of brain development. Neuropsychol. Rev. 20, 327–348. 10.1007/s11065-010-9148-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  413. Stilling R. M., Bordenstein S. R., Dinan T. G., Cryan J. F. (2014a). Friends with social benefits: host-microbe interactions as a driver of brain evolution and development? Front. Cell. Infect. Microbiol. 4:147. 10.3389/fcimb.2014.00147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  414. Stilling R. M., Dinan T. G., Cryan J. F. (2014b). Microbial genes, brain and behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav. 13, 69–86. 10.1111/gbb.12109 [DOI] [PubMed] [Google Scholar]
  415. Stilling R. M., Ryan F. J., Hoban A. E., Shanahan F., Clarke G., Claesson M. J., et al. (2015). Microbes and neurodevelopment–Absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain Behav. Immun. 50, 209–220. 10.1016/j.bbi.2015.07.009 [DOI] [PubMed] [Google Scholar]
  416. Strachan D. P. (1989). Hay fever, hygiene, and household size. BMJ 299, 1259–1260. 10.1136/bmj.299.6710.1259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Strang J. F., Kenworthy L., Daniolos P., Case L., Wills M. C., Martin A., et al. (2012). Depression and anxiety symptoms in children and adolescents with autism spectrum disorders without intellectual disability. Res. Autism Spectr. Disord. 6, 406–412. 10.1016/j.rasd.2011.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. Strati F., Cavalieri D., Albanese D., De Felice C., Donati C., Hayek J., et al. (2017). New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome 5:24. 10.1186/s40168-017-0242-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  419. Sudo N., Chida Y., Aiba Y., Sonoda J., Oyama N., Yu X. N., et al. (2004). Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 558, 263–275. 10.1113/jphysiol.2004.063388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Sugama S., Takenouchi T., Fujita M., Kitani H., Hashimoto M. (2011). Cold stress induced morphological microglial activation and increased IL-1beta expression in astroglial cells in rat brain. J. Neuroimmunol. 233, 29–36. 10.1016/j.jneuroim.2010.11.002 [DOI] [PubMed] [Google Scholar]
  421. Suhr M. J., Hallen-Adams H. E. (2015). The human gut mycobiome: pitfalls and potentials–a mycologist's perspective. Mycologia 107, 1057–1073. 10.3852/15-147 [DOI] [PubMed] [Google Scholar]
  422. Sullivan P. F., Kendler K. S., Neale M. C. (2003). Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. Arch. Gen. Psychiatry 60, 1187–1192. 10.1001/archpsyc.60.12.1187 [DOI] [PubMed] [Google Scholar]
  423. Susser E. S., Lin S. P. (1992). Schizophrenia after prenatal exposure to the Dutch Hunger Winter of 1944-1945. Arch. Gen. Psychiatry 49, 983–988. 10.1001/archpsyc.1992.01820120071010 [DOI] [PubMed] [Google Scholar]
  424. Suzuki K., Sugihara G., Ouchi Y., Nakamura K., Futatsubashi M., Takebayashi K., et al. (2013). Microglial activation in young adults with autism spectrum disorder. JAMA Psychiatry 70, 49–58. 10.1001/jamapsychiatry.2013.272 [DOI] [PubMed] [Google Scholar]
  425. Takeda K., Akira S. (2005). Toll-like receptors in innate immunity. Int. Immunol. 17, 1–14. 10.1093/intimm/dxh186 [DOI] [PubMed] [Google Scholar]
  426. Tan J., McKenzie C., Potamitis M., Thorburn A. N., Mackay C. R., Macia L. (2014). The role of short-chain fatty acids in health and disease. Adv. Immunol. 121, 91–119. 10.1016/B978-0-12-800100-4.00003-9 [DOI] [PubMed] [Google Scholar]
  427. Tay T. L., Savage J. C., Hui C. W., Bisht K., Tremblay M.-È. (2017). Microglia across the lifespan: from origin to function in brain development, plasticity and cognition. J. Physiol. 595, 1929–1945. 10.1113/JP272134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  428. Teng B. L., Nonneman R. J., Agster K. L., Nikolova V. D., Davis T. T., Riddick N. V., et al. (2013). Prosocial effects of oxytocin in two mouse models of autism spectrum disorders. Neuropharmacology 72, 187–196. 10.1016/j.neuropharm.2013.04.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  429. Thaiss C. A., Levy M., Korem T., Dohnalová,á L., Shapiro H., Jaitin D. A., et al. (2016). microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 167, 1495–1510.e12. 10.1016/j.cell.2016.11.003 [DOI] [PubMed] [Google Scholar]
  430. Thomas E. H. X., Bozaoglu K., Rossell S. L., Gurvich C. (2017). The influence of the glutamatergic system on cognition in schizophrenia: a systematic review. Neurosci. Biobehav. Rev. 77, 369–387. 10.1016/j.neubiorev.2017.04.005 [DOI] [PubMed] [Google Scholar]
  431. Thomas R. H., Meeking M. M., Mepham J. R., Tichenoff L., Possmayer F., Liu S., et al. (2012). The enteric bacterial metabolite propionic acid alters brain and plasma phospholipid molecular species: further development of a rodent model of autism spectrum disorders. J. Neuroinflammation 9:153. 10.1186/1742-2094-9-153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  432. Tick B., Bolton P., Happe F., Rutter M., Rijsdijk F. (2016). Heritability of autism spectrum disorders: a meta-analysis of twin studies. J. Child Psychol. Psychiatry 57, 585–595. 10.1111/jcpp.12499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Tillisch K., Labus J., Kilpatrick L., Jiang Z., Stains J., Ebrat B., et al. (2013). Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 144, 1394–1401. 10.1053/j.gastro.2013.02.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Tomova A., Husarova V., Lakatosova S., Bakos J., Vlkova B., Babinska K., et al. (2015). Gastrointestinal microbiota in children with autism in Slovakia. Physiol. Behav. 138, 179–187. 10.1016/j.physbeh.2014.10.033 [DOI] [PubMed] [Google Scholar]
  435. Too L. K., McGregor I. S., Baxter A. G., Hunt N. H. (2016). Altered behaviour and cognitive function following combined deletion of Toll-like receptors 2 and 4 in mice. Behav. Brain Res. 303, 1–8. 10.1016/j.bbr.2016.01.024 [DOI] [PubMed] [Google Scholar]
  436. Torrey E. F., Yolken R. H. (2003). Toxoplasma gondii and schizophrenia. Emerging Infect. Dis. 9, 1375–1380. 10.3201/eid0911.030143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  437. Trudler D., Farfara D., Frenkel D. (2010). Toll-like receptors expression and signaling in glia cells in neuro-amyloidogenic diseases: towards future therapeutic application. Mediators Inflamm. 2010:497987. 10.1155/2010/497987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Turnbaugh P. J., Hamady M., Yatsunenko T., Cantarel B. L., Duncan A., Ley R. E., et al. (2009). A core gut microbiome in obese and lean twins. Nature 457, 480–484. 10.1038/nature07540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  439. Tynan R. J., Naicker S., Hinwood M., Nalivaiko E., Buller K. M., Pow D. V., et al. (2010). Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav. Immun. 24, 1058–1068. 10.1016/j.bbi.2010.02.001 [DOI] [PubMed] [Google Scholar]
  440. Udina M., Castellvi P., Moreno-Espana J., Navines R., Valdes M., Forns X., et al. (2012). Interferon-induced depression in chronic hepatitis C: a systematic review and meta-analysis. J. Clin. Psychiatry 73, 1128–1138. 10.4088/JCP.12r07694 [DOI] [PubMed] [Google Scholar]
  441. Upthegrove R., Manzanares-Teson N., Barnes N. M. (2014). Cytokine function in medication-naive first episode psychosis: a systematic review and meta-analysis. Schizophr. Res. 155, 101–108. 10.1016/j.schres.2014.03.005 [DOI] [PubMed] [Google Scholar]
  442. Valkanova V., Ebmeier K. P., Allan C. L. (2013). CRP, IL-6 and depression: a systematic review and meta-analysis of longitudinal studies. J. Affect. Disord. 150, 736–744. 10.1016/j.jad.2013.06.004 [DOI] [PubMed] [Google Scholar]
  443. van Berckel B. N., Bossong M. G., Boellaard R., Kloet R., Schuitemaker A., Caspers E., et al. (2008). Microglia activation in recent-onset schizophrenia: a quantitative (R)-[11C]PK11195 positron emission tomography study. Biol. Psychiatry 64, 820–822. 10.1016/j.biopsych.2008.04.025 [DOI] [PubMed] [Google Scholar]
  444. van Os J. (2016). “Schizophrenia” does not exist. BMJ 352, i375 10.1136/bmj.i375 [DOI] [PubMed] [Google Scholar]
  445. van Steensel F. J. A., Bögels S. M., Perrin S. (2011). Anxiety disorders in children and adolescents with autistic spectrum disorders: a meta-analysis. Clin. Child Fam. Psychol. Rev. 14:302. 10.1007/s10567-011-0097-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  446. Varese F., Smeets F., Drukker M., Lieverse R., Lataster T., Viechtbauer W., et al. (2012). Childhood adversities increase the risk of psychosis: a meta-analysis of patient-control, prospective- and cross-sectional cohort studies. Schizophr. Bull. 38, 661–671. 10.1093/schbul/sbs050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  447. Vassos E., Pedersen C. B., Murray R. M., Collier D. A., Lewis C. M. (2012). Meta-Analysis of the Association of Urbanicity With Schizophrenia. Schizophr. Bull. 38, 1118–1123. 10.1093/schbul/sbs096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  448. Vela G., Stark P., Socha M., Sauer A. K., Hagmeyer S., Grabrucker A. M. (2015). Zinc in gut-brain interaction in autism and neurological disorders. Neural Plast. 2015:15. 10.1155/2015/972791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  449. Vijay N., Morris M. E. (2014). Role of monocarboxylate transporters in drug delivery to the brain. Curr. Pharm. Des. 20, 1487–1498. 10.2174/13816128113199990462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  450. Voreades N., Kozil A., Weir T. L. (2014). Diet and the development of the human intestinal microbiome. Front. Microbiol. 5:494. 10.3389/fmicb.2014.00494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  451. Vorstman J. A. S., Parr J. R., Moreno-De-Luca D., Anney R. J. L., Nurnberger J. I., Jr., Hallmayer J. F. (2017). Autism genetics: opportunities and challenges for clinical translation. Nat. Rev. Genet. 18, 362–376. 10.1038/nrg.2017.4 [DOI] [PubMed] [Google Scholar]
  452. Vuong H. E., Hsiao E. Y. (2017). Emerging roles for the gut microbiome in autism spectrum disorder. Biol Psychiatry 81, 411–423. 10.1016/j.biopsych.2016.08.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. Wang L., Christophersen C. T., Sorich M. J., Gerber J. P., Angley M. T., Conlon M. A. (2011). Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism. Appl. Environ. Microbiol. 77, 6718–6721. 10.1128/AEM.05212-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  454. Wang L., Christophersen C. T., Sorich M. J., Gerber J. P., Angley M. T., Conlon M. A. (2012). Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig. Dis. Sci. 57, 2096–2102. 10.1007/s10620-012-2167-7 [DOI] [PubMed] [Google Scholar]
  455. Wang L., Christophersen C. T., Sorich M. J., Gerber J. P., Angley M. T., Conlon M. A. (2013). Increased abundance of Sutterella spp. and Ruminococcus torques in feces of children with autism spectrum disorder. Mol. Autism. 4:42. 10.1186/2040-2392-4-42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  456. Wang S., Adolphs R. (2017). Reduced specificity in emotion judgment in people with autism spectrum disorder. Neuropsychologia 99, 286–295. 10.1016/j.neuropsychologia.2017.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  457. Wang S. X., Wu W. C. (2005). Effects of psychological stress on small intestinal motility and bacteria and mucosa in mice. World J. Gastroenterol. 11, 2016–2021. 10.3748/wjg.v11.i13.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  458. Whitaker-Azmitia P. M. (2001). Serotonin and brain development: role in human developmental diseases. Brain Res. Bull. 56, 479–485. 10.1016/S0361-9230(01)00615-3 [DOI] [PubMed] [Google Scholar]
  459. WHO (2015). Statement on caesarean section rates. Reprod. Health Matt. 23, 149–150. 10.1016/j.rhm.2015.07.007 [DOI] [PubMed] [Google Scholar]
  460. Wikoff W. R., Anfora A. T., Liu J., Schultz P. G., Lesley S. A., Peters E. C., et al. (2009). Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U.S.A. 106, 3698–3703. 10.1073/pnas.0812874106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  461. Williams B. L., Hornig M., Buie T., Bauman M. L., Cho Paik M., Wick I., et al. (2011). Impaired carbohydrate digestion and transport and mucosal dysbiosis in the intestines of children with autism and gastrointestinal disturbances. PLoS ONE 6:e24585. 10.1371/journal.pone.0024585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  462. Williamson L. L., McKenney E. A., Holzknecht Z. E., Belliveau C., Rawls J. F., Poulton S., et al. (2015). Got worms? Perinatal exposure to helminths prevents persistent immune sensitization and cognitive dysfunction induced by early-life infection. Brain Behav. Immun. 51, 14–28. 10.1016/j.bbi.2015.07.006 [DOI] [PubMed] [Google Scholar]
  463. Willsey A. J., Sanders S. J., Li M., Dong S., Tebbenkamp A. T., Muhle R. A., et al. (2013). Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 155, 997–1007. 10.1016/j.cell.2013.10.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  464. Wong M. L., Inserra A., Lewis M. D., Mastronardi C. A., Leong L., Choo J., et al. (2016). Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol. Psychiatry 21, 797–805. 10.1038/mp.2016.46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  465. Xiao J., Li Y., Gressitt K. L., He H., Kannan G., Schultz T. L., et al. (2016). Cerebral complement C1q activation in chronic Toxoplasma infection. Brain Behav. Immun. 58, 52–56. 10.1016/j.bbi.2016.04.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  466. Xu M.-Q., Sun W.-S., Liu B.-X., Feng G.-Y., Yu L., Yang L., et al. (2009). Prenatal malnutrition and adult schizophrenia: further evidence from the 1959-1961 chinese famine. Schizophr. Bull. 35, 568–576. 10.1093/schbul/sbn168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  467. Yano J. M., Yu K., Donaldson G. P., Shastri G. G., Ann P., Ma L., et al. (2015). Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161, 264–276. 10.1016/j.cell.2015.02.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  468. Yasuda H., Yoshida K., Yasuda Y., Tsutsui T. (2011). Infantile zinc deficiency: association with autism spectrum disorders. Sci. Rep. 1:129. 10.1038/srep00129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  469. Yolken R. H., Severance E. G., Sabunciyan S., Gressitt K. L., Chen O., Stallings C., et al. (2015). Metagenomic sequencing indicates that the oropharyngeal phageome of individuals with schizophrenia differs from that of controls. Schizophr. Bull. 41, 1153–1161. 10.1093/schbul/sbu197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  470. Yolken R. H., Torrey E. F. (2008). Are some cases of psychosis caused by microbial agents? A review of the evidence. Mol Psychiatry 13, 470–479. 10.1038/mp.2008.5 [DOI] [PubMed] [Google Scholar]
  471. Yorbik Ö., Akay C., Sayal A., Cansever A., Söhmen T., O. Çavdar A. (2004). Zinc status in autistic children. J. Trace Elements Exp. Med. 17, 101–107. 10.1002/jtra.20002 [DOI] [Google Scholar]
  472. Yu M., Jia H., Zhou C., Yang Y., Zhao Y., Yang M., et al. (2017). Variations in gut microbiota and fecal metabolic phenotype associated with depression by 16S rRNA gene sequencing and LC/MS-based metabolomics. J. Pharm. Biomed. Anal. 138, 231–239. 10.1016/j.jpba.2017.02.008 [DOI] [PubMed] [Google Scholar]
  473. Yuen R. K. C., Merico D., Bookman M., Howe J. L., Thiruvahindrapuram B., Patel R. V., et al. (2017). Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat. Neurosci. 20, 602–611. 10.1038/nn.4524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  474. Zackular J. P., Moore J. L., Jordan A. T., Juttukonda L. J., Noto M. J., Nicholson M. R., et al. (2016). Dietary zinc alters the microbiota and decreases resistance to clostridium difficile infection. Nat. Med. 22, 1330–1334. 10.1038/nm.4174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  475. Zheng P., Zeng B., Zhou C., Liu M., Fang Z., Xu X., et al. (2016). Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism. Mol. Psychiatry. 21, 786–796 10.1038/mp.2016.44 [DOI] [PubMed] [Google Scholar]
  476. Zhernakova A., Kurilshikov A., Bonder M. J., Tigchelaar E. F., Schirmer M., Vatanen T., et al. (2016). Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352, 565–569. 10.1126/science.aad3369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  477. Zijlmans M. A. C., Korpela K., Riksen-Walraven J. M., de Vos W. M., de Weerth C. (2015). Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 53, 233–245. 10.1016/j.psyneuen.2015.01.006 [DOI] [PubMed] [Google Scholar]
  478. Zilber-Rosenberg I., Rosenberg E. (2008). Role of microorganisms in the evolution of animals and plants: the hologenome theory of evolution. FEMS Microbiol. Rev. 32, 723–735. 10.1111/j.1574-6976.2008.00123.x [DOI] [PubMed] [Google Scholar]

Articles from Frontiers in Neuroscience are provided here courtesy of Frontiers Media SA

RESOURCES